PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 8120182-4 1994 Milk production was correlated positively with DMI and water intake within and across parities. Desipramine 47-50 Weaning weight-maternal milk Bos taurus 0-4 8120182-7 1994 Rumination and total time spent chewing per unit of DMI were correlated negatively (r = -.58) with milk production within and across parities. Desipramine 52-55 Weaning weight-maternal milk Bos taurus 99-103 7734110-2 1994 GPT exerted the same biphasic effect on basal and DA-stimulated enzyme activity in membranes from DMI-treated rats as on saline-treated rats. Desipramine 98-101 glutamic--pyruvic transaminase Rattus norvegicus 0-3 7734110-5 1994 D2 inhibition of adenylate cyclase by DA was attenuated by the IAP treatment in both DMI-and saline-treated rats; peak levels of DA plus GTP stimulation shifted from 1 microM to 100 microM GTP. Desipramine 85-88 Cd47 molecule Rattus norvegicus 63-66 7734110-6 1994 D1 stimulation of adenylate cyclase by DA was also attenuated by the IAP in the DMI-treated rats. Desipramine 80-83 Cd47 molecule Rattus norvegicus 69-72 7734110-7 1994 Since long-term treatment with DMI (15 mg/kg, once a day, for 3 weeks) resulted in suppression of D1 stimulation similar to that seen in the present findings, uncoupling between D2 receptors and Gi due to IAP treatment might accelerate DMI-induced adaptive changes of dual control of adenylate cyclase system by DA. Desipramine 31-34 Cd47 molecule Rattus norvegicus 205-208 7734110-7 1994 Since long-term treatment with DMI (15 mg/kg, once a day, for 3 weeks) resulted in suppression of D1 stimulation similar to that seen in the present findings, uncoupling between D2 receptors and Gi due to IAP treatment might accelerate DMI-induced adaptive changes of dual control of adenylate cyclase system by DA. Desipramine 236-239 Cd47 molecule Rattus norvegicus 205-208 7969855-4 1994 Chronic desipramine and lithium chloride treatment slightly improved catalase activity in the bulbectomised rats. Desipramine 8-19 catalase Rattus norvegicus 69-77 8130740-7 1994 Na+,K(+)-ATPase increase by acute DMI was inhibited when endogenous NA was depleted by the noradrenergic neurotoxin DSP-4 or the NA synthesis inhibitor alpha-methyl-p-tyrosine. Desipramine 34-37 dual specificity phosphatase 26 Homo sapiens 116-121 8374761-0 1993 Norepinephrine transporter mRNA is elevated in the locus coeruleus following short- and long-term desipramine treatment. Desipramine 98-109 solute carrier family 6 member 2 Homo sapiens 0-26 8310712-14 1993 CYP2D6 catalyses C-hydroxylation of imipramine to 2-hydroxyimipramine more efficiently than its N-demethylation to desipramine. Desipramine 115-126 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 8110987-4 1993 Acetylcholinesterase (AChE) was inhibited by CPZ, 3,7,8-trihydroxy-CPZ, acridine orange partially saturated desipramine, imipramine, trans-clopenthixol and tetrahydrocannabidiolic at 10(-4) to 10(-5). Desipramine 108-119 acetylcholinesterase Rattus norvegicus 0-20 8110987-4 1993 Acetylcholinesterase (AChE) was inhibited by CPZ, 3,7,8-trihydroxy-CPZ, acridine orange partially saturated desipramine, imipramine, trans-clopenthixol and tetrahydrocannabidiolic at 10(-4) to 10(-5). Desipramine 108-119 acetylcholinesterase Rattus norvegicus 22-26 8218295-9 1993 The binding was most potently inhibited by the ligands of the norepinephrine transporter (desipramine and nomifensine). Desipramine 90-101 solute carrier family 6 member 2 Homo sapiens 62-88 8354888-3 1993 Imipramine and desipramine were structurally modified by the attachment of spacer arms to the aromatic ring which were subsequently attached to bovine serum albumin. Desipramine 15-26 albumin Oryctolagus cuniculus 151-164 8374761-1 1993 In situ hybridization for the norepinephrine transporter (NET) mRNA was performed in animals receiving short-term (2 days) and long-term (4 weeks) treatment with desipramine (DMI; 10 mg/kg, intraperitoneal). Desipramine 162-173 solute carrier family 6 member 2 Homo sapiens 30-56 8374761-1 1993 In situ hybridization for the norepinephrine transporter (NET) mRNA was performed in animals receiving short-term (2 days) and long-term (4 weeks) treatment with desipramine (DMI; 10 mg/kg, intraperitoneal). Desipramine 162-173 solute carrier family 6 member 2 Homo sapiens 58-61 8374761-1 1993 In situ hybridization for the norepinephrine transporter (NET) mRNA was performed in animals receiving short-term (2 days) and long-term (4 weeks) treatment with desipramine (DMI; 10 mg/kg, intraperitoneal). Desipramine 175-178 solute carrier family 6 member 2 Homo sapiens 58-61 8374761-2 1993 Following short-term and long-term DMI treatment, a significant (P < 0.05) increase in hybridization of 35S-labeled oligonucleotides to NET mRNA in the locus coeruleus was observed compared to that observed in vehicle-treated animals. Desipramine 35-38 solute carrier family 6 member 2 Homo sapiens 139-142 8315492-6 1993 MIBG uptake in hNET transfected cells is inhibited by 3 x 10(-6) M norepinephrine (87% inhibition) and by hNET transport inhibitors: 10(-7) M desipramine (94% inhibition) and 10(-7) M mazindol (97% inhibition). Desipramine 142-153 solute carrier family 6 member 2 Homo sapiens 15-19 8315492-6 1993 MIBG uptake in hNET transfected cells is inhibited by 3 x 10(-6) M norepinephrine (87% inhibition) and by hNET transport inhibitors: 10(-7) M desipramine (94% inhibition) and 10(-7) M mazindol (97% inhibition). Desipramine 142-153 solute carrier family 6 member 2 Homo sapiens 106-110 8335704-3 1993 In addition, however, the human CYP2D6 isozyme preparation was found to be unequivocally involved in the N-dealkylation of IMI to desmethylimipramine (DMI). Desipramine 130-149 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 32-38 8335704-3 1993 In addition, however, the human CYP2D6 isozyme preparation was found to be unequivocally involved in the N-dealkylation of IMI to desmethylimipramine (DMI). Desipramine 151-154 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 32-38 1480137-3 1992 Treatment of transgenic animals with the tricyclic antidepressant desipramine increased hypothalamic glucocorticoid receptor mRNA concentration and dexamethasone-binding activity while decreasing plasma adrenocorticotropin hormone concentration and corticosterone levels. Desipramine 66-77 nuclear receptor subfamily 3, group C, member 1 Mus musculus 101-124 8415816-0 1993 Chronic, combined treatment with desipramine and mianserin: enhanced 5-HT1A receptor function and altered 5-HT1A/5-HT2 receptor interaction in rats. Desipramine 33-44 5-hydroxytryptamine receptor 1A Rattus norvegicus 69-75 8415816-0 1993 Chronic, combined treatment with desipramine and mianserin: enhanced 5-HT1A receptor function and altered 5-HT1A/5-HT2 receptor interaction in rats. Desipramine 33-44 5-hydroxytryptamine receptor 1A Rattus norvegicus 106-112 8359181-7 1993 There was evidence for linkage between the CYP2D6 gene and the gene regulating the hydroxylation of desipramine and debrisoquine. Desipramine 100-111 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 43-49 7870896-6 1993 These findings suggest a decrease in the functional response of beta 1, but not beta 2, receptors after treatment with desipramine. Desipramine 119-130 potassium calcium-activated channel subfamily M regulatory beta subunit 1 Homo sapiens 64-70 1362806-0 1992 Semiquantitative analysis of immunoreactivities of tyrosine hydroxylase and aromatic L-amino acid decarboxylase in the locus coeruleus of desipramine-treated mice. Desipramine 138-149 dopa decarboxylase Mus musculus 76-111 8415816-3 1993 Pretreatment with desipramine and mianserin in combination induced the most intense 5-HT syndrome and the greatest fall in colonic temperature after injection of the 5-HT1A agonist 8-hydroxy-2-(di-n-propylamino)-tetralin (8-OH-DPAT). Desipramine 18-29 5-hydroxytryptamine receptor 1A Rattus norvegicus 166-172 8336822-7 1993 Several studies have shown a functional upregulation of the 5-HT1A receptor after chronic treatment with DMI. Desipramine 105-108 5-hydroxytryptamine (serotonin) receptor 1A Mus musculus 60-75 8358025-1 1993 Several amphiphilic cations such as mepacrine, desipramine, didodecyldimethylamine, chlorpromazine, oleylamine and W-7 activated the phospholipase D (PLD) activity of cultured LA-N-2 cells. Desipramine 47-58 glycosylphosphatidylinositol specific phospholipase D1 Homo sapiens 133-148 8358025-1 1993 Several amphiphilic cations such as mepacrine, desipramine, didodecyldimethylamine, chlorpromazine, oleylamine and W-7 activated the phospholipase D (PLD) activity of cultured LA-N-2 cells. Desipramine 47-58 glycosylphosphatidylinositol specific phospholipase D1 Homo sapiens 150-153 8439771-1 1993 The effects of acute and chronic treatment with the tricyclic antidepressant drugs, imipramine, clomipramine and desipramine on levels of neuropeptide Y (NPY) and prepro-NPY-mRNA were studied in different areas of the rat brain. Desipramine 113-124 neuropeptide Y Rattus norvegicus 138-152 8439771-1 1993 The effects of acute and chronic treatment with the tricyclic antidepressant drugs, imipramine, clomipramine and desipramine on levels of neuropeptide Y (NPY) and prepro-NPY-mRNA were studied in different areas of the rat brain. Desipramine 113-124 neuropeptide Y Rattus norvegicus 154-157 8439771-1 1993 The effects of acute and chronic treatment with the tricyclic antidepressant drugs, imipramine, clomipramine and desipramine on levels of neuropeptide Y (NPY) and prepro-NPY-mRNA were studied in different areas of the rat brain. Desipramine 113-124 neuropeptide Y Rattus norvegicus 170-173 8513845-3 1993 This confirms that desipramine is extensively metabolized via the sparteine/debrisoquine oxidation polymorphism i.e. by CYP2D6. Desipramine 19-30 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 120-126 8513845-5 1993 The 5-fold decrease in clearance in EMs when desipramine was co-administered with paroxetine confirms that paroxetine is a potent inhibitor of CYP2D6. Desipramine 45-56 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 143-149 8464952-0 1993 Effect of desipramine on cerebrospinal fluid concentrations of corticotropin-releasing factor in human subjects. Desipramine 10-21 corticotropin releasing hormone Homo sapiens 63-93 8464952-1 1993 To assess the effect of desipramine (DMI) on corticotropin-releasing-factor (CRF) activity in the central nervous system, we measured cerebrospinal fluid (CSF) concentrations of CRF in healthy volunteers following short-term administration of DMI or placebo. Desipramine 37-40 corticotropin releasing hormone Homo sapiens 45-75 1336635-0 1992 The desipramine-induced growth hormone response and the dexamethasone suppression test in obsessive-compulsive disorder. Desipramine 4-15 growth hormone 1 Homo sapiens 24-38 1359398-0 1992 Long term treatment with desipramine increases the turnover of alpha 2-adrenoceptors in the rat brain. Desipramine 25-36 adrenoceptor alpha 2A Rattus norvegicus 63-84 1359398-1 1992 The aim of this study was to quantitate the turnover of alpha 2-adrenoceptors in different regions of the rat brain and its modulation during desipramine (a cyclic antidepressant drug)-induced receptor down-regulation. Desipramine 142-153 adrenoceptor alpha 2A Rattus norvegicus 56-77 1359398-5 1992 In the cerebral cortex and other brain regions, desipramine induced a time-dependent modulation of alpha 2-adrenoceptors, with significant decreases in the number of receptors (40-71%; p < 0.01) during the first 7-14 days of treatment and regulation to base-line values by 21-35 days. Desipramine 48-59 adrenoceptor alpha 2A Rattus norvegicus 99-120 1387021-10 1992 Rats treated with DMI but not sertraline showed a virtually complete suppression of the growth hormone (GH) secretion elicited by clonidine in controls, while the secretion of corticosterone was augmented. Desipramine 18-21 gonadotropin releasing hormone receptor Rattus norvegicus 88-102 1389951-7 1992 CYP2D6 activity was also diminished by the tricyclic antidepressant drugs clomipramine (2.2 microM), desipramine (2.3 microM) and amitriptyline (4.0 microM). Desipramine 101-112 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 0-6 1436127-7 1992 If desipramine (25 mg/kg) was administered before DSP-4 treatment, the DSP-4-induced changes both in noradrenaline uptake and CCK receptor binding were not present, suggesting that both effects were exerted after uptake of the neurotoxin by the nerve terminals. Desipramine 3-14 cholecystokinin Rattus norvegicus 126-129 1325288-6 1992 Desimipramine, but not reserpine, easily depleted IFN-gamma-treated LAN-5 cells of their MIBG content. Desipramine 0-13 interferon gamma Homo sapiens 50-59 1610412-1 1992 The effects of long-term (28-day) administration of several antidepressant/antipanic drugs [imipramine, desipramine, tranylcypromine and phenelzine (PLZ)] on gamma-aminobutyric acid-tranaminase (GABA-T) activity and GABA levels were investigated in rat frontal cortex. Desipramine 104-115 4-aminobutyrate aminotransferase Rattus norvegicus 158-193 1614406-1 1992 We have tested the hypothesis that antidepressants affect the expression of the glucocorticoid receptor gene, by looking at glucocorticoid receptor gene promoter activity, glucocorticoid receptor mRNA levels, and glucocorticoid-binding activity after treatment of different cell lines with desipramine. Desipramine 290-301 nuclear receptor subfamily 3, group C, member 1 Mus musculus 80-103 1614406-2 1992 Treatment of LTK- cells or Neuro 2A cells with desipramine produced a 50-200% increase in chloramphenicol acetyltransferase activity transcribed from a 2.7-kilobase glucocorticoid receptor gene promoter region. Desipramine 47-58 leukocyte tyrosine kinase Mus musculus 13-16 1614406-2 1992 Treatment of LTK- cells or Neuro 2A cells with desipramine produced a 50-200% increase in chloramphenicol acetyltransferase activity transcribed from a 2.7-kilobase glucocorticoid receptor gene promoter region. Desipramine 47-58 nuclear receptor subfamily 3, group C, member 1 Mus musculus 165-188 1614406-3 1992 In cell lines derived from both neuronal and non-neuronal sources, glucocorticoid receptor mRNA concentration doubled after desipramine treatment, and this was associated with a 2-fold higher functional glucocorticoid binding capacity and increased glucocorticoid sensitivity, as measured with the reporter plasmid pMMTVCAT. Desipramine 124-135 nuclear receptor subfamily 3, group C, member 1 Mus musculus 67-90 1387021-10 1992 Rats treated with DMI but not sertraline showed a virtually complete suppression of the growth hormone (GH) secretion elicited by clonidine in controls, while the secretion of corticosterone was augmented. Desipramine 18-21 gonadotropin releasing hormone receptor Rattus norvegicus 104-106 1387021-16 1992 These results show that DMI treatment blunted the sensitivity of post-synaptic alpha 2-adrenoceptors, accompanied by complex interactions manifested as increased responsiveness of alpha 1-adrenoceptors and 5-HT1A receptors. Desipramine 24-27 5-hydroxytryptamine receptor 1A Rattus norvegicus 180-212 1346258-0 1992 Analysis of the CYP2D6 gene in relation to debrisoquin and desipramine hydroxylation in a Swedish population. Desipramine 59-70 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 16-22 1744272-8 1991 Growth hormone-releasing factor increased DMI (23.2 kg/d) over that of controls (21.1 kg/d), whereas bST (21.5 kg/d) did not. Desipramine 42-45 growth hormone releasing hormone Bos taurus 0-31 1542151-3 1992 In this study, the influence of changing salt status and angiotensin II activity has been investigated by evaluating the QRS prolonging effects of the sodium channel blocking drug, desmethylimipramine. Desipramine 181-200 angiotensinogen Rattus norvegicus 57-71 1542151-10 1992 The QRS response to desmethylimipramine and salt-loaded rats on normal salt diets receiving captopril returned to the control pattern after a subpressor infusion of angiotensin II (3 ng/min), while a higher rate of angiotensin II (10 ng/min) further enhanced the QRS prolonging effect of desmethylimipramine. Desipramine 20-39 angiotensinogen Rattus norvegicus 165-179 1542151-11 1992 These data demonstrate that endogenous angiotensin II contributes to the regulation of the cardiac electro-physiological response to DMI. Desipramine 133-136 angiotensinogen Rattus norvegicus 39-53 1775194-4 1991 After inhibition of MAO, desipramine caused a further five-fold increase in the basal efflux of dopamine with no change in the basal efflux of DOPAC. Desipramine 25-36 monoamine oxidase A Rattus norvegicus 20-23 1839385-0 1991 Similar effects of treatment with desipramine and electroconvulsive shock on 5-hydroxytryptamine1A receptors in rat brain. Desipramine 34-45 5-hydroxytryptamine receptor 1A Rattus norvegicus 77-98 1839385-1 1991 The effect of chronic and acute treatment with desipramine (DMI) and electroconvulsive shock (ECS) on 5-hydroxytryptamine1A (5-HT1A) receptors was determined in the cortex and the hippocampus brain regions of rats. Desipramine 47-58 5-hydroxytryptamine receptor 1A Rattus norvegicus 102-123 1839385-1 1991 The effect of chronic and acute treatment with desipramine (DMI) and electroconvulsive shock (ECS) on 5-hydroxytryptamine1A (5-HT1A) receptors was determined in the cortex and the hippocampus brain regions of rats. Desipramine 47-58 5-hydroxytryptamine receptor 1A Rattus norvegicus 125-131 1839385-1 1991 The effect of chronic and acute treatment with desipramine (DMI) and electroconvulsive shock (ECS) on 5-hydroxytryptamine1A (5-HT1A) receptors was determined in the cortex and the hippocampus brain regions of rats. Desipramine 60-63 5-hydroxytryptamine receptor 1A Rattus norvegicus 102-123 1839385-1 1991 The effect of chronic and acute treatment with desipramine (DMI) and electroconvulsive shock (ECS) on 5-hydroxytryptamine1A (5-HT1A) receptors was determined in the cortex and the hippocampus brain regions of rats. Desipramine 60-63 5-hydroxytryptamine receptor 1A Rattus norvegicus 125-131 1839385-2 1991 We observed that chronic treatment with both DMI and ECS significantly decreased 5-HT1A receptors, as determined by [3H]8-hydroxy-2-(di-n-propylamino)tetralin [( 3H]8-OH-DPAT) binding, in the cortex but not in the hippocampus. Desipramine 45-48 5-hydroxytryptamine receptor 1A Rattus norvegicus 81-87 1912129-1 1991 Twelve patients with DSM-III-R major depressive illness were tested for growth hormone (GH) response to desipramine (DMI), a noradrenergic (NA) reuptake inhibitor. Desipramine 104-115 growth hormone 1 Homo sapiens 72-86 1912129-1 1991 Twelve patients with DSM-III-R major depressive illness were tested for growth hormone (GH) response to desipramine (DMI), a noradrenergic (NA) reuptake inhibitor. Desipramine 104-115 growth hormone 1 Homo sapiens 88-90 1912129-1 1991 Twelve patients with DSM-III-R major depressive illness were tested for growth hormone (GH) response to desipramine (DMI), a noradrenergic (NA) reuptake inhibitor. Desipramine 117-120 growth hormone 1 Homo sapiens 72-86 1912129-1 1991 Twelve patients with DSM-III-R major depressive illness were tested for growth hormone (GH) response to desipramine (DMI), a noradrenergic (NA) reuptake inhibitor. Desipramine 117-120 growth hormone 1 Homo sapiens 88-90 1912129-6 1991 Patients treated with fluoxetine showed a significant decrease in DMI-mediated GH release irrespective of therapeutic outcome. Desipramine 66-69 growth hormone 1 Homo sapiens 79-81 1782991-7 1991 Desipramine selectively inhibited the T-current and Cl- efflux for 1 min. Desipramine 0-11 nuclear receptor subfamily 1, group H, member 5 L homeolog Xenopus laevis 63-68 1663633-0 1991 Down-regulation of beta receptors by desipramine in vitro involves PKC/phospholipase A2. Desipramine 37-48 phospholipase A2 group IB Rattus norvegicus 71-87 1717808-0 1991 Repeated administration of desmethylimipramine blocks the reserpine-induced increase in tyrosine hydroxylase mRNA in locus coeruleus neurons of the rat. Desipramine 27-46 tyrosine hydroxylase Rattus norvegicus 88-108 1717808-4 1991 However, coadministration of desmethylimipramine with reserpine blocked the elevation in tyrosine hydroxylase mRNA induced by reserpine alone. Desipramine 29-48 tyrosine hydroxylase Rattus norvegicus 89-109 1849767-0 1991 Secretion of growth hormone elicited by intravenous desipramine in the conscious, unrestrained rat. Desipramine 52-63 gonadotropin releasing hormone receptor Rattus norvegicus 13-27 1849767-2 1991 Acute intravenous administration of either clonidine (Clon) (50 micrograms kg-1) or desipramine (DMI) (5 mg kg-1) elicited a pulse of growth hormone (GH) and corticosterone secretion in conscious, unrestrained rats. Desipramine 84-95 gonadotropin releasing hormone receptor Rattus norvegicus 134-148 1849767-2 1991 Acute intravenous administration of either clonidine (Clon) (50 micrograms kg-1) or desipramine (DMI) (5 mg kg-1) elicited a pulse of growth hormone (GH) and corticosterone secretion in conscious, unrestrained rats. Desipramine 84-95 gonadotropin releasing hormone receptor Rattus norvegicus 150-152 1849767-2 1991 Acute intravenous administration of either clonidine (Clon) (50 micrograms kg-1) or desipramine (DMI) (5 mg kg-1) elicited a pulse of growth hormone (GH) and corticosterone secretion in conscious, unrestrained rats. Desipramine 97-100 gonadotropin releasing hormone receptor Rattus norvegicus 134-148 1849767-2 1991 Acute intravenous administration of either clonidine (Clon) (50 micrograms kg-1) or desipramine (DMI) (5 mg kg-1) elicited a pulse of growth hormone (GH) and corticosterone secretion in conscious, unrestrained rats. Desipramine 97-100 gonadotropin releasing hormone receptor Rattus norvegicus 150-152 1849767-4 1991 The responses to DMI were similar to those with Clon, except that the GH pulse following DMI was delayed and was not dose-dependent. Desipramine 17-20 gonadotropin releasing hormone receptor Rattus norvegicus 70-72 1849767-4 1991 The responses to DMI were similar to those with Clon, except that the GH pulse following DMI was delayed and was not dose-dependent. Desipramine 89-92 gonadotropin releasing hormone receptor Rattus norvegicus 70-72 1849767-6 1991 The GH response to DMI was inhibited by prior administration of idazoxan (1 mg kg-1) or yohimbine (0.5 mg kg-1), but not by atropine (10 micrograms kg-1), sulpiride (5 mg kg-1) or prazosin (1 mg kg-1). Desipramine 19-22 gonadotropin releasing hormone receptor Rattus norvegicus 4-6 1849767-14 1991 DMI caused an activation through indirect mechanisms of alpha 2-adrenoceptors specifically involved in hypothalamic-pituitary regulation of GH release and also a distinct, independent and transient generalized activation of the pituitary-adrenal axis. Desipramine 0-3 gonadotropin releasing hormone receptor Rattus norvegicus 140-142 1647239-8 1991 However, the ability of NPY (0.05-0.5 microM) to inhibit forskolin (1 microM) stimulated adenylate cyclase via Y2 NPY-type receptors in rat frontal cortical slices was significantly reduced following chronic desipramine treatment. Desipramine 208-219 neuropeptide Y Rattus norvegicus 24-27 1775595-2 1991 We observed that DMI inhibited thrombin-stimulated 3H-inositol bisphosphate (IP2) and 3H-inositol trisphosphate (IP3) but not 3H-inositol monophosphate (IP1) formation in human platelets. Desipramine 17-20 coagulation factor II, thrombin Homo sapiens 31-39 1775595-2 1991 We observed that DMI inhibited thrombin-stimulated 3H-inositol bisphosphate (IP2) and 3H-inositol trisphosphate (IP3) but not 3H-inositol monophosphate (IP1) formation in human platelets. Desipramine 17-20 inhibitor of nuclear factor kappa B kinase regulatory subunit gamma Homo sapiens 77-80 1775595-2 1991 We observed that DMI inhibited thrombin-stimulated 3H-inositol bisphosphate (IP2) and 3H-inositol trisphosphate (IP3) but not 3H-inositol monophosphate (IP1) formation in human platelets. Desipramine 17-20 inhibitor of nuclear factor kappa B kinase regulatory subunit gamma Homo sapiens 153-156 1775595-3 1991 DMI also inhibited norepinephrine (NE) and serotonin (5-HT) stimulated 3H-IP1 formation in rat cerebral cortex. Desipramine 0-3 inhibitor of nuclear factor kappa B kinase regulatory subunit gamma Homo sapiens 74-77 1775595-4 1991 DMI increased levels of all three 3H-inositol phospholipids, 3H-phosphatidyl inositol (PI), 3H-PI-4-phosphate (PIP), and 3H-PI 4,5-bisphosphate (PIP2), in both platelets and rat cortex. Desipramine 0-3 prolactin induced protein Rattus norvegicus 92-109 1775595-4 1991 DMI increased levels of all three 3H-inositol phospholipids, 3H-phosphatidyl inositol (PI), 3H-PI-4-phosphate (PIP), and 3H-PI 4,5-bisphosphate (PIP2), in both platelets and rat cortex. Desipramine 0-3 prolactin induced protein Rattus norvegicus 111-114 1775595-5 1991 The decreased formation of inositol phosphates and increased levels of [3H]-PI, [3H]-PIP, and [3H]-PIP2 by DMI appears to be due to the inhibition of the enzyme phospholipase C rather than its effects on receptors. Desipramine 107-110 prolactin induced protein Rattus norvegicus 85-88 1811246-2 1991 Northern blot analysis of Type II GR mRNA showed that treatment of male rats with either desipramine or imipramine increased hypothalamic and hippocampal GR mRNA levels. Desipramine 89-100 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 34-36 1811246-2 1991 Northern blot analysis of Type II GR mRNA showed that treatment of male rats with either desipramine or imipramine increased hypothalamic and hippocampal GR mRNA levels. Desipramine 89-100 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 154-156 1811246-6 1991 We conclude that desipramine, imipramine, ketanserin and lithium can modulate GR mRNA in regions of rat brain involved in the control of the HPA axis and may have a common mechanism of action at the level of the GR gene. Desipramine 17-28 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 78-80 1811246-6 1991 We conclude that desipramine, imipramine, ketanserin and lithium can modulate GR mRNA in regions of rat brain involved in the control of the HPA axis and may have a common mechanism of action at the level of the GR gene. Desipramine 17-28 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 212-214 1540485-2 1992 The activity of N-sulphotransferase (N-ST) with desipramine (DMI) as substrate was measured in 118 human liver specimens, in platelets obtained from 105 subjects, in 12 specimens of human ileum and colon mucosa and in five specimens of human kidney and lung. Desipramine 48-59 sulfotransferase family 4A member 1 Homo sapiens 16-35 1540485-2 1992 The activity of N-sulphotransferase (N-ST) with desipramine (DMI) as substrate was measured in 118 human liver specimens, in platelets obtained from 105 subjects, in 12 specimens of human ileum and colon mucosa and in five specimens of human kidney and lung. Desipramine 48-59 sulfotransferase family 4A member 1 Homo sapiens 37-41 1540485-2 1992 The activity of N-sulphotransferase (N-ST) with desipramine (DMI) as substrate was measured in 118 human liver specimens, in platelets obtained from 105 subjects, in 12 specimens of human ileum and colon mucosa and in five specimens of human kidney and lung. Desipramine 61-64 sulfotransferase family 4A member 1 Homo sapiens 16-35 1540485-2 1992 The activity of N-sulphotransferase (N-ST) with desipramine (DMI) as substrate was measured in 118 human liver specimens, in platelets obtained from 105 subjects, in 12 specimens of human ileum and colon mucosa and in five specimens of human kidney and lung. Desipramine 61-64 sulfotransferase family 4A member 1 Homo sapiens 37-41 1647142-1 1991 Seven drug-free patients with bipolar affective disorder, currently in a manic phase, underwent desipramine-induced growth hormone (GH) stimulation, as did seven healthy age- and sex-matched subjects. Desipramine 96-107 growth hormone 1 Homo sapiens 116-130 1860973-5 1991 Milk to DMI ratio and milk energy to net energy intake ratio were improved by somidobove. Desipramine 8-11 Weaning weight-maternal milk Bos taurus 0-4 1672374-3 1991 Desipramine, imipramine, amitriptyline and iprindole inhibited thrombin-stimulated formation of [3H]IP2 and [3H]IP3 in human platelets but had no significant effect on [3H]IP1 formation. Desipramine 0-11 coagulation factor II, thrombin Homo sapiens 63-71 1672374-3 1991 Desipramine, imipramine, amitriptyline and iprindole inhibited thrombin-stimulated formation of [3H]IP2 and [3H]IP3 in human platelets but had no significant effect on [3H]IP1 formation. Desipramine 0-11 inhibitor of nuclear factor kappa B kinase regulatory subunit gamma Homo sapiens 100-103 1672374-3 1991 Desipramine, imipramine, amitriptyline and iprindole inhibited thrombin-stimulated formation of [3H]IP2 and [3H]IP3 in human platelets but had no significant effect on [3H]IP1 formation. Desipramine 0-11 inhibitor of nuclear factor kappa B kinase regulatory subunit gamma Homo sapiens 172-175 1663633-7 1991 Coincubation of C6 cells with either the phospholipase A2 (PLA2) inhibitor mepacrine or the protein kinase C (PKC) inhibitor H7, during chronic treatment with DMI, blocked the down-regulation of beta ARs. Desipramine 159-162 phospholipase A2 group IB Rattus norvegicus 41-57 1663633-7 1991 Coincubation of C6 cells with either the phospholipase A2 (PLA2) inhibitor mepacrine or the protein kinase C (PKC) inhibitor H7, during chronic treatment with DMI, blocked the down-regulation of beta ARs. Desipramine 159-162 phospholipase A2 group IB Rattus norvegicus 59-63 2076491-2 1990 In tissues previously incubated with [3H]-noradrenaline exposure to cocaine (0.1 to 10 microM) or desmethylimipramine (0.01 to 1 microM) produced a concentration-dependent increase (up to 2 fold) in electrically evoked (3 Hz, 2 ms, 20 mA, 120s every 20 min) fractional overflow of tritium from rat brain cortex slices but not from mouse vas deferens (2.5 Hz, 2 ms, 400 mA, for 90s every 14 min). Desipramine 98-117 arginine vasopressin Rattus norvegicus 337-340 19912776-4 1990 The catecholamine uptake blocker desipramine and the alpha(2)-adrenergic receptor agonist clonidine inhibited accumulation of newly synthesized catecholamines, but only after prolonged exposure to drug; NPY synthesis and accumulation were unaltered by these treatments. Desipramine 33-44 neuropeptide Y Rattus norvegicus 203-206 2128400-2 1990 Neurotensin was injected into the anterior chamber (AC) at a dose of 30 micrograms to 4 groups of albino rabbits which had previously undergone the following treatment: a) desmethylimipramine IM and, after 30 min, 6-hydroxydopamine IV 7 days prior to the neurotensin administration; b) haloperidol IM for 15 days; c) haloperidol AC 10 minutes before the neurotensin administration. Desipramine 172-191 neurotensin/neuromedin N Oryctolagus cuniculus 0-11 1980647-3 1990 The [Ca2+]i response to NMDA was blocked or reversed by selective antagonists such as 2-amino-5-phosphonovalerate (APV), MK801 and ketamine, as well as by desmethylimipramine and dextromethorphan. Desipramine 155-174 carbonic anhydrase 2 Gallus gallus 5-8 2162547-1 1990 The growth hormone (GH) response to desipramine was measured in ten patients meeting criteria for post-stroke depression (PSD), eight age-matched post-stroke (PS) non-depressed patients and eight healthy controls. Desipramine 36-47 growth hormone 1 Homo sapiens 4-18 2286698-5 1990 TRIM 4-week totals were significantly associated with total desipramine plus hydroxydesipramine plasma concentrations, r = -0.32, p less than 0.05, but HAM-D and MADS scores were not. Desipramine 60-71 tripartite motif containing 4 Homo sapiens 0-6 2164943-1 1990 Both 3-day and 15-day desipramine treatments (10 mg/kg, once daily) significantly reduced beta 1- but not beta 2-adrenoceptor and isoproterenol-stimulated adenylyl cyclase activities in rat cortical, hippocampal and amygdaloid membrane preparations. Desipramine 22-33 adrenoceptor beta 2 Rattus norvegicus 106-125 2095944-0 1990 Responses of growth hormone to desipramine in endogenous and non-endogenous depression. Desipramine 31-42 growth hormone 1 Homo sapiens 13-27 2095944-1 1990 Desipramine, a monoamine reuptake inhibitor, was used to stimulate release of growth hormone (GH) in 29 DSM-III major depressives and in 10 healthy controls. Desipramine 0-11 growth hormone 1 Homo sapiens 78-92 2139187-0 1990 Intrathecal administration of thiorphan, bestatin, desipramine and fluoxetine differentially potentiate the antinociceptive effects induced by beta-endorphin and morphine, administered intracerebroventricularly. Desipramine 51-62 pro-opiomelanocortin-alpha Mus musculus 143-157 2162547-1 1990 The growth hormone (GH) response to desipramine was measured in ten patients meeting criteria for post-stroke depression (PSD), eight age-matched post-stroke (PS) non-depressed patients and eight healthy controls. Desipramine 36-47 growth hormone 1 Homo sapiens 20-22 2176515-0 1990 The effect of reserpine, desipramine and thyroid hormone on alpha 1a- and alpha 1b-adrenoceptor binding sites: evidence for a subtype-specific regulation. Desipramine 25-36 adrenoceptor alpha 1A Rattus norvegicus 60-95 2207447-6 1990 Intracerebroventricular administration of the beta-2 adrenoceptor agonist salbutamol (1 microgram/rat) to rats pretreated with desipramine for 7 days, 15 min before immobilization, significantly relieved the inhibition by desipramine 5 and 10 min after the initiation of stress but the effect was not demonstrable after 20 and 40 min. Desipramine 127-138 adrenoceptor beta 2 Rattus norvegicus 46-65 2207447-6 1990 Intracerebroventricular administration of the beta-2 adrenoceptor agonist salbutamol (1 microgram/rat) to rats pretreated with desipramine for 7 days, 15 min before immobilization, significantly relieved the inhibition by desipramine 5 and 10 min after the initiation of stress but the effect was not demonstrable after 20 and 40 min. Desipramine 222-233 adrenoceptor beta 2 Rattus norvegicus 46-65 2170253-11 1990 Many classical pharmacological agents including guanethidine, clonidine, yohimbine, angiotensin II, nicotine and desipramine influence NPY release. Desipramine 113-124 neuropeptide Y Homo sapiens 135-138 1690278-3 1990 The activity of the enzyme marker system, glucose-6-phosphate dehydrogenase covalently conjugated to desipramine, is monitored by colorimetric detection of the rate of NADH formation at 340 nM. Desipramine 101-112 glucose-6-phosphate dehydrogenase Homo sapiens 42-75 2172531-1 1990 Desipramine, the monoamine reuptake inhibitor, acts predominantly on noradrenergic neurones, and via alpha-2 receptors brings about the release of growth hormone in normal healthy subjects. Desipramine 0-11 growth hormone 1 Homo sapiens 147-161 2398522-4 1990 A good correlation between the results of patient serum containing imipramine and desipramine simultaneously analyzed by ADx and gas liquid chromatography (GC) was observed, r2 = 0.964, n = 32. Desipramine 82-93 ferredoxin 1 Homo sapiens 121-124 2179970-3 1990 Studies with various receptor blockers have shown varying influences on the DMI-induced growth hormone, prolactin, and ACTH/cortisol secretion. Desipramine 76-79 proopiomelanocortin Homo sapiens 119-123 2248060-6 1990 This effect was almost completely eliminated by desipramine, suggesting that coadministration of a norepinephrine uptake inhibitor with a reversible MAO inhibitor is likely to reduce the risk of tyramine-induced hypertensive crisis. Desipramine 48-59 monoamine oxidase A Rattus norvegicus 149-152 2101962-0 1990 Influence of clenbuterol, a beta-adrenergic agonist, on desipramine induced growth hormone, prolactin and cortisol stimulation. Desipramine 56-67 amyloid beta precursor protein Homo sapiens 26-32 2101962-0 1990 Influence of clenbuterol, a beta-adrenergic agonist, on desipramine induced growth hormone, prolactin and cortisol stimulation. Desipramine 56-67 growth hormone 1 Homo sapiens 76-90 2101962-0 1990 Influence of clenbuterol, a beta-adrenergic agonist, on desipramine induced growth hormone, prolactin and cortisol stimulation. Desipramine 56-67 prolactin Homo sapiens 92-101 2101962-1 1990 We report herein the effects of the beta-adrenergic agonist clenbuterol on desipramine (DMI)-induced growth hormone (GH), prolactin (PRL) and cortisol secretion in healthy male subjects. Desipramine 75-86 growth hormone 1 Homo sapiens 101-115 2101962-1 1990 We report herein the effects of the beta-adrenergic agonist clenbuterol on desipramine (DMI)-induced growth hormone (GH), prolactin (PRL) and cortisol secretion in healthy male subjects. Desipramine 88-91 growth hormone 1 Homo sapiens 101-115 34597845-3 2021 In this study, we investigated the effects of heat stress and gestational status on ghrelin secretion and its possible associations with DMI in Holstein cattle. Desipramine 137-140 ghrelin and obestatin prepropeptide Bos taurus 84-91 2129310-2 1990 Subcutaneous administration of TRH reduced the total immobility time of rats tested in the despair (constrained swim) test and potentiated the anti-immobility effect of intraperitoneally administered desimipramine (DMI). Desipramine 200-213 thyrotropin releasing hormone Rattus norvegicus 31-34 2129310-2 1990 Subcutaneous administration of TRH reduced the total immobility time of rats tested in the despair (constrained swim) test and potentiated the anti-immobility effect of intraperitoneally administered desimipramine (DMI). Desipramine 215-218 thyrotropin releasing hormone Rattus norvegicus 31-34 2129310-4 1990 Hyperprolactinemia induced by pituitary homografts under the kidney capsule and the intracerebroventricular injection of PRL also potentiated the DMI-induced reduction of total immobility time of rats in the despair test and exerted "antidepressant" effects in aged rats. Desipramine 146-149 prolactin Rattus norvegicus 121-124 33945102-9 2021 Imipramine, desipramine, and fluoxetine suppress the production of IL-1beta, CCL2, as well as the expression of ICAM-1. Desipramine 12-23 interleukin 1 alpha Homo sapiens 67-75 33945102-9 2021 Imipramine, desipramine, and fluoxetine suppress the production of IL-1beta, CCL2, as well as the expression of ICAM-1. Desipramine 12-23 C-C motif chemokine ligand 2 Homo sapiens 77-81 33945102-9 2021 Imipramine, desipramine, and fluoxetine suppress the production of IL-1beta, CCL2, as well as the expression of ICAM-1. Desipramine 12-23 intercellular adhesion molecule 1 Homo sapiens 112-118 34965503-3 2022 Herein, expanding the previous findings on BDNFVal66Met polymorphism, we show that desipramine, a norepinephrine reuptake-inhibitor, rescues behavioral impairments, reduces the arterial thrombosis risk, abolishes pathological coagulation and platelet hyper-reactivity, normalizes leukocyte, platelet, and bone marrow megakaryocyte number and restores physiological norepinephrine levels in homozygous knock-in BDNF Val66Met (BDNFMet/Met) mice. Desipramine 83-94 brain derived neurotrophic factor Mus musculus 410-414 34969174-7 2022 The ASMase inhibitor desipramine decreased the level of S1P and the expression of alpha-smooth muscle actin (alpha-SMA) and NLRP3 in SiO2 dust-stained HBE cells, whereas the expression of E-cadherin (E-cad) increased. Desipramine 21-32 sphingomyelin phosphodiesterase 1 Homo sapiens 4-10 34969174-7 2022 The ASMase inhibitor desipramine decreased the level of S1P and the expression of alpha-smooth muscle actin (alpha-SMA) and NLRP3 in SiO2 dust-stained HBE cells, whereas the expression of E-cadherin (E-cad) increased. Desipramine 21-32 actin alpha 1, skeletal muscle Homo sapiens 109-118 34969174-7 2022 The ASMase inhibitor desipramine decreased the level of S1P and the expression of alpha-smooth muscle actin (alpha-SMA) and NLRP3 in SiO2 dust-stained HBE cells, whereas the expression of E-cadherin (E-cad) increased. Desipramine 21-32 NLR family pyrin domain containing 3 Homo sapiens 124-129 2550266-8 1989 Under our conditions the maximal effect occurred at 1 microM cAMP, revealing increased 32P incorporation in microtubule-associated protein 2 from a crude microtubule preparation obtained from the cerebral cortex of rats treated with desmethylimipramine. Desipramine 233-252 microtubule-associated protein 2 Rattus norvegicus 108-140 34337165-7 2021 Mechanistically, DMI treatment during reperfusion increased the expression level of the microtubule-associated protein 1A/1B-light chain 3 (LC3) processing enzymes, ATG4B and ATG7. Desipramine 17-20 annexin A3 Rattus norvegicus 140-143 34337165-7 2021 Mechanistically, DMI treatment during reperfusion increased the expression level of the microtubule-associated protein 1A/1B-light chain 3 (LC3) processing enzymes, ATG4B and ATG7. Desipramine 17-20 autophagy related 4B, cysteine peptidase Rattus norvegicus 165-170 34337165-7 2021 Mechanistically, DMI treatment during reperfusion increased the expression level of the microtubule-associated protein 1A/1B-light chain 3 (LC3) processing enzymes, ATG4B and ATG7. Desipramine 17-20 autophagy related 7 Rattus norvegicus 175-179 34337165-8 2021 Genetic knockdown of ATG4B abolished the cytoprotective effect of DMI. Desipramine 66-69 autophagy related 4B, cysteine peptidase Rattus norvegicus 21-26 34337165-9 2021 Together, these results indicate that DMI is a unique agent which enhances LC3 processing in an ATG4B-dependent way. Desipramine 38-41 annexin A3 Rattus norvegicus 75-78 34337165-9 2021 Together, these results indicate that DMI is a unique agent which enhances LC3 processing in an ATG4B-dependent way. Desipramine 38-41 autophagy related 4B, cysteine peptidase Rattus norvegicus 96-101 34232916-13 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 leptin Mus musculus 34-40 34232916-13 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 fatty acid binding protein 4, adipocyte Mus musculus 42-47 34232916-13 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 sterol regulatory element binding transcription factor 1 Mus musculus 49-56 34232916-13 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 peroxisome proliferator activated receptor gamma Mus musculus 58-67 34232916-13 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 CCAAT/enhancer binding protein (C/EBP), alpha Mus musculus 73-83 34232916-29 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 leptin Mus musculus 34-40 34232916-29 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 fatty acid binding protein 4, adipocyte Mus musculus 42-47 34232916-29 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 sterol regulatory element binding transcription factor 1 Mus musculus 49-56 34232916-29 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 peroxisome proliferator activated receptor gamma Mus musculus 58-67 34232916-29 2021 The elevated expression levels of Leptin, FABP4, SREBP1C, PPARgamma, and C/EBPalpha induced by the stimulation with DMI incubation were dramatically inhibited by the introduction of TSA, accompanied by the upregulation of phosphorylated AMP-activated protein kinase (p-AMPK). Desipramine 116-119 CCAAT/enhancer binding protein (C/EBP), alpha Mus musculus 73-83 34062902-7 2021 In contrast to their direct antagonistic effects, prolonged incubation with IMI and DMI increased the maximal response of the alpha1B-AR subtype, and the CIT of both the alpha1A- and the alpha1B-ARs. Desipramine 84-87 alpha-1-B glycoprotein Homo sapiens 126-133 34062902-7 2021 In contrast to their direct antagonistic effects, prolonged incubation with IMI and DMI increased the maximal response of the alpha1B-AR subtype, and the CIT of both the alpha1A- and the alpha1B-ARs. Desipramine 84-87 serpin family A member 1 Homo sapiens 170-177 34062902-7 2021 In contrast to their direct antagonistic effects, prolonged incubation with IMI and DMI increased the maximal response of the alpha1B-AR subtype, and the CIT of both the alpha1A- and the alpha1B-ARs. Desipramine 84-87 alpha-1-B glycoprotein Homo sapiens 187-194 35210489-5 2022 Our ex vivo efflux experiments reveal that the NE transporter (NET) blocker desipramine elevates both norepinephrine (NE) and dopamine (DA), but not 5-HT levels, in PFC tissue slices from wild-type (WT) and DAT-KO, but not NET-KO mice. Desipramine 76-87 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 47-61 35210489-5 2022 Our ex vivo efflux experiments reveal that the NE transporter (NET) blocker desipramine elevates both norepinephrine (NE) and dopamine (DA), but not 5-HT levels, in PFC tissue slices from wild-type (WT) and DAT-KO, but not NET-KO mice. Desipramine 76-87 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 207-210 35210489-7 2022 Systemic administration of desipramine or fluoxetine inhibits hyperactivity in DAT-KO mice, whereas local PFC infusion of desipramine alone produced this same effect. Desipramine 27-38 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 79-82 35210489-7 2022 Systemic administration of desipramine or fluoxetine inhibits hyperactivity in DAT-KO mice, whereas local PFC infusion of desipramine alone produced this same effect. Desipramine 122-133 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 79-82 35015730-6 2022 Combined treatment with desipramine and L-cycloserine (DC) lowered ceramide levels and exerted a protective effect on photoreceptors in AdipoR1-/- mice. Desipramine 24-35 adiponectin receptor 1 Mus musculus 136-143 35242795-7 2022 Furthermore, DMI, the CLA was significantly greater (P < 0.05) in the CA2 group than the other groups. Desipramine 13-16 carbonic anhydrase 2 Bos taurus 70-73 2533079-9 1989 It thus appears that 5-HT1 receptors in the VTA, presumably of the 5-HT1B type, act by preventing the anti-immobility effect of DMI. Desipramine 128-131 5-hydroxytryptamine receptor 1B Rattus norvegicus 67-73 34599336-9 2021 High allowance increased herbage mass and sites with greater canopy height which allow greater DMI, positively associated with cow BCS at weaning, calf ADG, BWW, and g of calf/kg DMI. Desipramine 95-98 ADG Bos taurus 152-155 2529959-4 1989 Analyses of the differences between GMI and DMI indicated that rapid forgetting was more apparent for DAT than for HD patients in the early stages of these disorders. Desipramine 44-47 solute carrier family 6 member 3 Homo sapiens 102-105 2553044-1 1989 We examined the effects of chronic treatment with antidepressants (imipramine or desipramine) or benzodiazepines (diazepam, alprazolam, or adinazolam) on modulation of corticotropin-releasing-factor (CRF) receptors in discrete areas of rat brain and in anterior pituitary. Desipramine 81-92 corticotropin releasing hormone Rattus norvegicus 168-198 2725706-3 1989 When both MAO and COMT were intact, most of the metabolism of 3H-noradrenaline was neuronal (i.e., desipramine-sensitive). Desipramine 99-110 monoamine oxidase A Rattus norvegicus 10-13 2725706-3 1989 When both MAO and COMT were intact, most of the metabolism of 3H-noradrenaline was neuronal (i.e., desipramine-sensitive). Desipramine 99-110 catechol-O-methyltransferase Rattus norvegicus 18-22 2566928-8 1989 Uptake1 blockade with desipramine (100 nM) or nisoxetine (100 nM) prior to the second stimulation significantly increased noradrenaline overflow and attenuated that of NPY; the attenuation of the stimulation-evoked overflow of NPY was abolished by yohimbine (1 microM). Desipramine 22-33 pro-neuropeptide Y Cavia porcellus 168-171 2496890-7 1989 Only rats injected with 6-OHDA without DMI pretreatment showed a significant reduction in ANG II-induced drinking responses. Desipramine 39-42 angiotensinogen Rattus norvegicus 90-96 2568728-14 1989 Pretreatment with PBZ significantly increased the SNS-evoked NA and NPY-LI overflow, while DMI enhanced both the functional response and NA release but reduced the NPY-LI overflow. Desipramine 91-94 neuropeptide Y Sus scrofa 164-167 2566928-8 1989 Uptake1 blockade with desipramine (100 nM) or nisoxetine (100 nM) prior to the second stimulation significantly increased noradrenaline overflow and attenuated that of NPY; the attenuation of the stimulation-evoked overflow of NPY was abolished by yohimbine (1 microM). Desipramine 22-33 pro-neuropeptide Y Cavia porcellus 227-230 3225754-7 1988 These data strongly suggest that the hydroxylation pathways of imipramine and desipramine and the demethylation pathways of imipramine and 2-hydroxyimipramine are each sharing the same species of cytochrome P-450. Desipramine 78-89 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 196-212 2615927-3 1989 The main findings were (1) sex differences in the growth hormone and cortisol response to desipramine and (2) a significant genetic component of the prolactin and cortisol response to desipramine as indicated by significantly (p less than 0.05) lower within-pair than between-pair variance in the sibling pairs but not random pairs of the experimental subjects. Desipramine 90-101 growth hormone 1 Homo sapiens 50-64 2615927-3 1989 The main findings were (1) sex differences in the growth hormone and cortisol response to desipramine and (2) a significant genetic component of the prolactin and cortisol response to desipramine as indicated by significantly (p less than 0.05) lower within-pair than between-pair variance in the sibling pairs but not random pairs of the experimental subjects. Desipramine 184-195 prolactin Homo sapiens 149-158 2667015-0 1989 Desipramine increases circulating growth hormone in elderly depressed patients: a pilot study. Desipramine 0-11 growth hormone 1 Homo sapiens 34-48 2975304-0 1988 Growth hormone response to desmethylimipramine in depressed and suicidal adolescents. Desipramine 27-46 growth hormone 1 Homo sapiens 0-14 3418509-0 1988 Effects of high dose alpha-1-acid glycoprotein on desipramine toxicity in rats. Desipramine 50-61 orosomucoid 1 Rattus norvegicus 21-46 3418509-9 1988 The serum DMI concentration did not change with albumin infusion but increased 4.7-fold (1.9 +/- 0.7 to 9.0 +/- 1.6 micrograms/ml) after AAG, 2.2 g/kg, and 6.7-fold (2.3 +/- 0.5 to 15.7 +/- 7.0 micrograms/ml) after AAG, 4.4 g/kg. Desipramine 10-13 orosomucoid 1 Rattus norvegicus 137-140 3418509-9 1988 The serum DMI concentration did not change with albumin infusion but increased 4.7-fold (1.9 +/- 0.7 to 9.0 +/- 1.6 micrograms/ml) after AAG, 2.2 g/kg, and 6.7-fold (2.3 +/- 0.5 to 15.7 +/- 7.0 micrograms/ml) after AAG, 4.4 g/kg. Desipramine 10-13 orosomucoid 1 Rattus norvegicus 215-218 2828545-11 1988 Decreasing 5-HT2 function by twice-a-day injection of 5 mg/kg of zimeldine or desipramine (DMI) produced 50% (p less than 0.01) and 56% (p less than 0.01), respectively, reductions in head-twitch behaviour. Desipramine 78-89 hypothermia due to alcohol sensitivity 2 Mus musculus 13-16 3132531-0 1988 Possible involvement of pertussis toxin substrates (Gi, Go) in desipramine-induced refractoriness of adenylate cyclase in cerebral cortices of rats. Desipramine 63-74 RAS like proto-oncogene B Rattus norvegicus 52-54 2828545-11 1988 Decreasing 5-HT2 function by twice-a-day injection of 5 mg/kg of zimeldine or desipramine (DMI) produced 50% (p less than 0.01) and 56% (p less than 0.01), respectively, reductions in head-twitch behaviour. Desipramine 91-94 hypothermia due to alcohol sensitivity 2 Mus musculus 13-16 3427414-0 1987 The modulatory effect of neurotensin on [3H]dopamine release from rat nucleus accumbens slices is enhanced after chronic desipramine treatment. Desipramine 121-132 neurotensin Rattus norvegicus 25-36 3427414-4 1987 Superfusion of the DMI-treated rat tissue with buffer-containing neurotensin (100 nM) resulted in an increase in the K+-evoked release of [3H]DA to a level which was higher than that observed with tissue obtained from untreated rats. Desipramine 19-22 neurotensin Rattus norvegicus 65-76 3427414-6 1987 These findings are consistent with DMI treatment giving rise to impaired DA release possibly due to DA autoreceptor-mediated inhibition of DA release and cancellation of this effect by neurotensin. Desipramine 35-38 neurotensin Rattus norvegicus 185-196 2887701-3 1987 Subchronic infusion of desipramine, a noradrenaline uptake inhibitor, effected a decrease in neocortical and hippocampal beta 1-adrenoceptor density. Desipramine 23-34 adrenoceptor beta 1 Rattus norvegicus 121-140 3108675-6 1987 Desipramine suppressed the NPY-LI release induced by tyramine apparently by the inhibition of the uptake of tyramine is suggested by the significant positive correlation between the reduction of 14C-tyramine uptake and the inhibition of NPY-LI release induced by desipramine (r = 0.946). Desipramine 0-11 neuropeptide Y Rattus norvegicus 27-30 3108675-6 1987 Desipramine suppressed the NPY-LI release induced by tyramine apparently by the inhibition of the uptake of tyramine is suggested by the significant positive correlation between the reduction of 14C-tyramine uptake and the inhibition of NPY-LI release induced by desipramine (r = 0.946). Desipramine 0-11 neuropeptide Y Rattus norvegicus 237-240 3108675-6 1987 Desipramine suppressed the NPY-LI release induced by tyramine apparently by the inhibition of the uptake of tyramine is suggested by the significant positive correlation between the reduction of 14C-tyramine uptake and the inhibition of NPY-LI release induced by desipramine (r = 0.946). Desipramine 263-274 neuropeptide Y Rattus norvegicus 27-30 3108675-6 1987 Desipramine suppressed the NPY-LI release induced by tyramine apparently by the inhibition of the uptake of tyramine is suggested by the significant positive correlation between the reduction of 14C-tyramine uptake and the inhibition of NPY-LI release induced by desipramine (r = 0.946). Desipramine 263-274 neuropeptide Y Rattus norvegicus 237-240 2882523-4 1987 After chronic administration of tricyclic antidepressants (imipramine and desipramine) the number of cortical beta 1 adrenergic receptors decreased without impairing the clenbuterol-induced decrease in spontaneous motor activity. Desipramine 74-85 hemoglobin, beta adult major chain Mus musculus 110-116 3666058-0 1987 The influence of desmethylimipramine on the hypothalamic and neurohypophysial vasopressin content in pinealectomized male rats. Desipramine 17-36 arginine vasopressin Rattus norvegicus 78-89 3666058-1 1987 The effect of desmethylimipramine (DMI) on the hypothalamic and neurohypophysial vasopressin content was investigated in normal and pinealectomized male rats. Desipramine 14-33 arginine vasopressin Rattus norvegicus 81-92 3666058-1 1987 The effect of desmethylimipramine (DMI) on the hypothalamic and neurohypophysial vasopressin content was investigated in normal and pinealectomized male rats. Desipramine 35-38 arginine vasopressin Rattus norvegicus 81-92 3666058-4 1987 injection of DMI (20 micrograms; dissolved in 10 microliters of normal saline) decreased vasopressin content in the neurohypophysis of non-operated and sham-operated animals. Desipramine 13-16 arginine vasopressin Rattus norvegicus 89-100 3628616-3 1987 Clinical studies have suggested that sleep deprivation could potentiate the therapeutic effects of antidepressants, and there were reports on DMI stimulation of GH. Desipramine 142-145 growth hormone 1 Homo sapiens 161-163 3628616-5 1987 The GH levels increased substantially (8- to 10-fold) after DMI in both experimental conditions. Desipramine 60-63 growth hormone 1 Homo sapiens 4-6 3628616-6 1987 Sleep deprivation neither changed GH basal levels nor potentiated the DMI-induced GH increase. Desipramine 70-73 growth hormone 1 Homo sapiens 82-84 3598027-1 1987 Adverse reactions to the tricyclic antidepressant drugs imipramine and desipramine have been described and include eosinophilia, pulmonary infiltrates with eosinophilia, and elevated total serum IgE levels. Desipramine 71-82 immunoglobulin heavy constant epsilon Homo sapiens 195-198 3598027-3 1987 This article describes a patient manifesting bronchospasm, profound eosinophilia, and elevated serum IgE levels after therapy with desipramine that resolved rapidly after withdrawal of the drug. Desipramine 131-142 immunoglobulin heavy constant epsilon Homo sapiens 101-104 3595714-4 1987 There was a positive correlation between the inhibitory effect of desipramine on the NPY release and on the uptake of [14C]tyramine into synaptosomes. Desipramine 66-77 neuropeptide Y Oryctolagus cuniculus 85-88 3595714-6 1987 These data suggest that tyramine enters into nerve terminals through a desipramine-sensitive mechanism, resulting in the co-release of NE and NPY which can be reduced by guanethidine. Desipramine 71-82 neuropeptide Y Oryctolagus cuniculus 142-145 3101979-0 1987 Chronic administration of desipramine or nialamide decreases wet-dog shakes in rats produced by the TRH-analog MK-771. Desipramine 26-37 thyrotropin releasing hormone Rattus norvegicus 100-103 3101979-1 1987 The effect of chronic administration of the tricyclic antidepressant, desipramine, or the monoamine oxidase inhibitor (MAOI), nialamide, on the ability of the TRH analog, MK-771, to induce wet-dog shakes in rats was examined. Desipramine 70-81 TRH Canis lupus familiaris 159-162 2455193-13 1987 The NA uptake inhibitor desipramine reduces NPY release. Desipramine 24-35 neuropeptide Y Homo sapiens 44-47 3769385-4 1986 For desipramine the apparent oral clearance (L X min-1) was 0.19 (0.12 to 0.24) in PM compared with 1.64 (1.46 to 1.80) and 1.03 (0.77 to 1.13) in rapid EM and slow EM. Desipramine 4-15 CD59 molecule (CD59 blood group) Homo sapiens 49-54 2872321-3 1986 Prolonged administration of desipramine and citalopram, but not amitriptyline, elevated the microsomal level of cytochrome P-450 and accelerated the rate of ethylmorphine demethylation. Desipramine 28-39 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 112-128 2418913-3 1986 In this study, we have determined the effect of neonatal 5,7-DHT and desipramine (DMI) treatment on the biochemical content and immunocytochemical localization of substance P and TRH throughout the brain. Desipramine 69-80 thyrotropin releasing hormone Rattus norvegicus 179-182 2874504-8 1986 co-administration of atenolol or practolol, beta 1-adrenoceptor antagonists, together with DMI attenuated both beta-adrenoceptor down regulation and the behavioural activation by chronic DMI. Desipramine 187-190 adrenoceptor beta 1 Rattus norvegicus 44-63 2418913-3 1986 In this study, we have determined the effect of neonatal 5,7-DHT and desipramine (DMI) treatment on the biochemical content and immunocytochemical localization of substance P and TRH throughout the brain. Desipramine 82-85 thyrotropin releasing hormone Rattus norvegicus 179-182 3028722-5 1986 The alpha 2-mediated growth hormone response to clonidine was increased after one week"s treatment with desipramine and then reduced during the second and third weeks of treatment. Desipramine 104-115 growth hormone 1 Homo sapiens 21-35 3709653-3 1986 2-hydroxylation of DMI is probably mediated by debrisoquine hydroxylase, a cytochrome P-450 isozyme that is monogenically controlled. Desipramine 19-22 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 75-91 3005903-4 1986 Since the inhibitors of the uptake of noradrenaline, desipramine and CPP 199 antagonized this preference for noradrenergic MAO it is concluded that these MAO inhibitors are accumulated in the noradrenergic neurones by the membranal uptake carrier. Desipramine 53-64 monoamine oxidase A Rattus norvegicus 123-126 3005903-4 1986 Since the inhibitors of the uptake of noradrenaline, desipramine and CPP 199 antagonized this preference for noradrenergic MAO it is concluded that these MAO inhibitors are accumulated in the noradrenergic neurones by the membranal uptake carrier. Desipramine 53-64 monoamine oxidase A Rattus norvegicus 154-157 3031716-2 1986 In this report the effects of various receptor blockers on desimipramine (DMI)-induced growth hormone (GH) secretion in healthy male subjects are presented. Desipramine 59-72 growth hormone 1 Homo sapiens 87-101 3031716-2 1986 In this report the effects of various receptor blockers on desimipramine (DMI)-induced growth hormone (GH) secretion in healthy male subjects are presented. Desipramine 59-72 growth hormone 1 Homo sapiens 103-105 3031716-2 1986 In this report the effects of various receptor blockers on desimipramine (DMI)-induced growth hormone (GH) secretion in healthy male subjects are presented. Desipramine 74-77 growth hormone 1 Homo sapiens 87-101 3031716-2 1986 In this report the effects of various receptor blockers on desimipramine (DMI)-induced growth hormone (GH) secretion in healthy male subjects are presented. Desipramine 74-77 growth hormone 1 Homo sapiens 103-105 3031716-6 1986 DMI-induced GH stimulation was not significantly different after DMI i.v. Desipramine 0-3 growth hormone 1 Homo sapiens 12-14 3031716-10 1986 ), GH secretion was significantly increased by 25 mg DMI (p less than 0.05) and 50 mg DMI (incomplete block design, n = 18). Desipramine 53-56 growth hormone 1 Homo sapiens 3-5 3031716-10 1986 ), GH secretion was significantly increased by 25 mg DMI (p less than 0.05) and 50 mg DMI (incomplete block design, n = 18). Desipramine 86-89 growth hormone 1 Homo sapiens 3-5 3031716-11 1986 GH secretion was significantly lower (p less than 0.01) after DMI in combination with 60 mg phentolamine i.v. Desipramine 62-65 growth hormone 1 Homo sapiens 0-2 3031716-15 1986 The DMI-induced GH increase following DMI plus 1 mg prazosin p.o. Desipramine 4-7 growth hormone 1 Homo sapiens 16-18 3031716-15 1986 The DMI-induced GH increase following DMI plus 1 mg prazosin p.o. Desipramine 38-41 growth hormone 1 Homo sapiens 16-18 3031716-17 1986 The results indicate that the GH-stimulating effect of DMI is primarily related to the ability of DMI to inhibit noradrenaline (NA) reuptake. Desipramine 55-58 growth hormone 1 Homo sapiens 30-32 3031716-17 1986 The results indicate that the GH-stimulating effect of DMI is primarily related to the ability of DMI to inhibit noradrenaline (NA) reuptake. Desipramine 98-101 growth hormone 1 Homo sapiens 30-32 3739365-5 1986 By monitoring the hydroxylation of DMI in various fractions during separation and purification of cytochrome P-450 from human liver microsomes we have purified a cytochrome P-450 which efficiently hydroxylates this drug. Desipramine 35-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 98-114 3739365-5 1986 By monitoring the hydroxylation of DMI in various fractions during separation and purification of cytochrome P-450 from human liver microsomes we have purified a cytochrome P-450 which efficiently hydroxylates this drug. Desipramine 35-38 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 162-178 3160963-7 1985 Pretreatment with the blocker of the uptake of norepinephrine, desipramine did not prevent the initial (30 min) effect but completely prevented the delayed (4 hr) effect of fenfluramine on plasma renin activity. Desipramine 63-74 renin Rattus norvegicus 196-201 3031718-2 1986 In this report the effects of various receptor blockers on the desimipramine (DMI)-induced cortisol (ACTH) secretion in healthy male subjects are presented. Desipramine 63-76 proopiomelanocortin Homo sapiens 101-105 3031718-2 1986 In this report the effects of various receptor blockers on the desimipramine (DMI)-induced cortisol (ACTH) secretion in healthy male subjects are presented. Desipramine 78-81 proopiomelanocortin Homo sapiens 101-105 3031718-6 1986 In addition, the effect of prazosin on DMI-induced ACTH stimulation was examined. Desipramine 39-42 proopiomelanocortin Homo sapiens 51-55 3031718-15 1986 n = 12), as was DMI-induced ACTH secretion (p less than 0.05) in these subjects. Desipramine 16-19 proopiomelanocortin Homo sapiens 28-32 3031718-16 1986 The findings of these trials, especially those of the prazosin trial, indicate that DMI-induced stimulation of cortisol and ACTH secretion is attributable to the noradrenaline (NA) reuptake inhibiting effect of DMI, and that the stimulus is transmitted with the aid of noradrenergic alpha-1 receptors. Desipramine 84-87 proopiomelanocortin Homo sapiens 124-128 3031718-16 1986 The findings of these trials, especially those of the prazosin trial, indicate that DMI-induced stimulation of cortisol and ACTH secretion is attributable to the noradrenaline (NA) reuptake inhibiting effect of DMI, and that the stimulus is transmitted with the aid of noradrenergic alpha-1 receptors. Desipramine 84-87 adrenoceptor alpha 1D Homo sapiens 283-290 3031718-16 1986 The findings of these trials, especially those of the prazosin trial, indicate that DMI-induced stimulation of cortisol and ACTH secretion is attributable to the noradrenaline (NA) reuptake inhibiting effect of DMI, and that the stimulus is transmitted with the aid of noradrenergic alpha-1 receptors. Desipramine 211-214 proopiomelanocortin Homo sapiens 124-128 3928818-9 1985 DMI pretreatment blocked the increase in hippocampal TRH, but not in brainstem TRH. Desipramine 0-3 thyrotropin releasing hormone Rattus norvegicus 53-56 3031716-18 1986 Should serotonergic receptors be involved in the DMI-induced GH secretion at all, they transmit a positive stimulus. Desipramine 49-52 growth hormone 1 Homo sapiens 61-63 3031717-2 1986 In this report the effects of various receptor blockers on desimipramine (DMI)-induced prolactin (PRL) secretion in healthy male subjects are presented. Desipramine 59-72 prolactin Homo sapiens 87-96 3031717-2 1986 In this report the effects of various receptor blockers on desimipramine (DMI)-induced prolactin (PRL) secretion in healthy male subjects are presented. Desipramine 74-77 prolactin Homo sapiens 87-96 6098913-5 1984 DMI acutely increased plasma PRL, whereas ZIM had an effect only after chronic pretreatment. Desipramine 0-3 prolactin Homo sapiens 29-32 4092710-0 1985 Growth hormone release after desipramine in depressive illness. Desipramine 29-40 growth hormone 1 Homo sapiens 0-14 4092710-2 1985 administration of 75 mg of desipramine on growth hormone (GH) secretion was investigated in a sample of 87 patients with major depressive disorders and in 31 normal controls. Desipramine 27-38 growth hormone 1 Homo sapiens 42-56 4092710-2 1985 administration of 75 mg of desipramine on growth hormone (GH) secretion was investigated in a sample of 87 patients with major depressive disorders and in 31 normal controls. Desipramine 27-38 growth hormone 1 Homo sapiens 58-60 4001280-2 1985 In previous studies it was shown that the tricyclic antidepressant desimipramine (DMI) had different stimulatory effects on growth hormone (GH), prolactin (PRL), ACTH and cortisol secretion in healthy subjects, depending on the mode of administration. Desipramine 67-80 growth hormone 1 Homo sapiens 124-138 4001280-2 1985 In previous studies it was shown that the tricyclic antidepressant desimipramine (DMI) had different stimulatory effects on growth hormone (GH), prolactin (PRL), ACTH and cortisol secretion in healthy subjects, depending on the mode of administration. Desipramine 67-80 prolactin Homo sapiens 145-154 4001280-2 1985 In previous studies it was shown that the tricyclic antidepressant desimipramine (DMI) had different stimulatory effects on growth hormone (GH), prolactin (PRL), ACTH and cortisol secretion in healthy subjects, depending on the mode of administration. Desipramine 67-80 prolactin Homo sapiens 156-159 4001280-2 1985 In previous studies it was shown that the tricyclic antidepressant desimipramine (DMI) had different stimulatory effects on growth hormone (GH), prolactin (PRL), ACTH and cortisol secretion in healthy subjects, depending on the mode of administration. Desipramine 67-80 proopiomelanocortin Homo sapiens 162-166 4001280-2 1985 In previous studies it was shown that the tricyclic antidepressant desimipramine (DMI) had different stimulatory effects on growth hormone (GH), prolactin (PRL), ACTH and cortisol secretion in healthy subjects, depending on the mode of administration. Desipramine 82-85 growth hormone 1 Homo sapiens 124-138 4001280-2 1985 In previous studies it was shown that the tricyclic antidepressant desimipramine (DMI) had different stimulatory effects on growth hormone (GH), prolactin (PRL), ACTH and cortisol secretion in healthy subjects, depending on the mode of administration. Desipramine 82-85 prolactin Homo sapiens 145-154 4001280-2 1985 In previous studies it was shown that the tricyclic antidepressant desimipramine (DMI) had different stimulatory effects on growth hormone (GH), prolactin (PRL), ACTH and cortisol secretion in healthy subjects, depending on the mode of administration. Desipramine 82-85 prolactin Homo sapiens 156-159 4001280-2 1985 In previous studies it was shown that the tricyclic antidepressant desimipramine (DMI) had different stimulatory effects on growth hormone (GH), prolactin (PRL), ACTH and cortisol secretion in healthy subjects, depending on the mode of administration. Desipramine 82-85 proopiomelanocortin Homo sapiens 162-166 4001280-6 1985 Compared to placebo, significant increases occurred in GH (p less than 0.05) and in PRL (p less than 0.05) from a dose of DMI 25 mg on, and in cortisol (p less than 0.05) from 15 mg on. Desipramine 122-125 prolactin Homo sapiens 84-87 2937389-2 1985 In rat but not in mouse thymocytes the ecto-ATPase was inhibited in vitro by HgCl2, tricyclohexyltin chloride, di-n-butyltin dichloride, DDT, gamma HCH and desipramine. Desipramine 156-167 CEA cell adhesion molecule 1 Rattus norvegicus 39-50 2857613-1 1985 In the present studies the effects of desipramine on the functional sensitivity of the prejunctional alpha 2-adrenoceptors of the vas deferens were studied in pithed rats; contractions of the tissue were evoked by electrical stimulation (6 Hz) of the spinal sympathetic outflow. Desipramine 38-49 arginine vasopressin Rattus norvegicus 130-133 2857613-4 1985 In acute studies desipramine (0.3-4.3 mg/kg, intravenously) inhibited electrically induced contractions of the vas deferens of pithed rats. Desipramine 17-28 arginine vasopressin Rattus norvegicus 111-114 6098913-6 1984 Chronic DMI but not ZIM increased baseline PRL. Desipramine 8-11 prolactin Homo sapiens 43-46 6237705-7 1984 Desipramine given orally (27 mg kg-1 per day, 14 days) decreased both the behaviour and number of 5-HT2 binding sites. Desipramine 0-11 hypothermia due to alcohol sensitivity 2 Mus musculus 100-103 6087965-4 1984 By contrast, the growth hormone response to clonidine tended to be increased after one week of desipramine, reduced after three weeks of treatment, and further reduced after discontinuation. Desipramine 95-106 growth hormone 1 Homo sapiens 17-31 6703855-4 1984 Both desipramine (N = 7) and amitriptyline (N = 6) significantly increased the PRL rise induced by tryptophan compared with a preceding placebo period. Desipramine 5-16 prolactin Homo sapiens 79-82 6735152-3 1984 Similarly inhibition of NE reuptake by imipramine or desmethylimipramine were followed by reduced GH secretion. Desipramine 53-72 growth hormone 1 Homo sapiens 98-100 6703855-5 1984 In contrast, following long-term amitriptyline and desipramine treatment, the ability of tryptophan to increase PRL was enhanced two weeks following abrupt cessation of amitriptyline therapy (N = 5), but not after discontinuation of desipramine therapy. Desipramine 51-62 prolactin Homo sapiens 112-115 6703855-5 1984 In contrast, following long-term amitriptyline and desipramine treatment, the ability of tryptophan to increase PRL was enhanced two weeks following abrupt cessation of amitriptyline therapy (N = 5), but not after discontinuation of desipramine therapy. Desipramine 233-244 prolactin Homo sapiens 112-115 6693882-4 1984 In aorta and vas deferens, about 45% and 65%, respectively, of the total [3H]XYL accumulation was blocked by 100 mumol/L desmethylimipramine (DMI), l-NA, or bretylium. Desipramine 121-140 arginine vasopressin Rattus norvegicus 13-16 6141282-7 1984 The decrease and recovery of enzyme activity in the heart after administration of the reversible noradrenergic neurotoxin N-(2-chloroethyl)-N-ethyl-2-bromobenzylamine (DSP4) were parallel to the recovery of norepinephrine content and desmethylimipramine binding. Desipramine 234-253 dual specificity phosphatase 26 Homo sapiens 168-172 6693882-4 1984 In aorta and vas deferens, about 45% and 65%, respectively, of the total [3H]XYL accumulation was blocked by 100 mumol/L desmethylimipramine (DMI), l-NA, or bretylium. Desipramine 142-145 arginine vasopressin Rattus norvegicus 13-16 6098855-3 1984 Desimipramine therapy (50-100 mg/day per os for 3-5 weeks) induced in parallel psychological improvement and fall in beta LPH-beta EP in 6 of 8 PAD patients treated, and a normalization of beta EP-beta LPH levels with minimal mood improvement in the 2 schizoaffective subjects. Desipramine 0-13 proopiomelanocortin Homo sapiens 117-125 6098855-3 1984 Desimipramine therapy (50-100 mg/day per os for 3-5 weeks) induced in parallel psychological improvement and fall in beta LPH-beta EP in 6 of 8 PAD patients treated, and a normalization of beta EP-beta LPH levels with minimal mood improvement in the 2 schizoaffective subjects. Desipramine 0-13 proopiomelanocortin Homo sapiens 197-205 6093182-2 1984 Desimipramine (DMI) (50 mg IV) significantly stimulated ACTH secretion. Desipramine 0-13 proopiomelanocortin Homo sapiens 56-60 6093182-2 1984 Desimipramine (DMI) (50 mg IV) significantly stimulated ACTH secretion. Desipramine 15-18 proopiomelanocortin Homo sapiens 56-60 6420832-0 1984 Comparison of growth hormone and prolactin stimulation induced by chlorimipramine and desimipramine in man in connection with chlorimipramine metabolism. Desipramine 86-99 prolactin Homo sapiens 33-42 6420832-1 1984 In order to compare the effects of chlorimipramine (CI) and desimipramine (DMI) on growth hormone (GH) and on prolactin (PRL) secretion equal doses of 25 mg CI or DMI were administered i.v. Desipramine 75-78 growth hormone 1 Homo sapiens 83-97 6644379-2 1983 The label is incorporated by reaction of C-11 methyl iodide (11CH3I) upon desipramine in dimethylsulfoxide. Desipramine 74-85 aldo-keto reductase family 1 member C4 Homo sapiens 41-45 6646243-8 1983 Since pretreatment of the rats with norzimeldine or desipramine antagonized the preferences for serotonergic or noradrenergic MAO, it is plausible to conclude that the compounds showing these preferences are accumulated in the neurons by the membranal uptake systems. Desipramine 52-63 monoamine oxidase A Rattus norvegicus 126-129 6299918-3 1983 When in these patients brain norepinephrine receptor activity was increased by desipramine, the normally negative differential feedback mechanism was converted into a positive one (paradoxical ACTH response). Desipramine 79-90 proopiomelanocortin Homo sapiens 193-197 6888570-7 1983 The liver/intestine ratio of DMI and 2-OH-IMP formation is proportional to the cytochrome P-450 ratio in the microsomes, in contrast to N-oxide formation. Desipramine 29-32 cytochrome P450 3A14 Cavia porcellus 79-95 6130811-3 1983 The beta(2)-adrenoceptor agonist, terbutaline (5.0 mg kg(-1)), produced a moderate increase in pineal melatonin content.2 Repeated daily administration of desmethylimipramine (10 mg kg(-1) for 10 days) and maprotiline (10 mg kg(-1) for 10 days), antidepressants predominantly inhibiting noradrenaline (NA) uptake, reduced the isoprenaline-induced increase in pineal melatonin content. Desipramine 155-174 adrenoceptor beta 2 Rattus norvegicus 4-24 6840904-5 1983 There was a difference between the drug-free period and desmethylimipramine or imipramine on the PEPc (P less than 0.05) and the PEP/LVET ratio (P less than 0.05); on the R-R (P less than 0.05), PR (P less than 0.05), QRS (P less than 0.05), and QTc (P less than 0.05) intervals; but no difference on the LVETc or shortening fraction or the mean velocity of circumferential shortening. Desipramine 56-75 peptidase C Homo sapiens 97-101 6840904-5 1983 There was a difference between the drug-free period and desmethylimipramine or imipramine on the PEPc (P less than 0.05) and the PEP/LVET ratio (P less than 0.05); on the R-R (P less than 0.05), PR (P less than 0.05), QRS (P less than 0.05), and QTc (P less than 0.05) intervals; but no difference on the LVETc or shortening fraction or the mean velocity of circumferential shortening. Desipramine 56-75 progestagen associated endometrial protein Homo sapiens 97-100 6840904-6 1983 Drugs such as desmethylimipramine and imipramine which prolong intraventricular conduction can probably be expected to prolong the PEP and PEP/LVET. Desipramine 14-33 progestagen associated endometrial protein Homo sapiens 131-134 6840904-6 1983 Drugs such as desmethylimipramine and imipramine which prolong intraventricular conduction can probably be expected to prolong the PEP and PEP/LVET. Desipramine 14-33 progestagen associated endometrial protein Homo sapiens 139-142 6959166-1 1982 The effect of 4 to 6 weeks of treatment with the antidepressants, desmethylimipramine and amitriptyline, on the growth hormone response to clonidine was studied in 12 depressed patients. Desipramine 66-85 growth hormone 1 Homo sapiens 112-126 6126334-7 1982 These data suggest that DMI decreases both transport of 5-HT across the pulmonary endothelium and MAO activity. Desipramine 24-27 monoamine oxidase A Rattus norvegicus 98-101 7090425-3 1982 The enzyme system involved was dependent on cytochrome P-450 and the main metabolites were desmethylimipramine (DMI), formaldehyde and p-chlorobenzoic acid. Desipramine 91-110 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 44-60 7090425-3 1982 The enzyme system involved was dependent on cytochrome P-450 and the main metabolites were desmethylimipramine (DMI), formaldehyde and p-chlorobenzoic acid. Desipramine 112-115 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 44-60 7178370-0 1982 Comparison of growth hormone stimulation induced by desimipramine, diazepam and metaclazepam in man. Desipramine 52-65 growth hormone 1 Homo sapiens 14-28 6457138-4 1981 Plasma renin activity was also reduced in rats that had received intraventricular 5,7-dihydroxytryptamine after pretreatment with desmethylimipramine. Desipramine 130-149 renin Rattus norvegicus 7-12 6314168-0 1983 Involvement of alpha- and beta 1-adrenergic mechanisms in the immobility-reducing action of desipramine in the forced swimming test. Desipramine 92-103 potassium calcium-activated channel subfamily M regulatory beta subunit 1 Homo sapiens 15-32 6135776-0 1983 Stereoselectivity of catecholamines: differential effects of cocaine and desipramine on catecholamine-induced contractions of the rat isolated vas deferens. Desipramine 73-84 arginine vasopressin Rattus norvegicus 143-146 6135776-1 1983 The effect of uptake inhibitors, cocaine and desipramine, on the contractile response of the rat isolated vas deferens to the enantiomers of noradrenaline and adrenaline and to the corresponding deoxy-derivatives, dopamine and epinephline, was investigated. Desipramine 45-56 arginine vasopressin Rattus norvegicus 106-109 6135776-4 1983 This selective antagonism of dopamine by desipramine was also observed for the separated epididymal and prostatic ends of the rat vas deferens. Desipramine 41-52 arginine vasopressin Rattus norvegicus 130-133 6124090-10 1982 It is likely that such studies will enrich our understanding of how these agents work, of the difference between agents which have been classed together on the basis of preclinical studies (e.g., DMI, which appears to increase PRL and GH, and NT which appears not to) and provided additional evidence to test current hypotheses about the biological basis of their antidepressant action. Desipramine 196-199 prolactin Homo sapiens 227-230 7320735-6 1981 Desipramine treatment increase NAT activity in 8-day-old animals; hydrocortisone-treated animals were least affected. Desipramine 0-11 bromodomain containing 2 Homo sapiens 31-34 6971560-8 1981 Desipramine, acutely, had no effect on any of the above paradigms but after chronic administration, potentiated the effect of low dose 5-HTP on prolactin secretion. Desipramine 0-11 prolactin Rattus norvegicus 144-153 6452790-4 1981 In the same individuals, desipramine concentrations were highly correlated with decreases of MHPG in the CSF. Desipramine 25-36 colony stimulating factor 2 Homo sapiens 105-108 119692-3 1979 TRH (10(-7)--10(--3)M) alone did not increase the DA release into the incubation medium, but it clearly enhanced the DA release in the concomitant presence of desipramine (5 x 10(-5)M). Desipramine 159-170 thyrotropin releasing hormone Rattus norvegicus 0-3 1000130-12 1976 5 The long-term effects of DSP 4 on the noradrenaline accumulation, the DBH activity and noradrenaline concentration in the rat brain were antagonized by desipramine (10 mg/kg i.p.). Desipramine 154-165 dopamine beta-hydroxylase Rattus norvegicus 72-75 215414-1 1978 Intravenous infusion of two separate doses of angiotensin II in pentobarbital-anesthetized, desipramine-treated animals produced dose-related increases in arterial blood pressure and caused significant potentiation of the cardioacceleration observed during the stimulation of the right postganglionic cardiac sympathetic nerve fibers. Desipramine 92-103 angiotensinogen Homo sapiens 46-60 365126-3 1978 The levels of both drugs were approximately equal in plasma, but desipramine predominated in CSF (imipramine/desipramine ratio of 0.8). Desipramine 65-76 colony stimulating factor 2 Homo sapiens 93-96 582390-0 1978 [Influence of single and repeated administration of desipramine (DMI) on the secretion of growth hormone (GH) (author"s transl)]. Desipramine 52-63 growth hormone 1 Homo sapiens 90-104 582390-0 1978 [Influence of single and repeated administration of desipramine (DMI) on the secretion of growth hormone (GH) (author"s transl)]. Desipramine 52-63 growth hormone 1 Homo sapiens 106-108 582390-0 1978 [Influence of single and repeated administration of desipramine (DMI) on the secretion of growth hormone (GH) (author"s transl)]. Desipramine 65-68 growth hormone 1 Homo sapiens 90-104 582390-0 1978 [Influence of single and repeated administration of desipramine (DMI) on the secretion of growth hormone (GH) (author"s transl)]. Desipramine 65-68 growth hormone 1 Homo sapiens 106-108 582390-1 1978 DMI leads to stimulation of GH in healthy male subjects 60--120 min after i.m. Desipramine 0-3 growth hormone 1 Homo sapiens 28-30 582390-3 1978 During a 20-day administration of 2 x 75 mg DMI per day a repeated stimulation of GH was provable on days 0, 10 and 20 in two male patients, whereas no stimulation of GH occurred in two female patients who underwent the same treatment. Desipramine 44-47 growth hormone 1 Homo sapiens 82-84 582390-6 1978 The increase in PRL and TSH, which is induced by the thyrotrophin releasing hormone (TRH), is provable after acute as well as after chronic administration of DMI. Desipramine 158-161 prolactin Homo sapiens 16-19 1012341-3 1976 In vas deferens from non-reserpinized rats the release of noradrenaline evoked by veratridine was partially antagonized by omission of Ca2+ in the incubation medium and partially inhibited by low concentrations of desipramine. Desipramine 214-225 arginine vasopressin Rattus norvegicus 3-6 958507-0 1976 Persistent enhancement of potassium-induced responses of the rat vas deferens by desipramine. Desipramine 81-92 arginine vasopressin Rattus norvegicus 65-68 991921-2 1976 Org 6582 was approximately twice as potent as fluoxetine, five times more potent than chlorimipramine and 14 times more potent than desipramine in blocking the ability of p-chloroamphetamine to lower rat brain 5-HT content. Desipramine 132-143 POU class 6 homeobox 1 Rattus norvegicus 204-211 186834-6 1976 By that time the MUA changes of PH and MRF showed similar characteristics in response to TRH administration which was observed following the injection of desipramine and imipramine. Desipramine 154-165 thyrotropin releasing hormone Felis catus 89-92 958507-1 1976 The effect of desipramine on the cumulative dose-response curves of noradrenaline and potassium (K+) was examined on the isolated rat vas deferens. Desipramine 14-25 arginine vasopressin Rattus norvegicus 134-137 5348466-4 1969 In slices of rat vas deferens incubated with various concentrations of (3)H-NA, cocaine and desipramine and phenoxybenzamine were also shown to act as competitive inhibitors of NA uptake. Desipramine 92-103 arginine vasopressin Rattus norvegicus 17-20 5067380-0 1972 Oxidation and glucuronidation of certain drugs in various subcellular fractions of rat liver: binding of desmethylimipramine and hexobarbital to cytochrome P-450 and oxidation and glucuronidation of desmethylimipramine, aminopyrine, p-nitrophenol and 1-naphthol. Desipramine 105-124 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 145-161 5314401-0 1970 Binding to cytochrome P-450 and metabolism of desmethylimipramine and metabolites in rat liver microsomes. Desipramine 46-65 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 11-27 476283-2 1979 Detection limits of 0.2 ng ml-1 for imipramine and 0.1 ng ml-1 for desipramine were demonstrated with a signal-to-noise ratio maintained at 2:1 or better. Desipramine 67-78 interleukin 17F Homo sapiens 58-62 1187753-2 1975 The administration of NE, DOPA, amphetamine, phenelzine, desipramine and clonidine induced the reappearance of the CAR. Desipramine 57-68 nuclear receptor subfamily 1, group I, member 3 Rattus norvegicus 115-118 33220685-2 2021 It poorly infects mice deficient in acid sphingomyelinase (ASM), a lysosomal enzyme critical for cholesterol efflux, and wild-type mice treated with desipramine that functionally inhibits ASM. Desipramine 149-160 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 188-191 33496295-4 2021 Layer-resolved DMI calculations revealed that the sign of the spin-orbit coupling (SOC) energy was changed for Au near the interface of Au/Fe under tensile stress, subsequently reversing the chirality of the i-DMI from left-handed to right-handed. Desipramine 15-18 spindlin 1 Homo sapiens 62-66 4382642-0 1967 Potentiation of noradrenaline isomers by cocaine and desipramine in the isolated vas deferens of the rat. Desipramine 53-64 arginine vasopressin Rattus norvegicus 81-84 33513512-8 2021 For example, IC50 value of desipramine for the zSERT was 1/200 of that for the hSERT. Desipramine 27-38 solute carrier family 6 member 4 Homo sapiens 79-84 33220685-8 2021 Infection was markedly lower in ASM-deficient and desipramine-treated wild-type mice versus desipramine-treated SCID mice. Desipramine 92-103 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 32-35 32294250-9 2020 Desipramine administration downregulated mRNA expressions of IL-1beta, iNOS, COX-2 and TIMP-1 when compared to vehicle alone in the ligature group (P<0.05). Desipramine 0-11 interleukin 1 alpha Rattus norvegicus 61-69 32294250-9 2020 Desipramine administration downregulated mRNA expressions of IL-1beta, iNOS, COX-2 and TIMP-1 when compared to vehicle alone in the ligature group (P<0.05). Desipramine 0-11 nitric oxide synthase 2 Rattus norvegicus 71-75 32294250-9 2020 Desipramine administration downregulated mRNA expressions of IL-1beta, iNOS, COX-2 and TIMP-1 when compared to vehicle alone in the ligature group (P<0.05). Desipramine 0-11 cytochrome c oxidase II, mitochondrial Rattus norvegicus 77-82 32294250-9 2020 Desipramine administration downregulated mRNA expressions of IL-1beta, iNOS, COX-2 and TIMP-1 when compared to vehicle alone in the ligature group (P<0.05). Desipramine 0-11 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 87-93 32294250-10 2020 MMP-9 expression and MMP-9/TIMP-1 ratio were similar among rats with ligature-induced periodontitis (P>0.05); however, MMP-9 activity was lower in the group treated with desipramine (P<0.05). Desipramine 170-181 matrix metallopeptidase 9 Rattus norvegicus 0-5 32294250-10 2020 MMP-9 expression and MMP-9/TIMP-1 ratio were similar among rats with ligature-induced periodontitis (P>0.05); however, MMP-9 activity was lower in the group treated with desipramine (P<0.05). Desipramine 170-181 matrix metallopeptidase 9 Rattus norvegicus 21-26 32294250-10 2020 MMP-9 expression and MMP-9/TIMP-1 ratio were similar among rats with ligature-induced periodontitis (P>0.05); however, MMP-9 activity was lower in the group treated with desipramine (P<0.05). Desipramine 170-181 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 27-33 32294250-10 2020 MMP-9 expression and MMP-9/TIMP-1 ratio were similar among rats with ligature-induced periodontitis (P>0.05); however, MMP-9 activity was lower in the group treated with desipramine (P<0.05). Desipramine 170-181 matrix metallopeptidase 9 Rattus norvegicus 21-26 32679212-1 2020 Single-crystal X-ray diffraction and DFT calculation have been carried out for an atomic-level understanding of beta-cyclodextrin (beta-CD) inclusion complexation with two tricyclic antidepressants (TCAs), desipramine and imipramine. Desipramine 206-217 ACD shelterin complex subunit and telomerase recruitment factor Homo sapiens 131-138 32697958-8 2020 Taken together with existing literature, concurrent blockade of OCTs and/or PMAT merits further investigation as an adjunctive therapeutic for desipramine-like antidepressant drugs. Desipramine 143-154 solute carrier family 29 member 4 Homo sapiens 76-80 32642907-12 2020 In contrast, treatment with desipramine, a NET selective inhibitor, resulted in profoundly decreased xenograft [125I]MIBG levels (p < 0.0001). Desipramine 28-39 solute carrier family 6 member 2 Homo sapiens 43-46 33117297-7 2020 Results indicate a positive response in terms of growth performance and gastrointestinal digestibility to REE supplementation, and 400 mg/kg DMI treatment presented the most average daily feed intake (ADFI), the best average daily weight gain (ADG), and the least F/G. Desipramine 141-144 ADFI Bos taurus 201-205 33117297-7 2020 Results indicate a positive response in terms of growth performance and gastrointestinal digestibility to REE supplementation, and 400 mg/kg DMI treatment presented the most average daily feed intake (ADFI), the best average daily weight gain (ADG), and the least F/G. Desipramine 141-144 ADG Bos taurus 244-247 31090092-6 2019 The predicted versus observed geometric mean ratio (GMR) of AUCinf for CYP2D6 substrates desipramine and metoprolol coadministered with mirabegron 100 or 160 mg once daily were 3.47 versus 3.41 and 2.97 versus 3.29, respectively, indicating that the mirabegron model can be used to predict clinical CYP2D6 inhibition. Desipramine 89-100 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 71-77 33344693-8 2020 Further, Adcy1 tg mice show reduced immobility under acute physical stress conditions in the forced swimming test and are more sensitive to the antidepressant desipramine. Desipramine 159-170 adenylate cyclase 1 Mus musculus 9-14 32425895-9 2020 Nonetheless, inhibition of ASMase by desipramine did not affect IAV infection. Desipramine 37-48 sphingomyelin phosphodiesterase 1 Homo sapiens 27-33 32481596-9 2020 Treatment of immortalized cell in vision research (ARPE-19) cells with high glucose resulted in a 25% +- 1.6% decrease in citrate synthase activity at 72 h. Inhibition of ASM with desipramine (15 muM, 1 h daily) abolished the decreases in metabolic functional parameters. Desipramine 180-191 citrate synthase Homo sapiens 122-138 32481596-9 2020 Treatment of immortalized cell in vision research (ARPE-19) cells with high glucose resulted in a 25% +- 1.6% decrease in citrate synthase activity at 72 h. Inhibition of ASM with desipramine (15 muM, 1 h daily) abolished the decreases in metabolic functional parameters. Desipramine 180-191 sphingomyelin phosphodiesterase 1 Homo sapiens 171-174 31788096-11 2019 Treatment with Desi inhibited the activation of insulin-like growth factor-1 receptor and inhibited the activation of proteins in the PI3K/Akt signaling pathway. Desipramine 15-19 insulin like growth factor 1 receptor Homo sapiens 48-85 31442583-7 2019 CD spectroscopic analysis of protein stability allowed identifying CHS and POPX as stabilizing components, which increased hSERT thermostability by 7 C. The kinetic dissociation constant KD of 2.8 muM (+-0.05) for of the inhibitor Desipramine was determined with a ka of 10,848 M - 1 s-1 (+-220) and a kd of 0.03 s-1 (+-4.7 x 10-5). Desipramine 232-243 solute carrier family 6 member 4 Homo sapiens 123-128 31090092-6 2019 The predicted versus observed geometric mean ratio (GMR) of AUCinf for CYP2D6 substrates desipramine and metoprolol coadministered with mirabegron 100 or 160 mg once daily were 3.47 versus 3.41 and 2.97 versus 3.29, respectively, indicating that the mirabegron model can be used to predict clinical CYP2D6 inhibition. Desipramine 89-100 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 299-305 31624238-9 2019 DMI treatment restored the mRNA expression of AVP in the SCN and enhanced GR-mediated signaling by upregulating GR expression and nuclear translocation in the hippocampus. Desipramine 0-3 arginine vasopressin Mus musculus 46-49 31090092-4 2019 Models predicted and verified mirabegron and desipramine (CYP2D6 substrate) and 5-HMT and ketoconazole (strong CYP3A inhibitor) DDIs. Desipramine 45-56 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 58-64 31624238-9 2019 DMI treatment restored the mRNA expression of AVP in the SCN and enhanced GR-mediated signaling by upregulating GR expression and nuclear translocation in the hippocampus. Desipramine 0-3 nuclear receptor subfamily 3, group C, member 1 Mus musculus 74-76 31624238-9 2019 DMI treatment restored the mRNA expression of AVP in the SCN and enhanced GR-mediated signaling by upregulating GR expression and nuclear translocation in the hippocampus. Desipramine 0-3 nuclear receptor subfamily 3, group C, member 1 Mus musculus 112-114 31624238-11 2019 Taken together, our data indicate that DMI treatment enhances GR-mediated signaling and restores the synchronization of peripheral clocks with the SCN and support the hypothesis that altered circadian entrainment is a modifiable risk factor for depression. Desipramine 39-42 nuclear receptor subfamily 3, group C, member 1 Mus musculus 62-64 30550872-3 2019 RAW 264.7 cells were pretreated with desipramine, an ASM inhibitor, prior to LPS challenge in vitro. Desipramine 37-48 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 53-56 30546074-5 2019 The neutral SMase inhibitor GW4869 as well as the genetic inhibition of nSMase with small interfering RNA (siRNA) against sphingomyelin phosphodiesterase 3 (SMPD3) prevented the inhibition by TNFalpha, but not the acid SMase inhibitor desipramine. Desipramine 235-246 sphingomyelin phosphodiesterase 2 Homo sapiens 72-78 30546074-5 2019 The neutral SMase inhibitor GW4869 as well as the genetic inhibition of nSMase with small interfering RNA (siRNA) against sphingomyelin phosphodiesterase 3 (SMPD3) prevented the inhibition by TNFalpha, but not the acid SMase inhibitor desipramine. Desipramine 235-246 sphingomyelin phosphodiesterase 3 Homo sapiens 157-162 30985942-7 2019 The PBPK model predicted that the AUC for mirabegron with itraconazole (a CYP3A4 inhibitor) was 4.12-times that in healthy elderly subjects administered mirabegron alone, and predicted that the proportional change in AUC for desipramine (a CYP2D6 substrate) with mirabegron was greater than that in healthy subjects. Desipramine 225-236 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 74-80 30985942-7 2019 The PBPK model predicted that the AUC for mirabegron with itraconazole (a CYP3A4 inhibitor) was 4.12-times that in healthy elderly subjects administered mirabegron alone, and predicted that the proportional change in AUC for desipramine (a CYP2D6 substrate) with mirabegron was greater than that in healthy subjects. Desipramine 225-236 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 240-246 30689930-5 2019 Calm heifers (mean REV minus 1 SD) had 4% greater (P < 0.001) initial BW, 12% greater (P < 0.001) ADG, 8% greater (P < 0.001) DMI, and 4% greater (P < 0.02) G:F than heifers with excitable temperaments (mean REV plus 1 SD). Desipramine 135-138 synaptosome associated protein 91 Homo sapiens 0-4 30728250-7 2019 5-HT elicited concentration-dependent contractions of the CE duct (pEC50 = 6.5 +- 0.1) that were potentiated with high potency by the norepinephrine transporter (NET) inhibitor desipramine and with low potency by the highly selective serotonin transporter inhibitor paroxetine, indicating that the NET is the major mediator of 5-HT reuptake in vitro. Desipramine 177-188 solute carrier family 6 member 2 Rattus norvegicus 134-160 30728250-7 2019 5-HT elicited concentration-dependent contractions of the CE duct (pEC50 = 6.5 +- 0.1) that were potentiated with high potency by the norepinephrine transporter (NET) inhibitor desipramine and with low potency by the highly selective serotonin transporter inhibitor paroxetine, indicating that the NET is the major mediator of 5-HT reuptake in vitro. Desipramine 177-188 solute carrier family 6 member 2 Rattus norvegicus 162-165 31262782-6 2019 Treatment of mice with pharmacological inhibitors of acid sphingomyelinase (ASMase), desipramine and imipramine, attenuated ATP-induced TF decryption. Desipramine 85-96 coagulation factor III Mus musculus 136-138 30910852-2 2019 Here, we show that elevated aSMase activity and ceramide content were reduced by desipramine treatment in diabetic animals. Desipramine 81-92 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 28-34 30910852-3 2019 The inhibitor of aSMase, desipramine, improved vascular dysfunction in db/db mice. Desipramine 25-36 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 17-23 30910852-4 2019 High glucose (HG)-induced up-regulation of aSMase activity and ceramide levels were restored by treatment with aSMase siRNA or desipramine in endothelial cells. Desipramine 127-138 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 43-49 30910852-5 2019 In addition, aSMase siRNA or desipramine treatment increased the release of nitric oxide (NO) and the phosphorylation of endothelial NO synthase (eNOS) in diabetic mouse aortas and aortic endothelial cells with HG.These results indicate that inhibition of aSMase/ceramide pathway improves endothelium-dependent vascular relaxation (EDR) largely through regulating the eNOS/NO pathway in diabetic animals. Desipramine 29-40 nitric oxide synthase 3, endothelial cell Mus musculus 121-144 30910852-5 2019 In addition, aSMase siRNA or desipramine treatment increased the release of nitric oxide (NO) and the phosphorylation of endothelial NO synthase (eNOS) in diabetic mouse aortas and aortic endothelial cells with HG.These results indicate that inhibition of aSMase/ceramide pathway improves endothelium-dependent vascular relaxation (EDR) largely through regulating the eNOS/NO pathway in diabetic animals. Desipramine 29-40 nitric oxide synthase 3, endothelial cell Mus musculus 146-150 30910852-5 2019 In addition, aSMase siRNA or desipramine treatment increased the release of nitric oxide (NO) and the phosphorylation of endothelial NO synthase (eNOS) in diabetic mouse aortas and aortic endothelial cells with HG.These results indicate that inhibition of aSMase/ceramide pathway improves endothelium-dependent vascular relaxation (EDR) largely through regulating the eNOS/NO pathway in diabetic animals. Desipramine 29-40 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 256-262 30910852-5 2019 In addition, aSMase siRNA or desipramine treatment increased the release of nitric oxide (NO) and the phosphorylation of endothelial NO synthase (eNOS) in diabetic mouse aortas and aortic endothelial cells with HG.These results indicate that inhibition of aSMase/ceramide pathway improves endothelium-dependent vascular relaxation (EDR) largely through regulating the eNOS/NO pathway in diabetic animals. Desipramine 29-40 nitric oxide synthase 3, endothelial cell Mus musculus 368-372 30550872-5 2019 Conversely, inhibition of ASM activity by desipramine diminished LPS induced ASM activities and TNF production of RAW 264.7 cells. Desipramine 42-53 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 26-29 30550872-5 2019 Conversely, inhibition of ASM activity by desipramine diminished LPS induced ASM activities and TNF production of RAW 264.7 cells. Desipramine 42-53 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 77-80 30550872-8 2019 Desipramine administration overrode ASM activities in colon, and ameliorated DSS-induced colitis evidenced with the reduced disease activities and the decreased cytokine levels. Desipramine 0-11 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 36-39 30284474-4 2018 The Dmi group was gavaged with the p38 MAPK inhibitor SB203580 for 9 weeks, and the effects on beta cell dysfunction and apoptosis were investigated. Desipramine 4-7 mitogen-activated protein kinase 14 Mus musculus 35-43 30326559-11 2018 Since we were also able to show that the functional inhibitor of SMPD1, desipramine, ameliorates downregulation of CYP mRNA expression and activities in the acute phase of sepsis in wild-type mice, SMPD1 might be an interesting pharmacological target, which should be further investigated. Desipramine 72-83 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 65-70 30326559-11 2018 Since we were also able to show that the functional inhibitor of SMPD1, desipramine, ameliorates downregulation of CYP mRNA expression and activities in the acute phase of sepsis in wild-type mice, SMPD1 might be an interesting pharmacological target, which should be further investigated. Desipramine 72-83 cytochrome P450, family 21, subfamily a, polypeptide 1 Mus musculus 115-118 30326559-11 2018 Since we were also able to show that the functional inhibitor of SMPD1, desipramine, ameliorates downregulation of CYP mRNA expression and activities in the acute phase of sepsis in wild-type mice, SMPD1 might be an interesting pharmacological target, which should be further investigated. Desipramine 72-83 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 198-203 29758252-8 2018 In contrast, selective noradrenaline reuptake inhibitor desipramine had a significant effect on RGS8tg in the FST. Desipramine 56-67 regulator of G-protein signaling 8 Mus musculus 96-100 30126999-6 2018 The antidepressant desipramine, which decreases ceramide levels by inhibiting acid sphingomyelinase, corrects EE defects in the RPE of Abca4-/- mice. Desipramine 19-30 ATP-binding cassette, sub-family A (ABC1), member 4 Mus musculus 135-140 29068103-2 2018 The level of LC3-II was elevated and that of p62 was reduced in desipramine-treated L929 cells, indicating the induction of autophagy by desipramine. Desipramine 64-75 nucleoporin 62 Mus musculus 45-48 29496989-6 2018 Results: The half-maximal inhibitory concentrations of desipramine, escitalopram, venlafaxine, and bupropion on 131I-MIBG cellular uptake were 11.9 nM, 7.5 muM, 4.92 muM, and 12.9 muM, respectively. Desipramine 55-66 latexin Homo sapiens 156-159 29496989-6 2018 Results: The half-maximal inhibitory concentrations of desipramine, escitalopram, venlafaxine, and bupropion on 131I-MIBG cellular uptake were 11.9 nM, 7.5 muM, 4.92 muM, and 12.9 muM, respectively. Desipramine 55-66 latexin Homo sapiens 166-169 29496989-6 2018 Results: The half-maximal inhibitory concentrations of desipramine, escitalopram, venlafaxine, and bupropion on 131I-MIBG cellular uptake were 11.9 nM, 7.5 muM, 4.92 muM, and 12.9 muM, respectively. Desipramine 55-66 latexin Homo sapiens 166-169 29401225-17 2018 It will be most effective if ADG over the period coinciding with intake recording and ADG over a much longer period of time are simultaneously included in a multiple-trait genetic evaluation with DMI and used in a selection index for efficiency. Desipramine 196-199 ADG Bos taurus 29-32 29068103-2 2018 The level of LC3-II was elevated and that of p62 was reduced in desipramine-treated L929 cells, indicating the induction of autophagy by desipramine. Desipramine 137-148 nucleoporin 62 Mus musculus 45-48 29068103-5 2018 Furthermore, desipramine-induced vacuolization was observed in autophagy-deficient Atg7-/- mouse embryonic fibroblasts (MEFs) as well as wild-type Atg7+/+ MEFs. Desipramine 13-24 autophagy related 7 Mus musculus 83-87 29068103-5 2018 Furthermore, desipramine-induced vacuolization was observed in autophagy-deficient Atg7-/- mouse embryonic fibroblasts (MEFs) as well as wild-type Atg7+/+ MEFs. Desipramine 13-24 autophagy related 7 Mus musculus 147-151 29414147-5 2018 RESULTS: In SH-SY5Y cells, [3H]-dopamine uptake was inhibited by desipramine (selective NET inhibitor), GBR-12909 (selective DAT inhibitor), and fluoxetine (selective inhibitor of the serotonin transporter, SERT) with IC50 values 37, 537, and 2800nM, respectively. Desipramine 65-76 solute carrier family 6 member 4 Homo sapiens 207-211 28359918-9 2017 Chronic administration of desipramine, fluoxetine, or tianeptine increased the BDNF mRNA expression and protein levels in the LPS-injected stressed rats. Desipramine 26-37 brain-derived neurotrophic factor Rattus norvegicus 79-83 29038231-4 2018 For static and dynamic predictions, midazolam, repaglinide, and desipramine were used as probe substrates for CYP3A4/5, CYP2C8, and CYP2D6, respectively. Desipramine 64-75 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 110-116 28955042-5 2017 Strikingly, similar to heterozygote expression of SMPD1, either preventative (p-smpd1+/+) or therapeutic (t-smpd1+/+) pharmacological treatment strategies with desipramine - a functional inhibitor of acid sphingomyelinase (FIASMA) - significantly improved liver function and survival. Desipramine 160-171 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 200-221 29468173-2 2018 Desipramine, a noradrenaline reuptake inhibitor, reduced the number of apneas in Mecp2-deficient mice, a model of RTT. Desipramine 0-11 methyl CpG binding protein 2 Mus musculus 81-86 28842312-8 2017 Moreover, desipramine treatment was able to reduce the oxidative damage to rat liver, and to increase the expression of telomerase reverse transcriptase (TERT), leading to correspondingly restored telomerase activity. Desipramine 10-21 telomerase reverse transcriptase Rattus norvegicus 120-152 28842312-8 2017 Moreover, desipramine treatment was able to reduce the oxidative damage to rat liver, and to increase the expression of telomerase reverse transcriptase (TERT), leading to correspondingly restored telomerase activity. Desipramine 10-21 telomerase reverse transcriptase Rattus norvegicus 154-158 28675459-10 2017 The combination of desipramine and G-CSF is safe and signals improved mobilization over G-CSF alone, providing a possible alternative means of mobilization that needs further investigation. Desipramine 19-30 colony stimulating factor 3 Homo sapiens 88-93 29169412-8 2017 Accordingly, Cer content was increase in MCF-7 cells treated by TNF-alpha, which was blocked by the pretreatment of ASM inhibitor Des. Desipramine 130-133 tumor necrosis factor Homo sapiens 64-73 29169412-8 2017 Accordingly, Cer content was increase in MCF-7 cells treated by TNF-alpha, which was blocked by the pretreatment of ASM inhibitor Des. Desipramine 130-133 sphingomyelin phosphodiesterase 1 Homo sapiens 116-119 29169412-10 2017 Des and DMS could respectively reverse the TNF-alpha-induced decrease and increase of proliferation in MCF-7 and MDA-MB-231 cells. Desipramine 0-3 tumor necrosis factor Homo sapiens 43-52 28627696-6 2017 In addition, desipramine activated MAPKs (ERK 1/2, JNK, and p38) and increased intracellular Ca2+ levels. Desipramine 13-24 mitogen-activated protein kinase 3 Homo sapiens 42-49 28627696-6 2017 In addition, desipramine activated MAPKs (ERK 1/2, JNK, and p38) and increased intracellular Ca2+ levels. Desipramine 13-24 mitogen-activated protein kinase 8 Homo sapiens 51-54 28627696-6 2017 In addition, desipramine activated MAPKs (ERK 1/2, JNK, and p38) and increased intracellular Ca2+ levels. Desipramine 13-24 mitogen-activated protein kinase 1 Homo sapiens 60-63 28627696-7 2017 Pro-apoptotic effects of desipramine were abolished after MAPK inhibitors (PD98059, SB203580, and SP600125) or N-acetyl-L-cysteine (NAC), as a ROS scavenger, treatments. Desipramine 25-36 mitogen-activated protein kinase 3 Homo sapiens 58-62 28627696-7 2017 Pro-apoptotic effects of desipramine were abolished after MAPK inhibitors (PD98059, SB203580, and SP600125) or N-acetyl-L-cysteine (NAC), as a ROS scavenger, treatments. Desipramine 25-36 X-linked Kx blood group Homo sapiens 132-135 28627696-8 2017 These findings suggest that desipramine shows anti-proliferative effects in Hep3B cells mediated by promotion of apoptosis, activation of MAPK signaling, and increase in intracellular Ca2+ levels. Desipramine 28-39 mitogen-activated protein kinase 3 Homo sapiens 138-142 31994095-9 2017 Chronic administration of desipramine, fluoxetine, or tianeptine increased the BDNF mRNA expression and protein levels in the LPS-injected stressed rats. Desipramine 26-37 brain-derived neurotrophic factor Rattus norvegicus 79-83 27654664-12 2017 RESULTS: We show dose-dependent inhibition of [125 I]MIBG uptake by dexmedetomidine, but at several logs lower potency than the known NET inhibitor desipramine. Desipramine 148-159 solute carrier family 6 member 2 Homo sapiens 134-137 28420138-8 2017 Furthermore, similar results in desipramine-pretreated SMPD1-/- littermates suggest an SMPD1-independent pathway. Desipramine 32-43 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 55-60 28420138-8 2017 Furthermore, similar results in desipramine-pretreated SMPD1-/- littermates suggest an SMPD1-independent pathway. Desipramine 32-43 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 87-92 27900470-0 2017 Influence of the selective antagonist of the NR2B subunit of the NMDA receptor, traxoprodil, on the antidepressant-like activity of desipramine, paroxetine, milnacipran, and bupropion in mice. Desipramine 132-143 glutamate receptor, ionotropic, NMDA2B (epsilon 2) Mus musculus 45-49 28520379-6 2012 Imipramine is primarily metabolized via CYP2C19 to active metabolites, including desipramine, also a tricyclic antidepressant. Desipramine 81-92 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 40-47 28352358-5 2017 Then, the effect of 28-day daily treatment with desipramine (DMI; 10 mg/kg), fluoxetine (5 mg/kg) or tianeptine (10 mg/kg) on the number of copies of VEGF mRNA in the amygdala, hippocampus and hypothalamus, and on serum VEGF protein levels, of rats subjected to chronic stress was determined. Desipramine 48-59 vascular endothelial growth factor A Rattus norvegicus 150-154 28352358-11 2017 Furthermore, the results identified that the stress-induced increase in VEGF mRNA expression in the amygdala and hypothalamus was attenuated by long-term administration of DMI. Desipramine 172-175 vascular endothelial growth factor A Rattus norvegicus 72-76 28352358-13 2017 In conclusion, the current study suggests that under conditions of stress, VEGF serves a role in the mechanism of action of DMI, through modulating activity of the norepinephrine system. Desipramine 124-127 vascular endothelial growth factor A Rattus norvegicus 75-79 27831486-5 2017 We investigated the relation between antidepressant-like response to the TCA desipramine using the tail suspension test and saturation binding of [3H]nisoxetine to the norepinephrine transporter (NET), the primary target of desipramine, in juvenile (21 days postnatal [P21]), adolescent (P28), and adult (P90) wild-type (SERT+/+) mice. Desipramine 77-88 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 196-199 27773824-8 2016 By contrast, cannabinoid CB1 and CB2 receptor antagonists partially attenuated the antinociceptive actions of desipramine in a manner that was restricted to the development phase of paclitaxel-induced neuropathic pain only. Desipramine 110-121 cannabinoid receptor 1 Homo sapiens 25-28 27773824-8 2016 By contrast, cannabinoid CB1 and CB2 receptor antagonists partially attenuated the antinociceptive actions of desipramine in a manner that was restricted to the development phase of paclitaxel-induced neuropathic pain only. Desipramine 110-121 cannabinoid receptor 2 Homo sapiens 33-36 27773824-10 2016 Notably, desipramine (1nM-1muM) alone did not alter tumor cell viability and did not prevent the cytotoxic effects of paclitaxel under identical conditions. Desipramine 9-20 latexin Homo sapiens 27-30 27571014-12 2016 The ischemia-induced increase in retinal TNF-alpha levels was suppressed by the administration of the ASMase inhibitor desipramine, or by reducing ASMase expression. Desipramine 119-130 tumor necrosis factor Mus musculus 41-50 27799387-5 2016 AHI was quantified off CPAP.Desipramine reduced active Pcrit (median (interquartile range) -5.2 (4.3) cmH2O on desipramine versus -1.9 (2.7) cmH2O on placebo; p=0.049) but not passive Pcrit (-2.2 (3.4) versus -0.7 (2.1) cmH2O; p=0.135). Desipramine 28-39 troponin T2, cardiac type Homo sapiens 102-106 27799387-5 2016 AHI was quantified off CPAP.Desipramine reduced active Pcrit (median (interquartile range) -5.2 (4.3) cmH2O on desipramine versus -1.9 (2.7) cmH2O on placebo; p=0.049) but not passive Pcrit (-2.2 (3.4) versus -0.7 (2.1) cmH2O; p=0.135). Desipramine 28-39 troponin T2, cardiac type Homo sapiens 141-145 26923251-9 2016 ASM was inhibited by siRNA or desipramine (DES), and/or ceramide was degraded by recombinant acid ceramidase (AC). Desipramine 30-41 sphingomyelin phosphodiesterase 1 Homo sapiens 0-3 26923251-9 2016 ASM was inhibited by siRNA or desipramine (DES), and/or ceramide was degraded by recombinant acid ceramidase (AC). Desipramine 43-46 sphingomyelin phosphodiesterase 1 Homo sapiens 0-3 27341515-9 2016 Hydrolysis of sphingomyelin, clustering of receptor complexes, such as the CD95L/Fas-receptor, as well as formation of ceramide enriched macrodomains was abrogated using functional inhibitors (desipramine and NB6). Desipramine 193-204 Fas ligand Homo sapiens 75-80 27341515-10 2016 Strikingly, the stimulation of HMECs with serum obtained from sepsis patients or mixture of proinflammatory cytokines resulted in cytotoxicity and ADAMTS13 downregulation which was abrogated using desipramine, amino bisphosphonates and genetic inhibitors. Desipramine 197-208 ADAM metallopeptidase with thrombospondin type 1 motif 13 Homo sapiens 147-155 27831486-5 2017 We investigated the relation between antidepressant-like response to the TCA desipramine using the tail suspension test and saturation binding of [3H]nisoxetine to the norepinephrine transporter (NET), the primary target of desipramine, in juvenile (21 days postnatal [P21]), adolescent (P28), and adult (P90) wild-type (SERT+/+) mice. Desipramine 224-235 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 168-194 27831486-5 2017 We investigated the relation between antidepressant-like response to the TCA desipramine using the tail suspension test and saturation binding of [3H]nisoxetine to the norepinephrine transporter (NET), the primary target of desipramine, in juvenile (21 days postnatal [P21]), adolescent (P28), and adult (P90) wild-type (SERT+/+) mice. Desipramine 224-235 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 196-199 27831486-7 2017 The potency and maximal antidepressant-like effect of desipramine was greater in P21 mice than in P90 mice and was SERT genotype independent. Desipramine 54-65 cyclin-dependent kinase inhibitor 1A (P21) Mus musculus 81-84 27831486-7 2017 The potency and maximal antidepressant-like effect of desipramine was greater in P21 mice than in P90 mice and was SERT genotype independent. Desipramine 54-65 transferrin receptor Mus musculus 98-101 27831486-7 2017 The potency and maximal antidepressant-like effect of desipramine was greater in P21 mice than in P90 mice and was SERT genotype independent. Desipramine 54-65 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 115-119 27831486-10 2017 These data show age-dependent shifts for desipramine to produce antidepressant-like effects that correlate with NET expression in the locus coeruleus and suggest that drugs with NET-blocking activity may be an effective alternative to SSRIs in juveniles. Desipramine 41-52 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 112-115 27831486-10 2017 These data show age-dependent shifts for desipramine to produce antidepressant-like effects that correlate with NET expression in the locus coeruleus and suggest that drugs with NET-blocking activity may be an effective alternative to SSRIs in juveniles. Desipramine 41-52 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 178-181 27440861-3 2016 Imipramine undergoes N-demethylation to form the active metabolite desipramine, and both imipramine and desipramine are converted to hydroxylated metabolites by the polymorphic enzyme CYP2D6. Desipramine 67-78 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 184-190 27440861-3 2016 Imipramine undergoes N-demethylation to form the active metabolite desipramine, and both imipramine and desipramine are converted to hydroxylated metabolites by the polymorphic enzyme CYP2D6. Desipramine 104-115 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 184-190 27440861-4 2016 The objective of the present study is to determine whether the human pharmacokinetics of desipramine following dosing of imipramine can be predicted using static and dynamic physiologically-based pharmacokinetic (PBPK) models from in vitro input data for CYP2D6 extensive metabolizer (EM) and poor metabolizer (PM) populations. Desipramine 89-100 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 255-261 27440861-9 2016 Moreover, the PBPK model was able to recover simultaneously plasma profiles of imipramine and desipramine in populations with different phenotypes of CYP2D6. Desipramine 94-105 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 150-156 27571014-12 2016 The ischemia-induced increase in retinal TNF-alpha levels was suppressed by the administration of the ASMase inhibitor desipramine, or by reducing ASMase expression. Desipramine 119-130 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 102-108 27259485-10 2016 Chronic (14 days) desipramine and fluoxetine, but not phenelzine, increased p-FADD (up to 59 %), p-FADD/FADD ratio (up to 1.8-fold), and pro-survival p-PEA-15 (up to 46 %) in rat brain cortex. Desipramine 18-29 proliferation and apoptosis adaptor protein 15 Rattus norvegicus 152-158 26970016-5 2016 In this study, we similarly show that Crtc1(-/-) mice are resistant to the antidepressant effect of chronic desipramine in a behavioral despair paradigm. Desipramine 108-119 CREB regulated transcription coactivator 1 Mus musculus 38-43 26785076-9 2016 The co-administration of DMI with the Src inhibitor SU6656, which blocks EphA4 forward signalling, could partially reverse the LTP attenuation, further supporting the targeting of the ephrinA3/EphA4 pathway by DMI. Desipramine 210-213 ephrin A3 Homo sapiens 184-192 27338163-0 2016 Desipramine improves depression-like behavior and working memory by up-regulating p-CREB in Alzheimer"s disease associated mice. Desipramine 0-11 cAMP responsive element binding protein 1 Mus musculus 84-88 27338163-8 2016 The neuroprotection of desipramine may be involved in the up-regulation of p-CREB level in the hippocampus of mice. Desipramine 23-34 cAMP responsive element binding protein 1 Mus musculus 77-81 26785076-0 2016 Desipramine targets astrocytes to attenuate synaptic plasticity via modulation of the ephrinA3/EphA4 signalling. Desipramine 0-11 ephrin A3 Homo sapiens 86-94 26785076-9 2016 The co-administration of DMI with the Src inhibitor SU6656, which blocks EphA4 forward signalling, could partially reverse the LTP attenuation, further supporting the targeting of the ephrinA3/EphA4 pathway by DMI. Desipramine 210-213 EPH receptor A4 Homo sapiens 193-198 26785076-0 2016 Desipramine targets astrocytes to attenuate synaptic plasticity via modulation of the ephrinA3/EphA4 signalling. Desipramine 0-11 EPH receptor A4 Homo sapiens 95-100 26785076-8 2016 DMI enhanced EphA4 clustering, which favoured an increased ephrinA3-mediated EphA4 phosphorylation and elevated EphA4 forward signalling. Desipramine 0-3 EPH receptor A4 Homo sapiens 13-18 26785076-8 2016 DMI enhanced EphA4 clustering, which favoured an increased ephrinA3-mediated EphA4 phosphorylation and elevated EphA4 forward signalling. Desipramine 0-3 ephrin A3 Homo sapiens 59-67 26785076-8 2016 DMI enhanced EphA4 clustering, which favoured an increased ephrinA3-mediated EphA4 phosphorylation and elevated EphA4 forward signalling. Desipramine 0-3 EPH receptor A4 Homo sapiens 77-82 26785076-8 2016 DMI enhanced EphA4 clustering, which favoured an increased ephrinA3-mediated EphA4 phosphorylation and elevated EphA4 forward signalling. Desipramine 0-3 EPH receptor A4 Homo sapiens 77-82 26785076-9 2016 The co-administration of DMI with the Src inhibitor SU6656, which blocks EphA4 forward signalling, could partially reverse the LTP attenuation, further supporting the targeting of the ephrinA3/EphA4 pathway by DMI. Desipramine 25-28 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 38-41 26785076-9 2016 The co-administration of DMI with the Src inhibitor SU6656, which blocks EphA4 forward signalling, could partially reverse the LTP attenuation, further supporting the targeting of the ephrinA3/EphA4 pathway by DMI. Desipramine 25-28 EPH receptor A4 Homo sapiens 73-78 26785076-9 2016 The co-administration of DMI with the Src inhibitor SU6656, which blocks EphA4 forward signalling, could partially reverse the LTP attenuation, further supporting the targeting of the ephrinA3/EphA4 pathway by DMI. Desipramine 25-28 ephrin A3 Homo sapiens 184-192 26785076-9 2016 The co-administration of DMI with the Src inhibitor SU6656, which blocks EphA4 forward signalling, could partially reverse the LTP attenuation, further supporting the targeting of the ephrinA3/EphA4 pathway by DMI. Desipramine 25-28 EPH receptor A4 Homo sapiens 193-198 26785076-9 2016 The co-administration of DMI with the Src inhibitor SU6656, which blocks EphA4 forward signalling, could partially reverse the LTP attenuation, further supporting the targeting of the ephrinA3/EphA4 pathway by DMI. Desipramine 210-213 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 38-41 26777153-3 2016 METHODS: We used diethylpyrocarbonate (DEPC) to specifically modify His(18) and obtained mono-ethyloxyformylated hIAPP (DMI). Desipramine 120-123 islet amyloid polypeptide Homo sapiens 113-118 26937856-0 2016 Desipramine administered chronically inhibits lipopolysaccharide-stimulated production of IL-1beta in the brain and plasma of rats. Desipramine 0-11 interleukin 1 beta Rattus norvegicus 90-98 26937856-2 2016 The present study investigated for the first time the effect of 21-day oral administration of desipramine on the lipopolysaccharide (LPS)-stimulated IL-1beta concentration in the olfactory bulb, hypothalamus, frontal cortex, hippocampus and plasma of rats, and on the LPS-induced IL-1beta mRNA level in the olfactory bulb. Desipramine 94-105 interleukin 1 beta Rattus norvegicus 149-157 26937856-2 2016 The present study investigated for the first time the effect of 21-day oral administration of desipramine on the lipopolysaccharide (LPS)-stimulated IL-1beta concentration in the olfactory bulb, hypothalamus, frontal cortex, hippocampus and plasma of rats, and on the LPS-induced IL-1beta mRNA level in the olfactory bulb. Desipramine 94-105 interleukin 1 beta Rattus norvegicus 280-288 26937856-6 2016 By using the alpha1/alpha2-adrenoceptor antagonist prazosin and the unspecific beta-adrenoceptor antagonist propranolol given prior to LPS, we found that the effect of desipramine on LPS-induced IL-1beta production was partially mediated by both adrenoceptors in the olfactory bulb and plasma, and that beta-adrenoceptors contributed also to its effect on the stimulated IL-1beta concentration in the hypothalamus. Desipramine 168-179 interleukin 1 beta Rattus norvegicus 195-203 26785076-10 2016 Thus, our findings suggest a putative novel mechanism for DMI to modulate LTP through the regulation of the ephrinA3/EphA4 signalling pathway. Desipramine 58-61 ephrin A3 Homo sapiens 108-116 26785076-10 2016 Thus, our findings suggest a putative novel mechanism for DMI to modulate LTP through the regulation of the ephrinA3/EphA4 signalling pathway. Desipramine 58-61 EPH receptor A4 Homo sapiens 117-122 26785076-11 2016 A further exploration of the molecular and behavioral consequences of targeting ephrinA3/EphA4 might help to improve the clinical use of DMI. Desipramine 137-140 ephrin A3 Homo sapiens 80-88 26785076-11 2016 A further exploration of the molecular and behavioral consequences of targeting ephrinA3/EphA4 might help to improve the clinical use of DMI. Desipramine 137-140 EPH receptor A4 Homo sapiens 89-94 26452722-11 2016 Similarly, quinidine-mediated CYP2D6 inhibition reduced clearance of dextromethorphan and desipramine by 71 and 22%, respectively. Desipramine 90-101 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 30-36 26809095-8 2016 Desipramine, a functional ASMase inhibitor, prevented RSV-induced ceramide increase. Desipramine 0-11 sphingomyelin phosphodiesterase 1 Homo sapiens 26-32 26250357-8 2015 Desipramine was less potent in middle-aged SERT(+/+) and SERT(+/-) mice than in adult SERT(+/+) or SERT(+/-) mice. Desipramine 0-11 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 43-47 26056032-6 2015 Previously defeated rats and controls were pre-treated with the norepinephrine transporter (NET) inhibitor desipramine (20 mg/kg, ip.) Desipramine 107-118 solute carrier family 6 member 2 Rattus norvegicus 64-90 26453893-9 2015 And desipramine significantly decreased AO fluorescence intensity and the expression of the p53 protein and gene. Desipramine 4-15 tumor protein p53 Homo sapiens 92-95 26305935-3 2015 Prevention of RGS9-2 action in the NAc increases the efficacy of the TCA desipramine and dramatically accelerates its onset of action. Desipramine 73-84 regulator of G-protein signaling 9 Mus musculus 14-20 26250357-8 2015 Desipramine was less potent in middle-aged SERT(+/+) and SERT(+/-) mice than in adult SERT(+/+) or SERT(+/-) mice. Desipramine 0-11 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 57-61 26250357-8 2015 Desipramine was less potent in middle-aged SERT(+/+) and SERT(+/-) mice than in adult SERT(+/+) or SERT(+/-) mice. Desipramine 0-11 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 57-61 26250357-8 2015 Desipramine was less potent in middle-aged SERT(+/+) and SERT(+/-) mice than in adult SERT(+/+) or SERT(+/-) mice. Desipramine 0-11 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 57-61 25639149-10 2015 We also found that desipramine inhibits the increase in membrane aquaporin-5 level triggered by carbachol and histamine treatments. Desipramine 19-30 aquaporin 5 Homo sapiens 65-76 26135316-5 2015 The ASM inhibitor desipramine significantly reduced EGCG-induced cell death. Desipramine 18-29 sphingomyelin phosphodiesterase 1 Homo sapiens 4-7 25873594-5 2015 We exemplify our approach by measuring the kinetics of cocaine, methylphenidate, and desipramine binding to SERT and DAT. Desipramine 85-96 solute carrier family 6 member 4 Homo sapiens 108-112 25873594-5 2015 We exemplify our approach by measuring the kinetics of cocaine, methylphenidate, and desipramine binding to SERT and DAT. Desipramine 85-96 solute carrier family 6 member 3 Homo sapiens 117-120 25873594-6 2015 Our analysis revealed that the selectivity of methylphenidate and desipramine for DAT and SERT, respectively, can be accounted for by their rate of association and not by the residence time in their respective binding sites. Desipramine 66-77 solute carrier family 6 member 3 Homo sapiens 82-85 25873594-6 2015 Our analysis revealed that the selectivity of methylphenidate and desipramine for DAT and SERT, respectively, can be accounted for by their rate of association and not by the residence time in their respective binding sites. Desipramine 66-77 solute carrier family 6 member 4 Homo sapiens 90-94 25840741-4 2015 The results showed that short- (3 days) or long-term (21 days) systemic treatment with the tricyclic ADs imipramine, clomipramine or desipramine increased BDNF levels in the DPAG. Desipramine 133-144 brain-derived neurotrophic factor Rattus norvegicus 155-159 25771452-6 2015 MMC and MDMA both induced concentration-dependently [(3)H]1-methyl-4-phenylpyridinium-release from NET-, DAT or SERT-expressing cells which was clearly transporter-mediated release as demonstrated by the selective inhibitory effects of nmolar to low micromolar concentrations of desipramine, GBR 12909 and fluoxetine, respectively. Desipramine 279-290 solute carrier family 6 member 4 Homo sapiens 112-116 25903729-0 2015 Changes in blood CD4+T and CD8+T lymphocytes in stressed rats pretreated chronically with desipramine are more pronounced after chronic open field stress challenge. Desipramine 90-101 Cd4 molecule Rattus norvegicus 17-20 25637091-3 2015 Systemic injections of antidepressants: the tricyclic antidepressant desipramine (DMI), the selective serotonin reuptake inhibitor fluoxetine, and the NMDA-antagonist ketamine selectively increased cFos expression (a marker of neuronal activity) in the deep layers of the ventromedial PFC (vmPFC) in rats. Desipramine 69-80 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-202 25637091-3 2015 Systemic injections of antidepressants: the tricyclic antidepressant desipramine (DMI), the selective serotonin reuptake inhibitor fluoxetine, and the NMDA-antagonist ketamine selectively increased cFos expression (a marker of neuronal activity) in the deep layers of the ventromedial PFC (vmPFC) in rats. Desipramine 82-85 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 198-202 25639149-11 2015 CONCLUSIONS: These results imply that desipramine blocks muscarinic and histamine receptor-mediated Ca(2+) signaling and the subsequent translocation of aquaporin-5 in human salivary gland cells, suggesting a novel mechanism for the xerogenic effects of desipramine. Desipramine 38-49 aquaporin 5 Homo sapiens 153-164 25639149-11 2015 CONCLUSIONS: These results imply that desipramine blocks muscarinic and histamine receptor-mediated Ca(2+) signaling and the subsequent translocation of aquaporin-5 in human salivary gland cells, suggesting a novel mechanism for the xerogenic effects of desipramine. Desipramine 254-265 aquaporin 5 Homo sapiens 153-164 25855965-7 2015 Inhibition of ceramide accumulation in lysosomes using an ASM inhibitor, desipramine, decreased cytosolic activation of CTSB by inhibiting its transfer into cytosol from the lysosome. Desipramine 73-84 H19 imprinted maternally expressed transcript Homo sapiens 58-61 25855965-7 2015 Inhibition of ceramide accumulation in lysosomes using an ASM inhibitor, desipramine, decreased cytosolic activation of CTSB by inhibiting its transfer into cytosol from the lysosome. Desipramine 73-84 cathepsin B Homo sapiens 120-124 25451113-3 2015 Moreover, biochemical studies have revealed the up-regulation of other monoamine transporters (dopamine and serotonin) in the brains of NET-KO mice, similar to the phenomenon observed after the chronic pharmacological blockade of norepinephrine transporter by desipramine in wild-type (WT) animals. Desipramine 260-271 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 230-256 25038868-0 2015 Effects of desipramine treatment on stress-induced up-regulation of norepinephrine transporter expression in rat brains. Desipramine 11-22 solute carrier family 6 member 2 Rattus norvegicus 68-94 25296725-0 2015 Evaluation of the likelihood of a selective CHK1 inhibitor (LY2603618) to inhibit CYP2D6 with desipramine as a probe substrate in cancer patients. Desipramine 94-105 checkpoint kinase 1 Homo sapiens 44-48 25296725-2 2015 The study described was to assess the potential interaction between LY2603618 and cytochrome P450 isoform 2D6 (CYP2D6) substrate desipramine in patients with cancer. Desipramine 129-140 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 82-109 25296725-2 2015 The study described was to assess the potential interaction between LY2603618 and cytochrome P450 isoform 2D6 (CYP2D6) substrate desipramine in patients with cancer. Desipramine 129-140 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 111-117 25038868-1 2015 RATIONALE: Many studies demonstrate down-regulation of the norepinephrine transporter (NET) by desipramine (DMI) in vitro and in stress-naive rats. Desipramine 95-106 solute carrier family 6 member 2 Rattus norvegicus 59-85 25038868-1 2015 RATIONALE: Many studies demonstrate down-regulation of the norepinephrine transporter (NET) by desipramine (DMI) in vitro and in stress-naive rats. Desipramine 108-111 solute carrier family 6 member 2 Rattus norvegicus 59-85 25463972-0 2014 Leptin receptor deficiency confers resistance to behavioral effects of fluoxetine and desipramine via separable substrates. Desipramine 86-97 leptin Mus musculus 0-6 25463972-5 2014 In this study, we determined the functional role of the leptin receptor (LepRb) in behavioral responses to the selective serotonergic antidepressant fluoxetine and the noradrenergic antidepressant desipramine. Desipramine 197-208 leptin Mus musculus 56-62 25463972-5 2014 In this study, we determined the functional role of the leptin receptor (LepRb) in behavioral responses to the selective serotonergic antidepressant fluoxetine and the noradrenergic antidepressant desipramine. Desipramine 197-208 leptin receptor Mus musculus 73-78 25463972-6 2014 While acute and chronic treatment with fluoxetine or desipramine in wild-type mice elicited antidepressant-like effects in the forced swim test, mice null for LepRb (db/db) displayed resistance to treatment with either fluoxetine or desipramine. Desipramine 233-244 leptin receptor Mus musculus 159-164 25463972-9 2014 Deletion of LepRb specifically from hippocampal and cortical neurons resulted in fluoxetine insensitivity in the forced swim test and tail suspension test while leaving the response to desipramine intact. Desipramine 185-196 leptin receptor Mus musculus 12-17 25463972-10 2014 These results suggest that functional LepRb is critically involved in regulating the antidepressant-like behavioral effects of both fluoxetine and desipramine. Desipramine 147-158 leptin receptor Mus musculus 38-43 25208966-0 2014 Norepinephrine transporter inhibition with desipramine exacerbates L-DOPA-induced dyskinesia: role for synaptic dopamine regulation in denervated nigrostriatal terminals. Desipramine 43-54 solute carrier family 6 member 2 Rattus norvegicus 0-26 25414111-2 2014 Circulating ghrelin concentrations could potentially be used as a predictor of DMI in cattle. Desipramine 79-82 ghrelin and obestatin prepropeptide Bos taurus 12-19 25414111-3 2014 The objective of this experiment was to determine the association of circulating ghrelin concentrations with DMI and other production traits. Desipramine 109-112 ghrelin and obestatin prepropeptide Bos taurus 81-88 25414111-6 2014 Active ghrelin was not correlated to DMI (P=0.36), but when DMI was modeled using a multivariate analysis including plasma metabolites and sex, active ghrelin was shown to be positively associated with DMI (P<0.01) and accounted for 6.2% of the variation accounted for by the regression model (R2=0.33). Desipramine 60-63 ghrelin and obestatin prepropeptide Bos taurus 151-158 25414111-6 2014 Active ghrelin was not correlated to DMI (P=0.36), but when DMI was modeled using a multivariate analysis including plasma metabolites and sex, active ghrelin was shown to be positively associated with DMI (P<0.01) and accounted for 6.2% of the variation accounted for by the regression model (R2=0.33). Desipramine 60-63 ghrelin and obestatin prepropeptide Bos taurus 151-158 25414111-7 2014 Total ghrelin was negatively correlated to DMI (P<0.01), but was not significant in a multivariate regression analysis (P=0.13). Desipramine 43-46 ghrelin and obestatin prepropeptide Bos taurus 6-13 25414111-8 2014 The ratio of active:total ghrelin was positively associated with DMI (P<0.01) and accounted for 10.2% of the variation in the model (R2=0.35). Desipramine 65-68 ghrelin and obestatin prepropeptide Bos taurus 26-33 25414111-14 2014 Results indicated that ghrelin concentrations are associated with DMI in beef cattle and that there is genetic variation that leads to differences in the amount and form of circulating ghrelin which could contribute to variation observed in DMI of beef cattle. Desipramine 66-69 ghrelin and obestatin prepropeptide Bos taurus 23-30 25414111-14 2014 Results indicated that ghrelin concentrations are associated with DMI in beef cattle and that there is genetic variation that leads to differences in the amount and form of circulating ghrelin which could contribute to variation observed in DMI of beef cattle. Desipramine 241-244 ghrelin and obestatin prepropeptide Bos taurus 185-192 25373053-8 2014 Vasopressin alone was shown to be the most effective treatment in the management of desipramine overdose. Desipramine 84-95 vasopressin Sus scrofa 0-11 24576687-3 2014 METHODS: This study assessed the following: 1) brain reward function in rats (using the intracranial self-stimulation procedure) and protein levels of brain-derived neurotrophic factor and related signaling molecules in response to chronic social defeat, 2) brain reward function during social defeat and long-term treatment with the antidepressants fluoxetine (5 mg/kg/day) and desipramine (10 mg/kg/day), and 3) forced swim test behavior after social defeat and fluoxetine treatment. Desipramine 379-390 brain-derived neurotrophic factor Rattus norvegicus 151-184 25129807-4 2014 This increase in prodynorphin immunoreactivity in the dBNST and CeL was blocked by desipramine and citalopram treatments. Desipramine 83-94 carboxyl ester lipase Mus musculus 64-67 25048970-9 2014 GSH loss was associated with an increase in acid sphingomyelinase (ASMase) activity and lipid raft formation, which could be inhibited by the ASMase inhibitor desipramine. Desipramine 159-170 sphingomyelin phosphodiesterase 1 Homo sapiens 44-65 25048970-9 2014 GSH loss was associated with an increase in acid sphingomyelinase (ASMase) activity and lipid raft formation, which could be inhibited by the ASMase inhibitor desipramine. Desipramine 159-170 sphingomyelin phosphodiesterase 1 Homo sapiens 67-73 25048970-9 2014 GSH loss was associated with an increase in acid sphingomyelinase (ASMase) activity and lipid raft formation, which could be inhibited by the ASMase inhibitor desipramine. Desipramine 159-170 sphingomyelin phosphodiesterase 1 Homo sapiens 142-148 25048970-10 2014 27OH-C and total lipids from LDL and oxLDL independently increased Abeta production by SH-SY5Y cells, and Abeta accumulation could be inhibited by desipramine and by N-acetylcysteine. Desipramine 147-158 amyloid beta precursor protein Homo sapiens 106-111 25129807-8 2014 These results suggest that regulation of prodynorphin in the dBNST and CeL before the test swim may be involved in the antidepressant-like effect of desipramine and citalopram in the FST and suggest that changes in pCREB immunoreactivity in these areas may not play an important role in the regulation of prodynorphin in the dBNST and CeA. Desipramine 149-160 carboxyl ester lipase Mus musculus 71-74 24880753-4 2014 Desipramine was selected as a model drug because it is mainly metabolized by CYP2D6, which is inhibited by almorexant in vitro. Desipramine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 77-83 24951586-5 2014 Moreover, treating cells with siRNA to ASM or with the indirect pharmacologic inhibitor desipramine resulted in significant inhibition of TNFalpha- and PMA-induced IL-6 production in MDA-MB-231 and HeLa cells. Desipramine 88-99 tumor necrosis factor Homo sapiens 138-146 24951586-5 2014 Moreover, treating cells with siRNA to ASM or with the indirect pharmacologic inhibitor desipramine resulted in significant inhibition of TNFalpha- and PMA-induced IL-6 production in MDA-MB-231 and HeLa cells. Desipramine 88-99 interleukin 6 Homo sapiens 164-168 24951586-7 2014 Further, ASM knockdown or desipramine blunted p38 MAPK activation in response to TNFalpha, revealing a key role for ASM in activating p38, a signaling pathway known to regulate IL-6 induction. Desipramine 26-37 mitogen-activated protein kinase 14 Homo sapiens 46-49 24951586-7 2014 Further, ASM knockdown or desipramine blunted p38 MAPK activation in response to TNFalpha, revealing a key role for ASM in activating p38, a signaling pathway known to regulate IL-6 induction. Desipramine 26-37 tumor necrosis factor Homo sapiens 81-89 25247592-8 2014 Serum levels of interleukin-1beta (IL-1beta) and IL-6 were elevated in the PTSD-like group compared with the control group, and were decreased by MPH, FLU, DES or the combination drug treatments, with the combination of DES+MPH producing the most complete rescue of the inflammatory response. Desipramine 156-159 interleukin 1 beta Rattus norvegicus 16-33 25247592-8 2014 Serum levels of interleukin-1beta (IL-1beta) and IL-6 were elevated in the PTSD-like group compared with the control group, and were decreased by MPH, FLU, DES or the combination drug treatments, with the combination of DES+MPH producing the most complete rescue of the inflammatory response. Desipramine 156-159 interleukin 1 beta Rattus norvegicus 35-43 25247592-8 2014 Serum levels of interleukin-1beta (IL-1beta) and IL-6 were elevated in the PTSD-like group compared with the control group, and were decreased by MPH, FLU, DES or the combination drug treatments, with the combination of DES+MPH producing the most complete rescue of the inflammatory response. Desipramine 156-159 interleukin 6 Rattus norvegicus 49-53 25247592-8 2014 Serum levels of interleukin-1beta (IL-1beta) and IL-6 were elevated in the PTSD-like group compared with the control group, and were decreased by MPH, FLU, DES or the combination drug treatments, with the combination of DES+MPH producing the most complete rescue of the inflammatory response. Desipramine 220-223 interleukin 1 beta Rattus norvegicus 16-33 25247592-8 2014 Serum levels of interleukin-1beta (IL-1beta) and IL-6 were elevated in the PTSD-like group compared with the control group, and were decreased by MPH, FLU, DES or the combination drug treatments, with the combination of DES+MPH producing the most complete rescue of the inflammatory response. Desipramine 220-223 interleukin 1 beta Rattus norvegicus 35-43 25247592-8 2014 Serum levels of interleukin-1beta (IL-1beta) and IL-6 were elevated in the PTSD-like group compared with the control group, and were decreased by MPH, FLU, DES or the combination drug treatments, with the combination of DES+MPH producing the most complete rescue of the inflammatory response. Desipramine 220-223 interleukin 6 Rattus norvegicus 49-53 24648162-1 2014 Proteasome modulator 9 (PSMD9) gene single nucleotide polymorphism (SNP) rs1043307/rs2514259 (E197G) is associated with significant clinical response to the anti-depressant desipramine. Desipramine 173-184 proteasome 26S subunit, non-ATPase 9 Homo sapiens 0-22 24648162-1 2014 Proteasome modulator 9 (PSMD9) gene single nucleotide polymorphism (SNP) rs1043307/rs2514259 (E197G) is associated with significant clinical response to the anti-depressant desipramine. Desipramine 173-184 proteasome 26S subunit, non-ATPase 9 Homo sapiens 24-29 25128275-3 2014 Our previous work established a particular TCA, desipramine, as an arrestin-biased alpha2AAR ligand driving receptor endocytosis and downregulation but not canonical heterotrimeric G protein-mediated signaling. Desipramine 48-59 adrenoceptor alpha 2A Homo sapiens 83-92 24657329-8 2014 Sertraline and desipramine tended to inhibit OCT2 activity via a competitive mechanism. Desipramine 15-26 solute carrier family 22 member 2 Homo sapiens 45-49 24880753-7 2014 Almorexant increased the exposure to desipramine 3.7-fold, suggesting that almorexant is a moderate inhibitor of desipramine metabolism through inhibition of CYP2D6. Desipramine 37-48 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 158-164 24880753-7 2014 Almorexant increased the exposure to desipramine 3.7-fold, suggesting that almorexant is a moderate inhibitor of desipramine metabolism through inhibition of CYP2D6. Desipramine 113-124 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 158-164 24748481-6 2014 These results suggest common and distinct regulation of CART by desipramine and citalopram treatments in the FST and raise the possibility that CART in the AcbSh, dBNST, and CeA may be involved in antidepressant-like effect in the FST. Desipramine 64-75 CART prepropeptide Mus musculus 56-60 24742525-0 2014 The tricyclic antidepressant desipramine inhibited the neurotoxic, kainate-induced [Ca(2+)]i increases in CA1 pyramidal cells in acute hippocampal slices. Desipramine 29-40 carbonic anhydrase 1 Rattus norvegicus 106-109 24742525-3 2014 We used [Ca(2+)]i imaging and patch clamp electrophysiology to decipher the mechanism of KA-evoked [Ca(2+)]i rise and its inhibition by the tricyclic antidepressant desipramine (DMI) in CA1 pyramidal cells in rat hippocampal slices and in cultured hippocampal cells. Desipramine 165-176 carbonic anhydrase 1 Rattus norvegicus 186-189 24528439-0 2014 Acute desipramine restores presynaptic cortical defects in murine experimental autoimmune encephalomyelitis by suppressing central CCL5 overproduction. Desipramine 6-17 chemokine (C-C motif) ligand 5 Mus musculus 131-135 24528439-11 2014 Acute, but not chronic, DMI reduced CCL5 levels in cortical homogenates of EAE mice at 13 d.p.i., but did not affect peripheral IL-17 and TNF-alpha contents or CCL5 plasma levels. Desipramine 24-27 chemokine (C-C motif) ligand 5 Mus musculus 36-40 24528439-12 2014 Acute DMI caused a long-lasting restoration of glutamate exocytosis, restored endogenous cAMP production and impeded the shift from inhibition to facilitation of the CCL5-mediated control of glutamate exocytosis. Desipramine 6-9 chemokine (C-C motif) ligand 5 Mus musculus 166-170 24663212-3 2014 Phenotypic RFI was calculated as actual DMI minus expected DMI. Desipramine 40-43 RFI Bos taurus 11-14 24663212-3 2014 Phenotypic RFI was calculated as actual DMI minus expected DMI. Desipramine 59-62 RFI Bos taurus 11-14 24663212-8 2014 Live weight and ADG were not correlated (P > 0.05), and DMI was positively correlated (r = 0.80; P < 0.001) with RFI. Desipramine 59-62 RFI Bos taurus 119-122 24748481-0 2014 Effect of desipramine and citalopram treatment on forced swimming test-induced changes in cocaine- and amphetamine-regulated transcript (CART) immunoreactivity in mice. Desipramine 10-21 CART prepropeptide Mus musculus 90-135 24748481-0 2014 Effect of desipramine and citalopram treatment on forced swimming test-induced changes in cocaine- and amphetamine-regulated transcript (CART) immunoreactivity in mice. Desipramine 10-21 CART prepropeptide Mus musculus 137-141 24748481-2 2014 To explore this possibility, we assessed the treatment effects of desipramine and citalopram, which inhibit the reuptake of norepinephrine and serotonin into the presynaptic terminals, respectively, on changes in levels of CART-IR before and after the test swim in mouse brain. Desipramine 66-77 CART prepropeptide Mus musculus 223-227 24748481-4 2014 This increase in CART-IR in the AcbSh, dBNST, and PVN before the test swim remained elevated by desipramine treatment after the test swim, but this increase in these brain areas returned to near control levels after test swim by citalopram treatment. Desipramine 96-107 CART prepropeptide Mus musculus 17-21 24461377-3 2014 Chronic desipramine pretreatment did not protect against OF stress-induced suppression of blood anti-tumor natural killer cell activity but increased plasma interleukin-10 and decreased interferon-gamma and corticosterone concentration. Desipramine 8-19 interleukin 10 Rattus norvegicus 157-171 24461377-3 2014 Chronic desipramine pretreatment did not protect against OF stress-induced suppression of blood anti-tumor natural killer cell activity but increased plasma interleukin-10 and decreased interferon-gamma and corticosterone concentration. Desipramine 8-19 interferon gamma Rattus norvegicus 186-202 24132698-0 2014 Dexamethasone in the presence of desipramine enhances MAPK/ERK1/2 signaling possibly via its interference with beta-arrestin. Desipramine 33-44 mitogen-activated protein kinase 3 Homo sapiens 59-65 24730055-0 2014 Chronic desipramine treatment rescues depression-related, social and cognitive deficits in Engrailed-2 knockout mice. Desipramine 8-19 engrailed 2 Mus musculus 91-102 24730055-4 2014 This study evaluated the ability of chronic treatment with desipramine (DMI), a selective norepinephrine (NE) reuptake inhibitor and classical antidepressant, to reverse behavioral abnormalities in En2-/- mice. Desipramine 59-70 engrailed 2 Mus musculus 198-201 24730055-4 2014 This study evaluated the ability of chronic treatment with desipramine (DMI), a selective norepinephrine (NE) reuptake inhibitor and classical antidepressant, to reverse behavioral abnormalities in En2-/- mice. Desipramine 72-75 engrailed 2 Mus musculus 198-201 24730055-5 2014 Desipramine treatment significantly reduced immobility in the tail suspension and forced swim tests, restored sociability in the three-chambered social approach task and reversed impairments in contextual fear conditioning in En2-/- mice. Desipramine 0-11 engrailed 2 Mus musculus 226-229 25562160-4 2014 The present study was designed to investigate effects of desipramine (DES), as an ASMase inhibitor, on Cr(VI)-induced hepatotoxicity. Desipramine 57-68 sphingomyelin phosphodiesterase 1 Homo sapiens 82-88 25562160-4 2014 The present study was designed to investigate effects of desipramine (DES), as an ASMase inhibitor, on Cr(VI)-induced hepatotoxicity. Desipramine 70-73 sphingomyelin phosphodiesterase 1 Homo sapiens 82-88 25562160-6 2014 Moreover, the study investigated the role of DES played in ASMase activities and ceramide levels. Desipramine 45-48 sphingomyelin phosphodiesterase 1 Homo sapiens 59-65 25562160-9 2014 The results demonstrated that apoptosis rates, ASMase activities and ceramide content decreased in groups treated with the combination of DES and Cr(VI) compared to Cr(VI) groups. Desipramine 138-141 sphingomyelin phosphodiesterase 1 Homo sapiens 47-53 24268836-12 2014 Milk production decreased on the 2 feather meal treatments, partly explained by a tendency for DMI to decrease. Desipramine 95-98 Weaning weight-maternal milk Bos taurus 0-4 24132698-3 2014 In the present study, we show that the glucocorticoid receptor (GR) agonist dexamethasone (DEX) increased MAPK/ERK1/2 signaling in the presence of the noradrenergic antidepressant, desipramine (DMI), while no such effect was induced by DEX or DMI alone in human neuroblastoma SH-SY5Y cells. Desipramine 181-192 nuclear receptor subfamily 3 group C member 1 Homo sapiens 39-62 24132698-3 2014 In the present study, we show that the glucocorticoid receptor (GR) agonist dexamethasone (DEX) increased MAPK/ERK1/2 signaling in the presence of the noradrenergic antidepressant, desipramine (DMI), while no such effect was induced by DEX or DMI alone in human neuroblastoma SH-SY5Y cells. Desipramine 181-192 nuclear receptor subfamily 3 group C member 1 Homo sapiens 64-66 24132698-3 2014 In the present study, we show that the glucocorticoid receptor (GR) agonist dexamethasone (DEX) increased MAPK/ERK1/2 signaling in the presence of the noradrenergic antidepressant, desipramine (DMI), while no such effect was induced by DEX or DMI alone in human neuroblastoma SH-SY5Y cells. Desipramine 194-197 nuclear receptor subfamily 3 group C member 1 Homo sapiens 39-62 24132698-3 2014 In the present study, we show that the glucocorticoid receptor (GR) agonist dexamethasone (DEX) increased MAPK/ERK1/2 signaling in the presence of the noradrenergic antidepressant, desipramine (DMI), while no such effect was induced by DEX or DMI alone in human neuroblastoma SH-SY5Y cells. Desipramine 194-197 nuclear receptor subfamily 3 group C member 1 Homo sapiens 64-66 24132698-3 2014 In the present study, we show that the glucocorticoid receptor (GR) agonist dexamethasone (DEX) increased MAPK/ERK1/2 signaling in the presence of the noradrenergic antidepressant, desipramine (DMI), while no such effect was induced by DEX or DMI alone in human neuroblastoma SH-SY5Y cells. Desipramine 243-246 nuclear receptor subfamily 3 group C member 1 Homo sapiens 39-62 24132698-3 2014 In the present study, we show that the glucocorticoid receptor (GR) agonist dexamethasone (DEX) increased MAPK/ERK1/2 signaling in the presence of the noradrenergic antidepressant, desipramine (DMI), while no such effect was induced by DEX or DMI alone in human neuroblastoma SH-SY5Y cells. Desipramine 243-246 nuclear receptor subfamily 3 group C member 1 Homo sapiens 64-66 24132698-10 2014 We suggest that such an interaction in the presence of DMI can enhance MAPK/ERK1/2 signaling, a key player in neural plasticity and neurogenesis processes, which is impaired in MDD, while stimulated by antidepressants. Desipramine 55-58 mitogen-activated protein kinase 3 Homo sapiens 76-82 23906970-4 2013 Twenty four-hour incubation of cells with antidepressants (0.05-10 muM) and DEX (10 muM) showed that imipramine, amitriptyline, desipramine, citalopram and fluoxetine at concentrations from 0.1 up to 1 muM, reboxetine (0.1 muM) and tianeptine (0.05 muM) prevented the DEX-induced decreases in cell viability and proliferation rate. Desipramine 128-139 latexin Homo sapiens 67-70 23906970-4 2013 Twenty four-hour incubation of cells with antidepressants (0.05-10 muM) and DEX (10 muM) showed that imipramine, amitriptyline, desipramine, citalopram and fluoxetine at concentrations from 0.1 up to 1 muM, reboxetine (0.1 muM) and tianeptine (0.05 muM) prevented the DEX-induced decreases in cell viability and proliferation rate. Desipramine 128-139 latexin Homo sapiens 84-87 23906970-4 2013 Twenty four-hour incubation of cells with antidepressants (0.05-10 muM) and DEX (10 muM) showed that imipramine, amitriptyline, desipramine, citalopram and fluoxetine at concentrations from 0.1 up to 1 muM, reboxetine (0.1 muM) and tianeptine (0.05 muM) prevented the DEX-induced decreases in cell viability and proliferation rate. Desipramine 128-139 latexin Homo sapiens 84-87 23906970-4 2013 Twenty four-hour incubation of cells with antidepressants (0.05-10 muM) and DEX (10 muM) showed that imipramine, amitriptyline, desipramine, citalopram and fluoxetine at concentrations from 0.1 up to 1 muM, reboxetine (0.1 muM) and tianeptine (0.05 muM) prevented the DEX-induced decreases in cell viability and proliferation rate. Desipramine 128-139 latexin Homo sapiens 84-87 23906970-4 2013 Twenty four-hour incubation of cells with antidepressants (0.05-10 muM) and DEX (10 muM) showed that imipramine, amitriptyline, desipramine, citalopram and fluoxetine at concentrations from 0.1 up to 1 muM, reboxetine (0.1 muM) and tianeptine (0.05 muM) prevented the DEX-induced decreases in cell viability and proliferation rate. Desipramine 128-139 latexin Homo sapiens 84-87 23486447-3 2013 These drugs and their metabolites, along with desipramine and nortriptyline, undergo hydroxylation by CYP2D6 to less active metabolites. Desipramine 46-57 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 102-108 23620084-5 2013 A significantly higher level of C/EBPalpha was measured at 48 h, but the levels of C/EBPbeta increased after 12 h. Levels of FABP4 significantly increased with the time of incubation after 12 h, but that of LPL was reduced (P < 0.05) at 6, 24 and 48 h. FABP4 was highly expressed in cells treated with CS, DMI + CS and DMIOA + CS compared to cells treated with FBS, DMI + FBS and DMIOA + FBS. Desipramine 309-312 fatty acid binding protein 4 Homo sapiens 125-130 23946692-0 2013 Changes in c-Fos Expression in the Forced Swimming Test: Common and Distinct Modulation in Rat Brain by Desipramine and Citalopram. Desipramine 104-115 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-16 23946692-5 2013 Both desipramine and citalopram treatment after the pretest swim significantly increased the number of c-Fos-like immunoreactive cells in the ventral lateral septum and ventrolateral periaqueductal gray before the test swim. Desipramine 5-16 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 103-108 23389997-7 2013 Furthermore, treatment with desipramine, an antidepressant with specific inhibitory effects on norepinephrine transport, prevented an increased dopamine beta-hydroxylase expression by chronic social defeat in the locus coeruleus and its main terminal regions such as the hippocampus, frontal cortex and amygdala. Desipramine 28-39 dopamine beta-hydroxylase Rattus norvegicus 144-169 24309563-7 2013 In DMI-treated mice, tumor VEGF, IL-6, and the prometastatic chemokines RANTES, M-CSF, and MIP-2 were reduced. Desipramine 3-6 vascular endothelial growth factor A Mus musculus 27-31 24309563-7 2013 In DMI-treated mice, tumor VEGF, IL-6, and the prometastatic chemokines RANTES, M-CSF, and MIP-2 were reduced. Desipramine 3-6 interleukin 6 Mus musculus 33-37 24309563-7 2013 In DMI-treated mice, tumor VEGF, IL-6, and the prometastatic chemokines RANTES, M-CSF, and MIP-2 were reduced. Desipramine 3-6 chemokine (C-C motif) ligand 5 Mus musculus 72-78 24309563-7 2013 In DMI-treated mice, tumor VEGF, IL-6, and the prometastatic chemokines RANTES, M-CSF, and MIP-2 were reduced. Desipramine 3-6 colony stimulating factor 1 (macrophage) Mus musculus 80-85 24309563-7 2013 In DMI-treated mice, tumor VEGF, IL-6, and the prometastatic chemokines RANTES, M-CSF, and MIP-2 were reduced. Desipramine 3-6 chemokine (C-X-C motif) ligand 2 Mus musculus 91-96 24161357-0 2013 Chronic antidepressant desipramine treatment increases open field-induced brain expression and spleen production of interleukin 10 in rats. Desipramine 23-34 interleukin 10 Rattus norvegicus 116-130 24161357-4 2013 In the desipramine pretreated groups, OF stress decreased spleen NKCC, behavioral activity, the Con A-stimulated splenocyte IFN-gamma response and plasma corticosterone concentration whereas it increased the brain and splenocyte IL-10 response. Desipramine 7-18 interferon gamma Rattus norvegicus 124-133 24161357-4 2013 In the desipramine pretreated groups, OF stress decreased spleen NKCC, behavioral activity, the Con A-stimulated splenocyte IFN-gamma response and plasma corticosterone concentration whereas it increased the brain and splenocyte IL-10 response. Desipramine 7-18 interleukin 10 Rattus norvegicus 229-234 24406179-12 2013 CONCLUSION: TNF-alpha can induce DUOX-1 expression increasing in lipid raft, then the DUOX-1 can be activated to increase reactive oxygen species level; acidic sphingomyelinase inhibitor desipramine can inhibit this process, the results disclose that the process will depend on the ceramide of lipid raft. Desipramine 187-198 tumor necrosis factor Homo sapiens 12-21 24406179-12 2013 CONCLUSION: TNF-alpha can induce DUOX-1 expression increasing in lipid raft, then the DUOX-1 can be activated to increase reactive oxygen species level; acidic sphingomyelinase inhibitor desipramine can inhibit this process, the results disclose that the process will depend on the ceramide of lipid raft. Desipramine 187-198 dual oxidase 1 Homo sapiens 33-39 24406179-12 2013 CONCLUSION: TNF-alpha can induce DUOX-1 expression increasing in lipid raft, then the DUOX-1 can be activated to increase reactive oxygen species level; acidic sphingomyelinase inhibitor desipramine can inhibit this process, the results disclose that the process will depend on the ceramide of lipid raft. Desipramine 187-198 dual oxidase 1 Homo sapiens 86-92 23620084-5 2013 A significantly higher level of C/EBPalpha was measured at 48 h, but the levels of C/EBPbeta increased after 12 h. Levels of FABP4 significantly increased with the time of incubation after 12 h, but that of LPL was reduced (P < 0.05) at 6, 24 and 48 h. FABP4 was highly expressed in cells treated with CS, DMI + CS and DMIOA + CS compared to cells treated with FBS, DMI + FBS and DMIOA + FBS. Desipramine 322-325 fatty acid binding protein 4 Homo sapiens 125-130 23508024-7 2013 Substitution of the G allele of SNP rs42670353 was associated with ADG (0.043 +- 0.02 kg/d; P < 0.01) and FCR (0.114 +- 0.05 kg BW gain kg(-1) DMI; P < 0.05). Desipramine 146-149 ADG Bos taurus 67-70 23508024-7 2013 Substitution of the G allele of SNP rs42670353 was associated with ADG (0.043 +- 0.02 kg/d; P < 0.01) and FCR (0.114 +- 0.05 kg BW gain kg(-1) DMI; P < 0.05). Desipramine 146-149 FCR Bos taurus 109-112 22422821-6 2012 Whereas desipramine did not by itself elicit circulating HSPCs, it increased G-CSF-triggered mobilization efficiency significantly and rescued mobilization in a model mimicking "poor mobilizers." Desipramine 8-19 peripheral blood stem cell response to granulocyte colony stimulating factor 1 Mus musculus 77-82 23582643-7 2013 Similarly, the cyclophilin D-inhibiting drug alisporivir and the acid sphingomyelinase-inactivating drug, desipramine, synergize to reverse susceptibility, suggesting the therapeutic potential of these orally active drugs against tuberculosis and possibly other TNF-mediated diseases. Desipramine 106-117 tumor necrosis factor b (TNF superfamily, member 2) Danio rerio 262-265 24399720-4 2013 RESULTS: The investigated antidepressant drugs added to control liver microsomes produced certain inhibitory effects on CYP2C11 activity, which were moderate (sertraline, nefazodone and clomipramine: Ki = 39, 56 and 66 muM, respectively), modest (fluoxetine and amitriptyline: Ki = 98 and 108 muM, respectively) or weak (imipramine and desipramine: Ki = 191 and 212 muM, respectively). Desipramine 336-347 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 120-127 24399720-6 2013 One-day exposure of rats to the antidepressant drugs did not significantly change the activity of CYP2C11 in liver microsomes; however, imipramine, desipramine and fluoxetine showed a tendency to diminish the activity of CYP2C11. Desipramine 148-159 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 221-228 24399720-7 2013 Of the antidepressants studied, only desipramine and fluoxetine administered chronically elevated CYP2C11 activity; those effects were positively correlated with the observed increases in the enzyme protein level. Desipramine 37-48 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 98-105 22250926-3 2012 Therefore, the present study aimed to investigate the effect of P-gp inhibition on the transport of the tricyclic antidepressant imipramine and its active metabolite desipramine across the BBB. Desipramine 166-177 ATP-binding cassette, subfamily B (MDR/TAP), member 1B Rattus norvegicus 64-68 23221858-1 2013 The results of two single-center, two-period, open-label trials that evaluated the effects of multiple doses of desvenlafaxine on the pharmacokinetics of desipramine, a cytochrome P450 (CYP) 2D6 enzyme substrate, are presented. Desipramine 154-165 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 169-194 22934693-4 2013 Our previous study revealed that DMI at the clinical relevant concentrations could induce CHOP-dependent apoptotic death in C6 glioma cells. Desipramine 33-36 DNA damage inducible transcript 3 Homo sapiens 90-94 22934693-6 2013 Treatment with DMI could induce autophagic cell death characterized by the formation of autophagosome and the elevated level of autophagic protein Beclin-1 and cellular redistribution of marker LC3. Desipramine 15-18 beclin 1 Homo sapiens 147-155 22934693-7 2013 Meanwhile, DMI inhibited the activation of PI3K-AKT-mTOR pathway which is considered as a negative regulator of autophagy. Desipramine 11-14 mechanistic target of rapamycin kinase Homo sapiens 52-56 22934693-8 2013 Furthermore, DMI activated PERK-eIF2alpha and ATF6 of endoplasmic reticulum (ER) stress pathway, while knockdown of PERK with the PERK-specific short interfering RNA (siRNA) could obviously attenuate the autophagy. Desipramine 13-16 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 27-31 22934693-8 2013 Furthermore, DMI activated PERK-eIF2alpha and ATF6 of endoplasmic reticulum (ER) stress pathway, while knockdown of PERK with the PERK-specific short interfering RNA (siRNA) could obviously attenuate the autophagy. Desipramine 13-16 eukaryotic translation initiation factor 2A Homo sapiens 32-41 22934693-8 2013 Furthermore, DMI activated PERK-eIF2alpha and ATF6 of endoplasmic reticulum (ER) stress pathway, while knockdown of PERK with the PERK-specific short interfering RNA (siRNA) could obviously attenuate the autophagy. Desipramine 13-16 activating transcription factor 6 Homo sapiens 46-50 22934693-9 2013 The results strongly suggested that DMI could induce autophagy through the PERK-ER stress pathway in C6 glioma cells. Desipramine 36-39 eukaryotic translation initiation factor 2 alpha kinase 3 Homo sapiens 75-79 23527713-12 2013 Desipramine also suppressed the serum OVA-specific IgE and IL-4 levels, but had no effect on IFN-gamma level. Desipramine 0-11 interleukin 4 Mus musculus 59-63 23527713-15 2013 These results suggest that the antidepressant drug, desipramine, also has anti-allergic action, which was possibly achieved by reducing allergen-specific IgE and Th2 cytokine production and maintaining a balance between Treg and Th17 cells. Desipramine 52-63 heart and neural crest derivatives expressed 2 Mus musculus 162-165 22806583-6 2012 At their upper therapeutic plasma concentrations, several drugs are expected to inhibit by more than 20 % hOCT1 and could thus interfere with the pharmacokinetics of hOCT1-transported drugs in the kidney and liver, namely trimipramine, desipramine and fluoxetine (by about 37 %), levomepromazine and nefazodone (by about 32 %), and clozapine and amitriptyline (by about 22 %). Desipramine 236-247 solute carrier family 22 member 1 Homo sapiens 106-111 22806583-6 2012 At their upper therapeutic plasma concentrations, several drugs are expected to inhibit by more than 20 % hOCT1 and could thus interfere with the pharmacokinetics of hOCT1-transported drugs in the kidney and liver, namely trimipramine, desipramine and fluoxetine (by about 37 %), levomepromazine and nefazodone (by about 32 %), and clozapine and amitriptyline (by about 22 %). Desipramine 236-247 solute carrier family 22 member 1 Homo sapiens 166-171 22369787-0 2012 Noradrenergic antidepressant responses to desipramine in vivo are reciprocally regulated by arrestin3 and spinophilin. Desipramine 42-53 protein phosphatase 1, regulatory subunit 9B Mus musculus 106-117 22369787-7 2012 As well, mice deficient in arrestin3 lack antidepressant response to DMI, while spinophilin-null mice have enhanced antidepressant response to DMI compared with wild-type controls, indicating that this alpha(2A)AR-mediated response is reciprocally regulated by arrestin and spinophilin. Desipramine 143-146 protein phosphatase 1, regulatory subunit 9B Mus musculus 80-91 22369787-9 2012 Our results implicate arrestin- and spinophilin-mediated regulation of the alpha(2A)AR in the pharmacology of the noradrenergic antidepressant DMI, and suggest that manipulation of this mode of receptor regulation may represent a novel and viable therapeutic strategy. Desipramine 143-146 protein phosphatase 1, regulatory subunit 9B Mus musculus 36-47 22342816-7 2012 Additionally, desipramine had an IC(50) value of 0.7 muM for the uptake of NE by OCT3, while the IC(50) value of sertraline was 2.3 muM for 5-HT uptake. Desipramine 14-25 OCTN3 Homo sapiens 81-85 22342816-8 2012 Both desipramine and sertraline appeared to inhibit OCT3 activity via a non-competitive mechanism. Desipramine 5-16 OCTN3 Homo sapiens 52-56 22405824-0 2012 Desipramine selectively potentiates norepinephrine-elicited ERK1/2 activation through the alpha2A adrenergic receptor. Desipramine 0-11 mitogen-activated protein kinase 3 Homo sapiens 60-66 22405824-0 2012 Desipramine selectively potentiates norepinephrine-elicited ERK1/2 activation through the alpha2A adrenergic receptor. Desipramine 0-11 adrenoceptor alpha 2A Homo sapiens 90-117 22405824-2 2012 We have recently identified the tricyclic antidepressant drug desipramine (DMI) as a direct ligand at the alpha(2A) adrenergic receptor (AR) without itself driving heterotrimeric G protein/downstream effector activation [5]. Desipramine 62-73 adrenoceptor alpha 2A Homo sapiens 106-135 22405824-2 2012 We have recently identified the tricyclic antidepressant drug desipramine (DMI) as a direct ligand at the alpha(2A) adrenergic receptor (AR) without itself driving heterotrimeric G protein/downstream effector activation [5]. Desipramine 75-78 adrenoceptor alpha 2A Homo sapiens 106-135 22405824-4 2012 DMI acted as a signaling potentiator, selectively enhancing NE-induced alpha(2A)AR-mediated ERK1/2 MAPK signaling. Desipramine 0-3 mitogen-activated protein kinase 3 Homo sapiens 92-98 22405824-4 2012 DMI acted as a signaling potentiator, selectively enhancing NE-induced alpha(2A)AR-mediated ERK1/2 MAPK signaling. Desipramine 0-3 mitogen-activated protein kinase 3 Homo sapiens 99-103 22405824-6 2012 DMI in a physiologically relevant ratio with NE effectively turned on ERK1/2 signaling that is lacking in response to physiological NE alone. Desipramine 0-3 mitogen-activated protein kinase 3 Homo sapiens 70-76 22405824-7 2012 Further, the DMI-induced ERK1/2 potentiation relied on heterotrimeric G(i/o) proteins and was arrestin-independent. Desipramine 13-16 mitogen-activated protein kinase 3 Homo sapiens 25-31 21976621-4 2012 The inhibition profiles of 20 known inhibitors of CYP2D6 were characterized in vitro against four clinically relevant CYP2D6 substrates (desipramine, dextromethorphan, metoprolol, and thioridazine) and bufuralol. Desipramine 137-148 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 50-56 22442074-6 2012 Finally, the depressive-like behaviors in BDNF(+/Met) mice could be selectively rescued by acute administration of desipramine but not fluoxetine. Desipramine 115-126 brain derived neurotrophic factor Mus musculus 42-46 22442074-8 2012 Importantly, desipramine but not fluoxetine has antidepressant effects on BDNF(+/Met) mice, suggesting that specific classes of antidepressant may be a more effective treatment option for depressive symptoms in humans with this genetic variant BDNF. Desipramine 13-24 brain derived neurotrophic factor Mus musculus 74-78 22442074-8 2012 Importantly, desipramine but not fluoxetine has antidepressant effects on BDNF(+/Met) mice, suggesting that specific classes of antidepressant may be a more effective treatment option for depressive symptoms in humans with this genetic variant BDNF. Desipramine 13-24 brain derived neurotrophic factor Homo sapiens 244-248 22089316-12 2012 This study was registered with ClinicalTrials.gov, registration number NCT00338962 and URL: http://clinicaltrials.gov/ct2/show/NCT00338962?term=desipramine+AND+alcohol+dependence+AND+depression&recr=Closed&rank=1. Desipramine 144-155 solute carrier family 6 member 10, pseudogene Homo sapiens 118-121 21976621-9 2012 The in vivo sensitivities of the clinically relevant CYP2D6 substrates desipramine, dextromethorphan, and metoprolol were determined on the basis of literature drug-drug interaction (DDI) outcomes. Desipramine 71-82 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 53-59 21976621-11 2012 Finally, the magnitude of in vivo CYP2D6 DDIs caused by quinidine was predicted using desipramine, dextromethorphan, and metoprolol. Desipramine 86-97 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 34-40 21918844-6 2012 The desipramine/cisplatin combination resulted in increased levels of p53 as well as mitochondrial damage, caspase activation, and poly(ADP ribose) polymerase cleavage, suggesting that desipramine may synergize with cisplatin more than with other platinum chemotherapeutics partly by activating distinct apoptotic pathways. Desipramine 4-15 tumor protein p53 Homo sapiens 70-73 21918844-6 2012 The desipramine/cisplatin combination resulted in increased levels of p53 as well as mitochondrial damage, caspase activation, and poly(ADP ribose) polymerase cleavage, suggesting that desipramine may synergize with cisplatin more than with other platinum chemotherapeutics partly by activating distinct apoptotic pathways. Desipramine 4-15 poly(ADP-ribose) polymerase 1 Homo sapiens 131-158 21918844-6 2012 The desipramine/cisplatin combination resulted in increased levels of p53 as well as mitochondrial damage, caspase activation, and poly(ADP ribose) polymerase cleavage, suggesting that desipramine may synergize with cisplatin more than with other platinum chemotherapeutics partly by activating distinct apoptotic pathways. Desipramine 185-196 tumor protein p53 Homo sapiens 70-73 21918844-6 2012 The desipramine/cisplatin combination resulted in increased levels of p53 as well as mitochondrial damage, caspase activation, and poly(ADP ribose) polymerase cleavage, suggesting that desipramine may synergize with cisplatin more than with other platinum chemotherapeutics partly by activating distinct apoptotic pathways. Desipramine 185-196 poly(ADP-ribose) polymerase 1 Homo sapiens 131-158 21867719-12 2012 and desipramine (16 mg/kg, i.p) prevented the depressant-like effect induced by TNF-alpha (0.1 fg/site, i.c.v.) Desipramine 4-15 tumor necrosis factor Mus musculus 80-89 22197911-8 2012 Our data show that fluoxetine is a selective inhibitor of GluN2B-containing NMDARs, whereas desipramine inhibits both GluN1/GluN2A and GluN1/GluN2B subtypes. Desipramine 92-103 glutamate ionotropic receptor NMDA type subunit 1 Homo sapiens 118-123 22197911-8 2012 Our data show that fluoxetine is a selective inhibitor of GluN2B-containing NMDARs, whereas desipramine inhibits both GluN1/GluN2A and GluN1/GluN2B subtypes. Desipramine 92-103 glutamate ionotropic receptor NMDA type subunit 2A Homo sapiens 124-130 22197911-8 2012 Our data show that fluoxetine is a selective inhibitor of GluN2B-containing NMDARs, whereas desipramine inhibits both GluN1/GluN2A and GluN1/GluN2B subtypes. Desipramine 92-103 glutamate ionotropic receptor NMDA type subunit 1 Homo sapiens 135-140 22197911-8 2012 Our data show that fluoxetine is a selective inhibitor of GluN2B-containing NMDARs, whereas desipramine inhibits both GluN1/GluN2A and GluN1/GluN2B subtypes. Desipramine 92-103 glutamate ionotropic receptor NMDA type subunit 2B Homo sapiens 141-147 21963947-0 2011 Regulation of CCL2/MCP-1 production in astrocytes by desipramine and atomoxetine: involvement of alpha2 adrenergic receptors. Desipramine 53-64 C-C motif chemokine ligand 2 Rattus norvegicus 14-18 22814024-10 2012 Desipramine decreased also relative weight of inguinal lymph nodes and spleens whereas desipramine and fluoxetine increased production of IL-10 in comparison to positive control. Desipramine 87-98 interleukin 10 Homo sapiens 138-143 23209658-0 2012 Desipramine protects neuronal cell death and induces heme oxygenase-1 expression in Mes23.5 dopaminergic neurons. Desipramine 0-11 heme oxygenase 1 Homo sapiens 53-69 23209658-6 2012 Desipramine induces endogenous anti-oxidative enzyme, heme oxygenase-1 (HO-1) protein and mRNA expression in concentration- and time-dependent manners. Desipramine 0-11 heme oxygenase 1 Homo sapiens 54-70 23209658-6 2012 Desipramine induces endogenous anti-oxidative enzyme, heme oxygenase-1 (HO-1) protein and mRNA expression in concentration- and time-dependent manners. Desipramine 0-11 heme oxygenase 1 Homo sapiens 72-76 23209658-7 2012 A different type of antidepressant SSRI (selective serotonin reuptake inhibitor), fluoxetine also shows similar effects of desipramine on HO-1 expression. Desipramine 123-134 heme oxygenase 1 Homo sapiens 138-142 23209658-8 2012 Moreover, desipramine induces HO-1 expression through activation of ERK and JNK signaling pathways. Desipramine 10-21 heme oxygenase 1 Homo sapiens 30-34 23209658-8 2012 Moreover, desipramine induces HO-1 expression through activation of ERK and JNK signaling pathways. Desipramine 10-21 mitogen-activated protein kinase 1 Homo sapiens 68-71 23209658-8 2012 Moreover, desipramine induces HO-1 expression through activation of ERK and JNK signaling pathways. Desipramine 10-21 mitogen-activated protein kinase 8 Homo sapiens 76-79 23209658-9 2012 Desipramine also increases NF-E2-related factor-2 (Nrf2) accumulation in the nucleus and enhances Nrf2-DNA binding activity. Desipramine 0-11 NFE2 like bZIP transcription factor 2 Homo sapiens 27-49 23209658-9 2012 Desipramine also increases NF-E2-related factor-2 (Nrf2) accumulation in the nucleus and enhances Nrf2-DNA binding activity. Desipramine 0-11 NFE2 like bZIP transcription factor 2 Homo sapiens 51-55 23209658-9 2012 Desipramine also increases NF-E2-related factor-2 (Nrf2) accumulation in the nucleus and enhances Nrf2-DNA binding activity. Desipramine 0-11 NFE2 like bZIP transcription factor 2 Homo sapiens 98-102 23209658-10 2012 Moreover, desipramine-mediated increase of HO-1 expression is reduced by transfection with siRNA against Nrf2. Desipramine 10-21 heme oxygenase 1 Homo sapiens 43-47 23209658-10 2012 Moreover, desipramine-mediated increase of HO-1 expression is reduced by transfection with siRNA against Nrf2. Desipramine 10-21 NFE2 like bZIP transcription factor 2 Homo sapiens 105-109 23209658-12 2012 Furthermore, inhibition of HO-1 activity by a HO-1 pharmacological inhibitor, ZnPP IX, attenuates the neuroprotective effect of desipramine. Desipramine 128-139 heme oxygenase 1 Homo sapiens 27-31 23209658-12 2012 Furthermore, inhibition of HO-1 activity by a HO-1 pharmacological inhibitor, ZnPP IX, attenuates the neuroprotective effect of desipramine. Desipramine 128-139 heme oxygenase 1 Homo sapiens 46-50 23209658-14 2012 CONCLUSIONS/SIGNIFICANCE: These findings suggest that desipramine-increased HO-1 expression is mediated by Nrf2 activation through the ERK and JNK signaling pathways. Desipramine 54-65 heme oxygenase 1 Homo sapiens 76-80 23209658-14 2012 CONCLUSIONS/SIGNIFICANCE: These findings suggest that desipramine-increased HO-1 expression is mediated by Nrf2 activation through the ERK and JNK signaling pathways. Desipramine 54-65 NFE2 like bZIP transcription factor 2 Homo sapiens 107-111 23209658-14 2012 CONCLUSIONS/SIGNIFICANCE: These findings suggest that desipramine-increased HO-1 expression is mediated by Nrf2 activation through the ERK and JNK signaling pathways. Desipramine 54-65 mitogen-activated protein kinase 1 Homo sapiens 135-138 23209658-14 2012 CONCLUSIONS/SIGNIFICANCE: These findings suggest that desipramine-increased HO-1 expression is mediated by Nrf2 activation through the ERK and JNK signaling pathways. Desipramine 54-65 mitogen-activated protein kinase 8 Homo sapiens 143-146 22563449-0 2012 Desipramine inhibits histamine H1 receptor-induced Ca2+ signaling in rat hypothalamic cells. Desipramine 0-11 histamine receptor H 1 Rattus norvegicus 21-42 22563449-4 2012 In primary cultured rat hypothalamic cells, desipramine markedly suppressed the elevation of intracellular Ca(2+) evoked by histamine H1 receptor activation. Desipramine 44-55 histamine receptor H 1 Rattus norvegicus 124-145 22563449-8 2012 Our results reveal that desipramine and sibutramine have different effects on histamine H1 receptor signaling in hypothalamic cells and suggest that distinct regulation of hypothalamic histamine signaling might underlie the differential regulation of food intake between antidepressants. Desipramine 24-35 histamine receptor H 1 Rattus norvegicus 78-99 21963947-0 2011 Regulation of CCL2/MCP-1 production in astrocytes by desipramine and atomoxetine: involvement of alpha2 adrenergic receptors. Desipramine 53-64 C-C motif chemokine ligand 2 Rattus norvegicus 19-24 21963947-2 2011 The treatment of primary rat astrocyte cultures with the noradrenaline transporter inhibitors desipramine or atomoxetine induced the expression and synthesis of CCL2/MCP-1 in these cells. Desipramine 94-105 C-C motif chemokine ligand 2 Rattus norvegicus 161-165 21963947-2 2011 The treatment of primary rat astrocyte cultures with the noradrenaline transporter inhibitors desipramine or atomoxetine induced the expression and synthesis of CCL2/MCP-1 in these cells. Desipramine 94-105 chemokine (C-C motif) ligand 2 Mus musculus 166-171 21859713-0 2011 The antidepressant desipramine is an arrestin-biased ligand at the alpha(2A)-adrenergic receptor driving receptor down-regulation in vitro and in vivo. Desipramine 19-30 adrenoceptor alpha 2A Homo sapiens 67-96 23049887-11 2012 Importantly, we have found that two drugs known to inhibit mammalian ASM activities, desipramine and clomipramine, markedly extend the lifespan of wild-type animals, in a manner similar to that achieved by genetic inactivation of the asm genes. Desipramine 85-96 sphingomyelin phosphodiesterase 1 Homo sapiens 69-72 23049887-11 2012 Importantly, we have found that two drugs known to inhibit mammalian ASM activities, desipramine and clomipramine, markedly extend the lifespan of wild-type animals, in a manner similar to that achieved by genetic inactivation of the asm genes. Desipramine 85-96 sphingomyelin phosphodiesterase 1 Homo sapiens 234-237 21859713-4 2011 In the present study, we have characterized the direct interaction between DMI and the alpha(2A)-adrenergic receptor (alpha(2A)AR), a key regulator of noradrenergic neurotransmission with altered expression and function in depression. Desipramine 75-78 adrenoceptor alpha 2A Homo sapiens 87-116 21859713-4 2011 In the present study, we have characterized the direct interaction between DMI and the alpha(2A)-adrenergic receptor (alpha(2A)AR), a key regulator of noradrenergic neurotransmission with altered expression and function in depression. Desipramine 75-78 adrenoceptor alpha 2A Homo sapiens 118-129 21859713-5 2011 DMI alone was found to be sufficient to drive receptor internalization acutely and a robust down-regulation of alpha(2A)AR expression and signaling following prolonged stimulation in vitro. Desipramine 0-3 adrenoceptor alpha 2A Homo sapiens 111-122 21859713-8 2011 Prolonged in vivo administration of DMI resulted in significant down-regulation of synaptic alpha(2A)AR expression, a response that was lost in arrestin3-null animals. Desipramine 36-39 adrenoceptor alpha 2A Homo sapiens 92-103 21442465-7 2011 The administration of desipramine, a specific inhibitor of noradrenaline reuptake, prevented the increase in Nur77-like immunoreactivity and mRNA induced by stress in rats subjected to repeated exposure to immobilization stress. Desipramine 22-33 nuclear receptor subfamily 4, group A, member 1 Rattus norvegicus 109-114 21208501-0 2011 Chronic desipramine treatment alters tyrosine hydroxylase but not norepinephrine transporter immunoreactivity in norepinephrine axons in the rat prefrontal cortex. Desipramine 8-19 tyrosine hydroxylase Rattus norvegicus 37-57 21208501-4 2011 Following DMI treatment (21 d, 15 mg/kg.d), NET-immunoreactive (ir) axons were significantly less likely to co-localize TH. Desipramine 10-13 solute carrier family 6 member 2 Rattus norvegicus 44-47 21481537-4 2011 Using an in situ brain/choroid plexus perfusion method, we tested the hypothesis that the antidepressant desipramine increases glucocorticoid accumulation in the mouse brain by inhibiting P-gp, following either chronic treatment (8 days, 20 mg/kg/day, IP) or acute administration (20 min brain perfusion in the presence of either 0.9 muM or 10 muM desipramine). Desipramine 105-116 phosphoglycolate phosphatase Mus musculus 188-192 21832094-3 2011 Importantly, desipramine belongs to a group of tricyclic antidepressant compounds that can simultaneously block hERG and inhibit its surface expression. Desipramine 13-24 ETS transcription factor ERG Homo sapiens 112-116 21832094-5 2011 Here, we have studied in detail how desipramine inhibits hERG surface expression. Desipramine 36-47 ETS transcription factor ERG Homo sapiens 57-61 21832094-6 2011 We find a previously unrecognized combination of two entirely different mechanisms; desipramine increases hERG endocytosis and degradation as a consequence of drug-induced channel ubiquitination and simultaneously inhibits hERG forward trafficking from the endoplasmic reticulum. Desipramine 84-95 ETS transcription factor ERG Homo sapiens 106-110 21832094-6 2011 We find a previously unrecognized combination of two entirely different mechanisms; desipramine increases hERG endocytosis and degradation as a consequence of drug-induced channel ubiquitination and simultaneously inhibits hERG forward trafficking from the endoplasmic reticulum. Desipramine 84-95 ETS transcription factor ERG Homo sapiens 223-227 21540358-5 2011 Moreover, the transfection of siRNA for nuclear factor erythroid 2-related factor 2 (Nrf2) to decrease the cellular expression level of Nrf2 exacerbated the cytotoxicity of some drugs (troglitazone, flutamide, acetaminophen, clozapine, terbinafine, and desipramine), suggesting that the genes regulated by Nrf2 are associated with the detoxification of the cytotoxicities mediated by CYP3A4. Desipramine 253-264 NFE2 like bZIP transcription factor 2 Homo sapiens 40-83 21605631-8 2011 Administration of desipramine (a norepinephrine reuptake inhibitor) to Sprague-Dawley rats was sufficient either alone or in combination with stress to increase IL-1beta mRNA in the hypothalamus and desipramine combined with stress was sufficient to increase IL-1R2 mRNA in the hypothalamus. Desipramine 18-29 interleukin 1 beta Rattus norvegicus 161-169 21605631-8 2011 Administration of desipramine (a norepinephrine reuptake inhibitor) to Sprague-Dawley rats was sufficient either alone or in combination with stress to increase IL-1beta mRNA in the hypothalamus and desipramine combined with stress was sufficient to increase IL-1R2 mRNA in the hypothalamus. Desipramine 18-29 interleukin 1 receptor type 2 Rattus norvegicus 259-265 21540358-5 2011 Moreover, the transfection of siRNA for nuclear factor erythroid 2-related factor 2 (Nrf2) to decrease the cellular expression level of Nrf2 exacerbated the cytotoxicity of some drugs (troglitazone, flutamide, acetaminophen, clozapine, terbinafine, and desipramine), suggesting that the genes regulated by Nrf2 are associated with the detoxification of the cytotoxicities mediated by CYP3A4. Desipramine 253-264 NFE2 like bZIP transcription factor 2 Homo sapiens 85-89 21540358-5 2011 Moreover, the transfection of siRNA for nuclear factor erythroid 2-related factor 2 (Nrf2) to decrease the cellular expression level of Nrf2 exacerbated the cytotoxicity of some drugs (troglitazone, flutamide, acetaminophen, clozapine, terbinafine, and desipramine), suggesting that the genes regulated by Nrf2 are associated with the detoxification of the cytotoxicities mediated by CYP3A4. Desipramine 253-264 NFE2 like bZIP transcription factor 2 Homo sapiens 136-140 21540358-5 2011 Moreover, the transfection of siRNA for nuclear factor erythroid 2-related factor 2 (Nrf2) to decrease the cellular expression level of Nrf2 exacerbated the cytotoxicity of some drugs (troglitazone, flutamide, acetaminophen, clozapine, terbinafine, and desipramine), suggesting that the genes regulated by Nrf2 are associated with the detoxification of the cytotoxicities mediated by CYP3A4. Desipramine 253-264 NFE2 like bZIP transcription factor 2 Homo sapiens 136-140 21389273-6 2011 Functionally, TNF-alpha-stimulated H(2)O(2) production was also blocked by MCD and DES (194.6 +- 15.4% vs. 90.6 +- 15.9% and 148.8 +- 20.4%), and the activation of the pivotal proinflammatory transcription factor, NF-kappaB, by TNF-alpha was reversed by MCD and DES as well as by small interfering RNAs of Duox1 or ASMase. Desipramine 83-86 tumor necrosis factor Homo sapiens 14-23 21621196-5 2011 We found that CUS led to depressive-like behavior and reduced the amplitude of PER2 oscillation in the SCN, which were blocked by 3 weeks of desipramine (DMI) treatment. Desipramine 141-152 period circadian regulator 2 Homo sapiens 79-83 21621196-5 2011 We found that CUS led to depressive-like behavior and reduced the amplitude of PER2 oscillation in the SCN, which were blocked by 3 weeks of desipramine (DMI) treatment. Desipramine 154-157 period circadian regulator 2 Homo sapiens 79-83 21389273-6 2011 Functionally, TNF-alpha-stimulated H(2)O(2) production was also blocked by MCD and DES (194.6 +- 15.4% vs. 90.6 +- 15.9% and 148.8 +- 20.4%), and the activation of the pivotal proinflammatory transcription factor, NF-kappaB, by TNF-alpha was reversed by MCD and DES as well as by small interfering RNAs of Duox1 or ASMase. Desipramine 83-86 nuclear factor kappa B subunit 1 Homo sapiens 214-223 21389273-6 2011 Functionally, TNF-alpha-stimulated H(2)O(2) production was also blocked by MCD and DES (194.6 +- 15.4% vs. 90.6 +- 15.9% and 148.8 +- 20.4%), and the activation of the pivotal proinflammatory transcription factor, NF-kappaB, by TNF-alpha was reversed by MCD and DES as well as by small interfering RNAs of Duox1 or ASMase. Desipramine 83-86 tumor necrosis factor Homo sapiens 228-237 21389273-6 2011 Functionally, TNF-alpha-stimulated H(2)O(2) production was also blocked by MCD and DES (194.6 +- 15.4% vs. 90.6 +- 15.9% and 148.8 +- 20.4%), and the activation of the pivotal proinflammatory transcription factor, NF-kappaB, by TNF-alpha was reversed by MCD and DES as well as by small interfering RNAs of Duox1 or ASMase. Desipramine 83-86 dual oxidase 1 Homo sapiens 306-311 21389273-6 2011 Functionally, TNF-alpha-stimulated H(2)O(2) production was also blocked by MCD and DES (194.6 +- 15.4% vs. 90.6 +- 15.9% and 148.8 +- 20.4%), and the activation of the pivotal proinflammatory transcription factor, NF-kappaB, by TNF-alpha was reversed by MCD and DES as well as by small interfering RNAs of Duox1 or ASMase. Desipramine 83-86 sphingomyelin phosphodiesterase 1 Homo sapiens 315-321 21248719-9 2011 This effect of DMI on spine density was severely attenuated following ATR deletion. Desipramine 15-18 ataxia telangiectasia and Rad3 related Mus musculus 70-73 21278119-12 2011 Because RFI is the error of the DMI equation, any inaccuracy when estimating intake will lead to a bias in the prediction of RFI. Desipramine 32-35 RFI Bos taurus 8-11 21278119-12 2011 Because RFI is the error of the DMI equation, any inaccuracy when estimating intake will lead to a bias in the prediction of RFI. Desipramine 32-35 RFI Bos taurus 125-128 21120454-0 2011 hERG K+ channel-associated cardiac effects of the antidepressant drug desipramine. Desipramine 70-81 ETS transcription factor ERG Homo sapiens 0-4 21052032-11 2011 CONCLUSION: The MC1R gene might be associated with major depressive disorder and with treatment response to desipramine. Desipramine 108-119 melanocortin 1 receptor Homo sapiens 16-20 21229608-5 2011 Moreover, desipramine attenuated THDF-mediated cell death, indicating a crucial role of ASMase in the mechanism of cell death. Desipramine 10-21 sphingomyelin phosphodiesterase 1 Homo sapiens 88-94 21130737-6 2011 Both Desipramine and FTY720 treatment reduced ASMase without significant inhibition of other lysosomal hydrolases but most hydrolases showed increased secretion (up to a 50% increase) providing more evidence of lysosomal disruption by these drugs. Desipramine 5-16 sphingomyelin phosphodiesterase 1 Homo sapiens 46-52 21178181-11 2011 Feeding fat and infusion of GLP-1 tended (linear, P = 0.12; quadratic, P = 0.13) to decrease DMI. Desipramine 93-96 glucagon like peptide 1 receptor Homo sapiens 28-33 20549303-0 2011 Desipramine induces apoptosis in rat glioma cells via endoplasmic reticulum stress-dependent CHOP pathway. Desipramine 0-11 DNA-damage inducible transcript 3 Rattus norvegicus 93-97 20549303-3 2011 DMI induced typical apoptotic morphology of chromatin condensation in rat glioma C6 cells and activated intracellular caspase 9 and caspase 3 with no change in mitochondrial membrane potential. Desipramine 0-3 caspase 9 Rattus norvegicus 118-127 20549303-3 2011 DMI induced typical apoptotic morphology of chromatin condensation in rat glioma C6 cells and activated intracellular caspase 9 and caspase 3 with no change in mitochondrial membrane potential. Desipramine 0-3 caspase 3 Rattus norvegicus 132-141 20549303-5 2011 However, knockdown of CHOP by CHOP-specific short interfering RNA (siRNA) could decrease the activity of intracellular caspase 3 and the cytotoxicity of DMI to C6 cells. Desipramine 153-156 DNA-damage inducible transcript 3 Rattus norvegicus 22-26 20549303-5 2011 However, knockdown of CHOP by CHOP-specific short interfering RNA (siRNA) could decrease the activity of intracellular caspase 3 and the cytotoxicity of DMI to C6 cells. Desipramine 153-156 DNA-damage inducible transcript 3 Rattus norvegicus 30-34 20549303-6 2011 These results revealed that the CHOP-dependent endoplasmic reticulum (ER) stress pathway is responsible for DMI-induced apoptosis in C6 cells. Desipramine 108-111 DNA-damage inducible transcript 3 Rattus norvegicus 32-36 22076148-6 2011 In vivo, chronic treatment with the non-selective antidepressant imipramine as well as the norepinephrine-selective reuptake inhibitor desipramine or the serotonin-selective reuptake inhibitor fluoxetine all decrease p21 expression, and this was associated with increased neurogenesis. Desipramine 135-146 cyclin-dependent kinase inhibitor 1A (P21) Mus musculus 217-220 21120454-8 2011 We found that desipramine reduced hERG currents by binding to a receptor site inside the channel pore. Desipramine 14-25 ETS transcription factor ERG Homo sapiens 34-38 21120454-12 2011 Finally, desipramine triggered apoptosis in cells expressing hERG channels. Desipramine 9-20 ETS transcription factor ERG Homo sapiens 61-65 21120454-13 2011 Desipramine exerts at least four different cellular effects: (1) direct hERG channel block, (2) acute reduction of hERG surface expression, (3) chronic disruption of hERG trafficking, and (4) induction of apoptosis. Desipramine 0-11 ETS transcription factor ERG Homo sapiens 72-76 21120454-13 2011 Desipramine exerts at least four different cellular effects: (1) direct hERG channel block, (2) acute reduction of hERG surface expression, (3) chronic disruption of hERG trafficking, and (4) induction of apoptosis. Desipramine 0-11 ETS transcription factor ERG Homo sapiens 115-119 21120454-13 2011 Desipramine exerts at least four different cellular effects: (1) direct hERG channel block, (2) acute reduction of hERG surface expression, (3) chronic disruption of hERG trafficking, and (4) induction of apoptosis. Desipramine 0-11 ETS transcription factor ERG Homo sapiens 115-119 20878594-7 2010 Evidence of cytochrome P450 (CYP)2D6 inhibition in clinical trials highlighted a concern for pharmacokinetic interaction with other drugs that are CYP2D6 substrates, as confirmed by a rise in the pharmacokinetic parameters of desipramine with concomitant administration of mirabegron. Desipramine 226-237 cytochrome P450, family 2, subfamily d, polypeptide 4 Rattus norvegicus 12-36 20926613-7 2010 In HEPES buffer the pH-sensitive current expressed by RTN astrocytes reversed near E(K(+)) (the equilibrium potential for K(+)) and was inhibited by Ba(2+) and desipramine (blocker of Kir4.1-containing channels), characteristics most consistent with heteromeric Kir4.1-Kir5.1 channels. Desipramine 160-171 potassium inwardly rectifying channel subfamily J member 10 Homo sapiens 184-190 20926613-7 2010 In HEPES buffer the pH-sensitive current expressed by RTN astrocytes reversed near E(K(+)) (the equilibrium potential for K(+)) and was inhibited by Ba(2+) and desipramine (blocker of Kir4.1-containing channels), characteristics most consistent with heteromeric Kir4.1-Kir5.1 channels. Desipramine 160-171 potassium inwardly rectifying channel subfamily J member 10 Homo sapiens 262-268 20974006-5 2010 Functional knockdown of ASMase with either the chemical inhibitor, desipramine, or siRNA markedly reduces alpha-TEA-induced cell surface membrane accumulation of ceramide and its co-localization with Fas, DR5, and FADD, cleavage of caspases-8 and -9 and apoptosis, suggesting an early and critical role for ASMase in alpha-TEA-induced apoptosis. Desipramine 67-78 sphingomyelin phosphodiesterase 1 Homo sapiens 24-30 20840444-0 2010 Desipramine, substrate for CYP2D6 activity: population pharmacokinetic model and design elements of drug-drug interaction trials. Desipramine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 27-33 20840444-2 2010 Additional objectives were to develop a semi-mechanistic population pharmacokinetic model for desipramine, which allowed simulation of CYP2D6-mediated inhibition, when using desipramine as a probe substrate, and to evaluate certain study design elements, such as duration of desipramine pharmacokinetic sampling, required sample size and optimal pharmacokinetic sampling schedule for intermediate, extensive and ultrarapid metabolizers of CYP2D6 substrates. Desipramine 94-105 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 135-141 20840444-2 2010 Additional objectives were to develop a semi-mechanistic population pharmacokinetic model for desipramine, which allowed simulation of CYP2D6-mediated inhibition, when using desipramine as a probe substrate, and to evaluate certain study design elements, such as duration of desipramine pharmacokinetic sampling, required sample size and optimal pharmacokinetic sampling schedule for intermediate, extensive and ultrarapid metabolizers of CYP2D6 substrates. Desipramine 94-105 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 439-445 20878594-7 2010 Evidence of cytochrome P450 (CYP)2D6 inhibition in clinical trials highlighted a concern for pharmacokinetic interaction with other drugs that are CYP2D6 substrates, as confirmed by a rise in the pharmacokinetic parameters of desipramine with concomitant administration of mirabegron. Desipramine 226-237 cytochrome P450, family 2, subfamily d, polypeptide 4 Rattus norvegicus 147-153 20435089-4 2010 We have found that desipramine (DMI), a tricyclic antidepressant, acts as an inhibitor of thyroid hormone transport by MCT8. Desipramine 19-30 solute carrier family 16 member 2 Homo sapiens 119-123 20435089-4 2010 We have found that desipramine (DMI), a tricyclic antidepressant, acts as an inhibitor of thyroid hormone transport by MCT8. Desipramine 32-35 solute carrier family 16 member 2 Homo sapiens 119-123 20435089-7 2010 In a heterologous expression system, both human MCT8 and its close homolog, MCT10, were sensitive to inhibition by DMI. Desipramine 115-118 solute carrier family 16 member 2 Homo sapiens 48-52 20435089-7 2010 In a heterologous expression system, both human MCT8 and its close homolog, MCT10, were sensitive to inhibition by DMI. Desipramine 115-118 solute carrier family 16 member 10 Homo sapiens 76-81 19749024-3 2010 Feed conversion ratio (FCR) is the inverse of gross feed efficiency and is the ratio of DMI to ADG. Desipramine 88-91 FCR Bos taurus 0-21 20136837-4 2010 KEY RESULTS: NTUA fluorescence accumulated linearly in nerve terminals, an effect that was prevented with NET inhibition with desipramine (1 microM). Desipramine 126-137 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 106-109 20158296-7 2010 Chronic administration of the antidepressant drugs fluoxetine and desipramine produced robust elevations in hippocampal cell proliferation and brain-derived neurotrophic factor (BDNF) protein levels in MRL/MpJ mice. Desipramine 66-77 brain derived neurotrophic factor Mus musculus 143-176 20158296-7 2010 Chronic administration of the antidepressant drugs fluoxetine and desipramine produced robust elevations in hippocampal cell proliferation and brain-derived neurotrophic factor (BDNF) protein levels in MRL/MpJ mice. Desipramine 66-77 brain derived neurotrophic factor Mus musculus 178-182 21364631-3 2010 Inhibition of JNK activation with sphingomyelinase inhibitors (20 muM desipramine, 20 muM imipramine), with the protein kinase C-delta (PKCdelta) inhibitor rottlerin (10 muM), and with the specific PKCtheta pseudosubstrate inhibitor (30 muM) indicates that ceramide and phosphorylation by PKCdelta and PKCtheta mediate gal-1-induced JNK activation. Desipramine 70-81 mitogen-activated protein kinase 8 Homo sapiens 14-17 19635928-9 2010 Inhalation of the Asm inhibitors amitriptyline, trimipramine, desipramine, chlorprothixene, fluoxetine, amlodipine, or sertraline restored normal ceramide concentrations in murine bronchial epithelial cells, reduced inflammation in the lung of CF mice and prevented infection with Pseudomonas aeruginosa. Desipramine 62-73 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 18-21 20428789-1 2010 Desipramine (DMI) has been reported to induce glucocorticoid receptor-mediated signal transduction in recent studies. Desipramine 0-11 nuclear receptor subfamily 3, group C, member 1 Mus musculus 46-69 20428789-1 2010 Desipramine (DMI) has been reported to induce glucocorticoid receptor-mediated signal transduction in recent studies. Desipramine 13-16 nuclear receptor subfamily 3, group C, member 1 Mus musculus 46-69 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 125-128 B cell leukemia/lymphoma 2 Mus musculus 248-253 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 125-128 baculoviral IAP repeat-containing 5 Mus musculus 258-266 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 125-128 BCL2-like 1 Mus musculus 275-281 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 125-128 BCL2-associated X protein Mus musculus 335-338 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 125-128 apoptotic peptidase activating factor 1 Mus musculus 340-346 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 125-128 caspase 3 Mus musculus 348-357 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 125-128 caspase 7 Mus musculus 362-371 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 223-226 B cell leukemia/lymphoma 2 Mus musculus 248-253 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 223-226 baculoviral IAP repeat-containing 5 Mus musculus 258-266 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 223-226 BCL2-like 1 Mus musculus 275-281 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 223-226 BCL2-associated X protein Mus musculus 335-338 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 223-226 apoptotic peptidase activating factor 1 Mus musculus 340-346 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 223-226 caspase 3 Mus musculus 348-357 20428789-5 2010 Apoptotic bodies in the nuclei of cells and DNA fragmentation were observed when the Ca3/7 cells were treated with 20 microM DMI for 24 h. Quantitative RT-PCR (reverse transcriptional-polymerase chain reaction) showed that DMI caused a decrease in Bcl-2 and survivin but not Bcl-xL gene expression and an increase in the expression of Bax, Apaf-1, caspase-3 and caspase-7 in a dose- and time-dependent manner. Desipramine 223-226 caspase 7 Mus musculus 362-371 20428789-7 2010 Quantitative RT-PCR revealed that DMI decreased the expression of the PCNA gene and caused an increase in the expression of the p21 and p27 genes in the Ca3/7 cells. Desipramine 34-37 proliferating cell nuclear antigen Mus musculus 70-74 20428789-7 2010 Quantitative RT-PCR revealed that DMI decreased the expression of the PCNA gene and caused an increase in the expression of the p21 and p27 genes in the Ca3/7 cells. Desipramine 34-37 cyclin-dependent kinase inhibitor 1A (P21) Mus musculus 128-131 20428789-7 2010 Quantitative RT-PCR revealed that DMI decreased the expression of the PCNA gene and caused an increase in the expression of the p21 and p27 genes in the Ca3/7 cells. Desipramine 34-37 cyclin-dependent kinase inhibitor 1B Mus musculus 136-139 19884907-1 2010 CYP2C19 converts the tricyclic antidepressant imipramine to its active metabolite desipramine, which is subsequently inactivated by CYP2D6. Desipramine 82-93 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 0-7 19884907-1 2010 CYP2C19 converts the tricyclic antidepressant imipramine to its active metabolite desipramine, which is subsequently inactivated by CYP2D6. Desipramine 82-93 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 132-138 19884907-6 2010 In a multivariate analysis, we found a significant, but limited effect (P=0.035, eta(2)=0.031) of the CYP2C19*17 genotype on imipramine+desipramine concentrations. Desipramine 136-147 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 102-109 20211602-0 2010 Block of the human ether-a-go-go-related gene (hERG) K+ channel by the antidepressant desipramine. Desipramine 86-97 ETS transcription factor ERG Homo sapiens 47-51 20211602-2 2010 Since blockade of cardiac human ether-a-go-go-related gene (hERG) channels is an important cause of acquired long QT syndrome, we investigated the acute effects of desipramine on hERG channels to determine the electrophysiological basis for its pro-arrhythmic potential. Desipramine 164-175 ETS transcription factor ERG Homo sapiens 179-183 20211602-3 2010 We examined the effects of desipramine on the hERG channels expressed in Xenopus oocytes using two-microelectrode voltage-clamp techniques. Desipramine 27-38 ETS transcription factor ERG Homo sapiens 46-50 20211602-4 2010 Desipramine-induced concentration-dependent decreases in the current amplitude at the end of the voltage steps and hERG tail currents. Desipramine 0-11 ETS transcription factor ERG Homo sapiens 115-119 20211602-5 2010 The IC(50) for desipramine needed to block the hERG current in Xenopus oocytes decreased progressively relative to the degree of depolarization. Desipramine 15-26 ETS transcription factor ERG Homo sapiens 47-51 20211602-7 2010 The S6 domain mutations, Tyr-652 located in the S6 domain of the hERG channel reduced the potency of the channel block by desipramine more than a mutation of Phe-656 in the same region. Desipramine 122-133 ETS transcription factor ERG Homo sapiens 65-69 20211602-8 2010 These results suggest that desipramine is a blocker of the hERG channels, providing a molecular mechanism for the arrhythmogenic side effects during the clinical administration of desipramine. Desipramine 27-38 ETS transcription factor ERG Homo sapiens 59-63 20211602-8 2010 These results suggest that desipramine is a blocker of the hERG channels, providing a molecular mechanism for the arrhythmogenic side effects during the clinical administration of desipramine. Desipramine 180-191 ETS transcription factor ERG Homo sapiens 59-63 20132473-8 2010 Desipramine (an inhibitor of NA reuptake) also caused an increase of MCP-1 in cortex. Desipramine 0-11 C-C motif chemokine ligand 2 Rattus norvegicus 69-74 19749024-3 2010 Feed conversion ratio (FCR) is the inverse of gross feed efficiency and is the ratio of DMI to ADG. Desipramine 88-91 FCR Bos taurus 23-26 20722995-1 2010 OBJECTIVES: In rats with diabetes mellitus induced by alloxan (DMIA) or streptozocin (DMIS), changes in the cytochrome P450 (CYP) isozymes in the liver, lung, kidney, intestine, brain, and testis have been reported based on Western blot analysis, Northern blot analysis, and various enzyme activities. Desipramine 86-90 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 108-123 20722995-1 2010 OBJECTIVES: In rats with diabetes mellitus induced by alloxan (DMIA) or streptozocin (DMIS), changes in the cytochrome P450 (CYP) isozymes in the liver, lung, kidney, intestine, brain, and testis have been reported based on Western blot analysis, Northern blot analysis, and various enzyme activities. Desipramine 86-90 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 125-128 19995332-0 2009 Difference in desipramine metabolic profile between wild-type and CYP2D6-humanized mice. Desipramine 14-25 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 66-72 21200377-7 2010 MAO-A was inhibited by the following drugs: pargyline > clorgyline > iproniazid > fluoxetine > desipramine > amitriptyline > imipramine > citalopram > venlafaxine > reboxetine > olanzapine > mirtazapine > tianeptine > moclobemide, cocaine >> lithium, valproate. Desipramine 107-118 monoamine oxidase A Sus scrofa 0-5 21200377-8 2010 MAO-B was inhibited by the following drugs: pargyline > clorgyline > iproniazid > fluoxetine > venlafaxine > amitriptyline > olanzapine > citalopram > desipramine > reboxetine > imipramine > tianeptine > mirtazapine, cocaine >> moclobemide, lithium, valproate. Desipramine 175-186 monoamine oxidase B Sus scrofa 0-5 19818834-0 2009 Desipramine induced changes in the norepinephrine transporter, alpha- and gamma-synuclein in the hippocampus, amygdala and striatum. Desipramine 0-11 solute carrier family 6 member 2 Rattus norvegicus 35-61 19818834-0 2009 Desipramine induced changes in the norepinephrine transporter, alpha- and gamma-synuclein in the hippocampus, amygdala and striatum. Desipramine 0-11 synuclein alpha Rattus norvegicus 63-89 19995332-1 2009 Desipramine (DMI), a CYP2D6 probe, was used as a model drug to test whether CYP2D6-humanized (Tg-CYP2D6) and wild-type control mice could be used as preclinical animal models to identify the effects of CYP2D6 genotype/phenotype on drug metabolic profiles. Desipramine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 21-27 19995332-4 2009 Three metabolites, 2-hydroxyl-, 10-hydroxyl, and N-desmethyl-desipramine were identified in the incubations of DMI with both wild-type and Tg-CYP2D6 MLM, as well as in human CYP2D6 extensive metabolizer liver microsomes. Desipramine 111-114 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 142-148 19995332-4 2009 Three metabolites, 2-hydroxyl-, 10-hydroxyl, and N-desmethyl-desipramine were identified in the incubations of DMI with both wild-type and Tg-CYP2D6 MLM, as well as in human CYP2D6 extensive metabolizer liver microsomes. Desipramine 111-114 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 174-180 19575421-0 2009 Desipramine inhibits the growth of a mouse skin squamous cell carcinoma cell line and affects glucocorticoid receptor-mediated transcription. Desipramine 0-11 nuclear receptor subfamily 3, group C, member 1 Mus musculus 94-117 19575421-1 2009 The purpose of this study was to examine the effect of tricyclic antidepressant desipramine (DMI) on the growth inhibition and translocation of the glucocorticoid receptor (GR) from the cytoplasm to the nucleus in cancerous and noncancerous cell lines and the effect of DMI on GR-mediated transcription. Desipramine 80-91 nuclear receptor subfamily 3, group C, member 1 Mus musculus 148-171 19575421-1 2009 The purpose of this study was to examine the effect of tricyclic antidepressant desipramine (DMI) on the growth inhibition and translocation of the glucocorticoid receptor (GR) from the cytoplasm to the nucleus in cancerous and noncancerous cell lines and the effect of DMI on GR-mediated transcription. Desipramine 80-91 nuclear receptor subfamily 3, group C, member 1 Mus musculus 173-175 19575421-1 2009 The purpose of this study was to examine the effect of tricyclic antidepressant desipramine (DMI) on the growth inhibition and translocation of the glucocorticoid receptor (GR) from the cytoplasm to the nucleus in cancerous and noncancerous cell lines and the effect of DMI on GR-mediated transcription. Desipramine 93-96 nuclear receptor subfamily 3, group C, member 1 Mus musculus 148-171 19575421-1 2009 The purpose of this study was to examine the effect of tricyclic antidepressant desipramine (DMI) on the growth inhibition and translocation of the glucocorticoid receptor (GR) from the cytoplasm to the nucleus in cancerous and noncancerous cell lines and the effect of DMI on GR-mediated transcription. Desipramine 93-96 nuclear receptor subfamily 3, group C, member 1 Mus musculus 173-175 19575421-1 2009 The purpose of this study was to examine the effect of tricyclic antidepressant desipramine (DMI) on the growth inhibition and translocation of the glucocorticoid receptor (GR) from the cytoplasm to the nucleus in cancerous and noncancerous cell lines and the effect of DMI on GR-mediated transcription. Desipramine 93-96 nuclear receptor subfamily 3, group C, member 1 Mus musculus 277-279 19575421-1 2009 The purpose of this study was to examine the effect of tricyclic antidepressant desipramine (DMI) on the growth inhibition and translocation of the glucocorticoid receptor (GR) from the cytoplasm to the nucleus in cancerous and noncancerous cell lines and the effect of DMI on GR-mediated transcription. Desipramine 270-273 nuclear receptor subfamily 3, group C, member 1 Mus musculus 148-171 19575421-1 2009 The purpose of this study was to examine the effect of tricyclic antidepressant desipramine (DMI) on the growth inhibition and translocation of the glucocorticoid receptor (GR) from the cytoplasm to the nucleus in cancerous and noncancerous cell lines and the effect of DMI on GR-mediated transcription. Desipramine 270-273 nuclear receptor subfamily 3, group C, member 1 Mus musculus 173-175 19575421-6 2009 The translocation of GR was induced by FA alone, DMI alone, and combination of both agents. Desipramine 49-52 nuclear receptor subfamily 3, group C, member 1 Mus musculus 21-23 19575421-8 2009 However, DMI inhibited FA-induced, GR-mediated transcription when both agents were given together. Desipramine 9-12 nuclear receptor subfamily 3, group C, member 1 Mus musculus 35-37 19575421-9 2009 Pretreatment with DMI followed by combination of DMI and FA decreased GR-mediated transcription more than pretreatment with FA. Desipramine 18-21 nuclear receptor subfamily 3, group C, member 1 Mus musculus 70-72 19575421-9 2009 Pretreatment with DMI followed by combination of DMI and FA decreased GR-mediated transcription more than pretreatment with FA. Desipramine 49-52 nuclear receptor subfamily 3, group C, member 1 Mus musculus 70-72 19575421-10 2009 The expression of metallothionein-1 (Mt-1) gene, which is regulated by GR, was induced significantly by the combination of DMI and FA, and enhanced significantly by pretreatment with FA but not DMI. Desipramine 123-126 metallothionein 1 Mus musculus 18-35 19575421-10 2009 The expression of metallothionein-1 (Mt-1) gene, which is regulated by GR, was induced significantly by the combination of DMI and FA, and enhanced significantly by pretreatment with FA but not DMI. Desipramine 123-126 metallothionein 1 Mus musculus 37-41 19575421-10 2009 The expression of metallothionein-1 (Mt-1) gene, which is regulated by GR, was induced significantly by the combination of DMI and FA, and enhanced significantly by pretreatment with FA but not DMI. Desipramine 123-126 nuclear receptor subfamily 3, group C, member 1 Mus musculus 71-73 19575421-10 2009 The expression of metallothionein-1 (Mt-1) gene, which is regulated by GR, was induced significantly by the combination of DMI and FA, and enhanced significantly by pretreatment with FA but not DMI. Desipramine 194-197 metallothionein 1 Mus musculus 18-35 19575421-10 2009 The expression of metallothionein-1 (Mt-1) gene, which is regulated by GR, was induced significantly by the combination of DMI and FA, and enhanced significantly by pretreatment with FA but not DMI. Desipramine 194-197 metallothionein 1 Mus musculus 37-41 19575421-11 2009 DMI is suggested to inhibit the growth of Ca3/7 cells and to affect GR-mediated transcription. Desipramine 0-3 nuclear receptor subfamily 3, group C, member 1 Mus musculus 68-70 19420132-6 2009 The K(m) values for N-sulfation mediated by hSULT2A1 were as follows: 1.08 +/- 0.03 mM for ciprofloxacin, 0.53 +/- 0.01 mM for moxifloxacin, 0.19 +/- 0.01 mM for garenoxacin, 0.054 +/- 0.001 mM for desipramine, and 2.32 +/- 0.12 mM for metoclopramide. Desipramine 198-209 sulfotransferase family 2A member 1 Homo sapiens 44-52 20081247-10 2009 The prometastatic effect of desipramine in young animals was connected with an increase of VEGF and MMP-9 plasma levels. Desipramine 28-39 vascular endothelial growth factor A Mus musculus 91-95 20081247-10 2009 The prometastatic effect of desipramine in young animals was connected with an increase of VEGF and MMP-9 plasma levels. Desipramine 28-39 matrix metallopeptidase 9 Mus musculus 100-105 19015855-0 2009 Disposition of desipramine, a sensitive cytochrome P450 2D6 substrate, when coadministered with intravenous temsirolimus. Desipramine 15-26 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 40-59 19328214-4 2009 HTC cells were found to be sensitive to exogenous ceramide and significantly protected against TNF by desipramine, an inhibitor of lysosomal acid sphingomyelinase. Desipramine 102-113 tumor necrosis factor Rattus norvegicus 95-98 19015855-4 2009 METHODS: This 2-period study in healthy subjects investigated the pharmacokinetics of a single oral 50-mg dose of the CYP2D6 substrate desipramine, first without and subsequently with a single coadministered i.v. Desipramine 135-146 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 118-124 19177066-10 2009 Protein levels of brain-derived neurotrophic factor (BDNF) in MRL/MpJ mice were elevated significantly in the frontal cortex, hippocampus, and amygdala after chronic FLX treatment, but increased only in the frontal cortex by chronic DMI. Desipramine 233-236 brain derived neurotrophic factor Mus musculus 18-51 19457081-2 2009 We report here that PACAP-deficient (PACAP-/-) mice showed increased immobility in a forced swimming test, which was reduced by the antidepressant desipramine, to a similar extent as in wild-type mice. Desipramine 147-158 adenylate cyclase activating polypeptide 1 Mus musculus 20-25 19457081-2 2009 We report here that PACAP-deficient (PACAP-/-) mice showed increased immobility in a forced swimming test, which was reduced by the antidepressant desipramine, to a similar extent as in wild-type mice. Desipramine 147-158 adenylate cyclase activating polypeptide 1 Mus musculus 37-42 19177066-10 2009 Protein levels of brain-derived neurotrophic factor (BDNF) in MRL/MpJ mice were elevated significantly in the frontal cortex, hippocampus, and amygdala after chronic FLX treatment, but increased only in the frontal cortex by chronic DMI. Desipramine 233-236 brain derived neurotrophic factor Mus musculus 53-57 19177066-11 2009 In contrast, BDNF levels in C57BL/6J mice were decreased in the hippocampus and increased in the amygdala after chronic FLX, and were decreased in the brain stem after chronic DMI. Desipramine 176-179 brain derived neurotrophic factor Mus musculus 13-17 18923403-1 2009 The antidepressant desipramine inhibits the reuptake of norepinephrine (NE), leading to activation of both pre- and postsynaptic adrenergic receptors, including alpha-1, alpha-2, beta-1, and beta-2 subtypes. Desipramine 19-30 hemoglobin, beta adult major chain Mus musculus 179-185 18923402-1 2009 Previous work has shown that repeated desipramine treatment causes downregulation of the norepinephrine transporter (NET) and persistent antidepressant-like effects on behavior, ie effects observed 2 days after discontinuation of drug treatment when acute effects are minimized. Desipramine 38-49 solute carrier family 6 member 2 Rattus norvegicus 89-115 18986328-0 2009 Desipramine-induced Ca-independent apoptosis in Mg63 human osteosarcoma cells: dependence on P38 mitogen-activated protein kinase-regulated activation of caspase 3. Desipramine 0-11 mitogen-activated protein kinase 14 Homo sapiens 93-129 18986328-0 2009 Desipramine-induced Ca-independent apoptosis in Mg63 human osteosarcoma cells: dependence on P38 mitogen-activated protein kinase-regulated activation of caspase 3. Desipramine 0-11 caspase 3 Homo sapiens 154-163 18986328-6 2009 Cells treated with 100-800 mmol/L desipramine showed typical apoptotic features, including an increase in sub-diploid nuclei and activation of caspase 3, indicating that these cells underwent apoptosis. Desipramine 34-45 caspase 3 Homo sapiens 143-152 18986328-7 2009 Immunoblotting revealed that 100 mmol/L desipramine activated extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 40-51 mitogen-activated protein kinase 1 Homo sapiens 62-99 18986328-7 2009 Immunoblotting revealed that 100 mmol/L desipramine activated extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 40-51 mitogen-activated protein kinase 1 Homo sapiens 101-104 18986328-7 2009 Immunoblotting revealed that 100 mmol/L desipramine activated extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 40-51 mitogen-activated protein kinase 8 Homo sapiens 107-130 18986328-7 2009 Immunoblotting revealed that 100 mmol/L desipramine activated extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 40-51 mitogen-activated protein kinase 8 Homo sapiens 132-135 18986328-7 2009 Immunoblotting revealed that 100 mmol/L desipramine activated extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 40-51 mitogen-activated protein kinase 14 Homo sapiens 141-177 18986328-7 2009 Immunoblotting revealed that 100 mmol/L desipramine activated extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 40-51 mitogen-activated protein kinase 1 Homo sapiens 179-183 18986328-9 2009 Desipramine-induced caspase 3 activation required p38 MAPK activation. Desipramine 0-11 caspase 3 Homo sapiens 20-29 18986328-9 2009 Desipramine-induced caspase 3 activation required p38 MAPK activation. Desipramine 0-11 mitogen-activated protein kinase 14 Homo sapiens 50-53 18986328-9 2009 Desipramine-induced caspase 3 activation required p38 MAPK activation. Desipramine 0-11 mitogen-activated protein kinase 1 Homo sapiens 54-58 18986328-13 2009 The results of the present study suggest that, in MG63 human osteosarcoma cells, desipramine causes Ca(2+)-independent apoptosis by inducing p38 MAPK-associated activation of caspase 3. Desipramine 81-92 mitogen-activated protein kinase 14 Homo sapiens 141-144 18986328-13 2009 The results of the present study suggest that, in MG63 human osteosarcoma cells, desipramine causes Ca(2+)-independent apoptosis by inducing p38 MAPK-associated activation of caspase 3. Desipramine 81-92 caspase 3 Homo sapiens 175-184 18952723-6 2009 Relationships between ADG and G:F with DMI as a percentage of BW and NE(g) intake also were examined in the same data set. Desipramine 39-42 ADG Bos taurus 22-25 18952723-7 2009 Across the wide range of average shrunk BW in the database (334.4 to 548.0 kg), ADG was positively related to DMI as a percentage of BW (P < 0.01); however, this relationship was not strong (r(2) = 0.17). Desipramine 110-113 ADG Bos taurus 80-83 19106229-0 2009 Desipramine reduces stress-activated dynorphin expression and CREB phosphorylation in NAc tissue. Desipramine 0-11 cAMP responsive element binding protein 1 Rattus norvegicus 62-66 19106229-7 2009 In primary cultures of NAc and striatum, DMI decreases basal and stimulated CREB phosphorylation by causing reductions in intracellular calcium (Ca(2+)) availability that are independent of norepinephrine or other monoaminergic inputs, identifying a potential mechanism for alterations in CREB-mediated gene expression. Desipramine 41-44 cAMP responsive element binding protein 1 Rattus norvegicus 76-80 19106229-7 2009 In primary cultures of NAc and striatum, DMI decreases basal and stimulated CREB phosphorylation by causing reductions in intracellular calcium (Ca(2+)) availability that are independent of norepinephrine or other monoaminergic inputs, identifying a potential mechanism for alterations in CREB-mediated gene expression. Desipramine 41-44 cAMP responsive element binding protein 1 Rattus norvegicus 289-293 19106229-9 2009 These findings raise the possibility that a therapeutically relevant mechanism of action of DMI occurs through attenuation of CREB-mediated gene transcription, which is mediated via previously uncharacterized mechanisms that occur directly within the NAc. Desipramine 92-95 cAMP responsive element binding protein 1 Rattus norvegicus 126-130 18800063-7 2009 DMI administration increased sst(1/4) receptors levels in the CA1 hippocampal region. Desipramine 0-3 carbonic anhydrase 1 Rattus norvegicus 62-65 18800064-0 2009 Desipramine modulation of alpha-, gamma-synuclein, and the norepinephrine transporter in an animal model of depression. Desipramine 0-11 synuclein, gamma Rattus norvegicus 34-49 18800064-0 2009 Desipramine modulation of alpha-, gamma-synuclein, and the norepinephrine transporter in an animal model of depression. Desipramine 0-11 solute carrier family 6 member 2 Rattus norvegicus 59-85 18800064-6 2009 In WKY rats there was an overexpression of gamma-Syn which was reduced following DMI treatment. Desipramine 81-84 synuclein, gamma Rattus norvegicus 43-52 18800064-7 2009 In parallel, DMI caused an increase in both alpha-Syn and NET in the frontal cortex. Desipramine 13-16 synuclein alpha Rattus norvegicus 44-53 19424057-3 2009 Here, we investigated whether chronic restraint stress (14 days) and antidepressant treatment [desipramine (DMI), 10 mg/kg intraperitoneally] induce changes in animal behavior and hippocampal levels of phospho-ERK1/2 and its substrate phospho-cAMP response element-binding protein (CREB). Desipramine 95-106 mitogen-activated protein kinase 3 Homo sapiens 210-216 19424057-3 2009 Here, we investigated whether chronic restraint stress (14 days) and antidepressant treatment [desipramine (DMI), 10 mg/kg intraperitoneally] induce changes in animal behavior and hippocampal levels of phospho-ERK1/2 and its substrate phospho-cAMP response element-binding protein (CREB). Desipramine 95-106 cAMP responsive element binding protein 1 Homo sapiens 282-286 19424057-3 2009 Here, we investigated whether chronic restraint stress (14 days) and antidepressant treatment [desipramine (DMI), 10 mg/kg intraperitoneally] induce changes in animal behavior and hippocampal levels of phospho-ERK1/2 and its substrate phospho-cAMP response element-binding protein (CREB). Desipramine 108-111 mitogen-activated protein kinase 3 Homo sapiens 210-216 19424057-7 2009 Bcl-2 mRNA was reduced in all hippocampal regions by stress, an effect independent of DMI treatment. Desipramine 86-89 BCL2 apoptosis regulator Homo sapiens 0-5 19424057-8 2009 However, immunoblot from hippocampal extracts revealed that stress increased BCL-2 levels, an effect prevented by chronic DMI. Desipramine 122-125 BCL2 apoptosis regulator Homo sapiens 77-82 19424057-9 2009 These results suggest that ERKs and BDNF may be altered in depressive disorder, modifications that are sensitive to DMI action. Desipramine 116-119 mitogen-activated protein kinase 3 Homo sapiens 27-31 19424057-9 2009 These results suggest that ERKs and BDNF may be altered in depressive disorder, modifications that are sensitive to DMI action. Desipramine 116-119 brain derived neurotrophic factor Homo sapiens 36-40 18923403-1 2009 The antidepressant desipramine inhibits the reuptake of norepinephrine (NE), leading to activation of both pre- and postsynaptic adrenergic receptors, including alpha-1, alpha-2, beta-1, and beta-2 subtypes. Desipramine 19-30 hemoglobin, beta adult minor chain Mus musculus 191-197 18923403-11 2009 In addition, desipramine (3-30 mg/kg) produced an antidepressant-like effect on behavior under a DRL 36-s schedule in mice deficient in both beta-1 and beta-2 adrenergic receptors. Desipramine 13-24 hemoglobin, beta adult major chain Mus musculus 141-147 18923403-11 2009 In addition, desipramine (3-30 mg/kg) produced an antidepressant-like effect on behavior under a DRL 36-s schedule in mice deficient in both beta-1 and beta-2 adrenergic receptors. Desipramine 13-24 hemoglobin, beta adult minor chain Mus musculus 152-158 19001559-0 2009 The effects of desvenlafaxine and paroxetine on the pharmacokinetics of the cytochrome P450 2D6 substrate desipramine in healthy adults. Desipramine 106-117 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 76-95 19001559-3 2009 The CYP2D6 substrate desipramine (50 mg) was administered alone on day 1 and coadministered on day 6 of dosing with either desvenlafaxine or paroxetine. Desipramine 21-32 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 4-10 18975044-5 2009 Zn-deficient mice showed exaggerated stress-evoked immediate-early gene expression in the amygdala which was normalised following DMI treatment. Desipramine 130-133 jun proto-oncogene Mus musculus 51-66 19067522-5 2009 Pretreatment of the monkey with the selective NET inhibitor, desipramine, decreased the specific binding for both [(11)C]1 and [(11)C]4. Desipramine 61-72 solute carrier family 6 member 2 Macaca mulatta 46-49 18507712-3 2009 Desipramine, a monoamine uptake blocker was able to abolish the toxic effects of MPP(+) but not H(2)O(2) in reduction of cell viability. Desipramine 0-11 M-phase phosphoprotein 6 Homo sapiens 81-84 18809731-0 2008 An assessment of drug-drug interactions: the effect of desvenlafaxine and duloxetine on the pharmacokinetics of the CYP2D6 probe desipramine in healthy subjects. Desipramine 129-140 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 116-122 18393297-1 2008 It has been reported that metformin was primarily metabolized via hepatic CYP2C11, 2D1, and 3A1/2 in rats, and the expression and mRNA levels of hepatic CYP2C11 and 3A1 decreased and increased, respectively, whereas the expression of CYP2D1 was not changed in rat model of diabetes induced by streptozotocin (DMIS). Desipramine 309-313 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 74-81 18393297-1 2008 It has been reported that metformin was primarily metabolized via hepatic CYP2C11, 2D1, and 3A1/2 in rats, and the expression and mRNA levels of hepatic CYP2C11 and 3A1 decreased and increased, respectively, whereas the expression of CYP2D1 was not changed in rat model of diabetes induced by streptozotocin (DMIS). Desipramine 309-313 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 153-160 18682257-8 2008 DMI and FLU increased FBP immunoreactivity in both cortical and hippocampal neurons. Desipramine 0-3 far upstream element binding protein 1 Rattus norvegicus 22-25 18791062-9 2008 Desipramine at the dose of 20 mg/kg produced more decrease in the level of TNF-alpha compared with the effect of the smaller dose, but fluoxetine at 10 mg/kg diminished more in the level of IL-1beta compared with the effect of the larger dose. Desipramine 0-11 tumor necrosis factor Rattus norvegicus 75-84 18418364-0 2008 Norepinephrine transporter regulation mediates the long-term behavioral effects of the antidepressant desipramine. Desipramine 102-113 solute carrier family 6 member 2 Rattus norvegicus 0-26 18418364-1 2008 The relationship between the ability of repeated desipramine treatment to cause downregulation of the norepinephrine transporter (NET) and produce antidepressant-like effects on behavior was determined. Desipramine 49-60 solute carrier family 6 member 2 Rattus norvegicus 102-128 18418364-1 2008 The relationship between the ability of repeated desipramine treatment to cause downregulation of the norepinephrine transporter (NET) and produce antidepressant-like effects on behavior was determined. Desipramine 49-60 solute carrier family 6 member 2 Rattus norvegicus 130-133 18418364-2 2008 Treatment of rats with 15 mg/kg per day desipramine reduced NET expression, measured by (3)H-nisoxetine binding and SDS-PAGE/immunoblotting, in cerebral cortex and hippocampus and reduced the time of immobility in the forced-swim test. Desipramine 40-51 solute carrier family 6 member 2 Rattus norvegicus 60-63 18815045-7 2008 Desipramine and fluoxetine, specific inhibitors of NET and SERT, respectively, both enhanced the inhibitory effects of propofol but reduced the inhibitory effects of ketamine on NET and SERT functions. Desipramine 0-11 solute carrier family 6 member 4 Homo sapiens 59-63 18418364-7 2008 The present results suggest an important role for NET regulation in the long-term behavioral effects of desipramine and are consistent with clinical data suggesting that enhanced noradrenergic neurotransmission is necessary, but not sufficient, for its antidepressant actions. Desipramine 104-115 solute carrier family 6 member 2 Rattus norvegicus 50-53 19090348-6 2008 Biochemically, citalopram and desipramine treatment significantly restored depleted reduced glutathione, catalase activity, attenuated raised lipid peroxidation and nitrite level as compared to control (72 hr sleep-deprived) animals. Desipramine 30-41 catalase Mus musculus 105-113 18815045-7 2008 Desipramine and fluoxetine, specific inhibitors of NET and SERT, respectively, both enhanced the inhibitory effects of propofol but reduced the inhibitory effects of ketamine on NET and SERT functions. Desipramine 0-11 solute carrier family 6 member 4 Homo sapiens 186-190 18478316-1 2008 PURPOSE: It has been reported that telithromycin is primarily metabolized via hepatic CYP3A1/2 in rats, the expression and/or mRNA level of hepatic CYP3A1/2 increase in rat model of diabetes mellitus induced by alloxan (DMIA) or streptozotocin (DMIS), and intestinal CYP3A1/2 enzyme activity decreases in rat model of DMIS. Desipramine 245-249 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 148-154 18698001-9 2008 Nisoxetine and desipramine, selective norepinephrine transporter inhibitors, also reduce locomotor activity in wild-type and knockin mice. Desipramine 15-26 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 38-64 18794257-6 2008 Data were compared with those obtained in 10 sham mice and correlated with the cardiac and hepatic tissue expression of norepinephrine transporter (NET) as assessed with a (3)H-desipramine saturation binding assay. Desipramine 177-188 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 120-146 18794257-6 2008 Data were compared with those obtained in 10 sham mice and correlated with the cardiac and hepatic tissue expression of norepinephrine transporter (NET) as assessed with a (3)H-desipramine saturation binding assay. Desipramine 177-188 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 148-151 18664374-4 2008 RESULTS: Compared to controls, significant reduction of mRNA levels of GR-beta under imipramine and of c-jun under desipramine treatment were found. Desipramine 115-126 Jun proto-oncogene, AP-1 transcription factor subunit Homo sapiens 103-108 18791062-8 2008 Desipramine and fluoxetine, at either dose, significantly reduced TNF-alpha and IL-beta. Desipramine 0-11 tumor necrosis factor Rattus norvegicus 66-75 18606486-0 2008 Desipramine-induced apoptosis in human PC3 prostate cancer cells: activation of JNK kinase and caspase-3 pathways and a protective role of [Ca2+]i elevation. Desipramine 0-11 chromobox 8 Homo sapiens 39-42 18606486-0 2008 Desipramine-induced apoptosis in human PC3 prostate cancer cells: activation of JNK kinase and caspase-3 pathways and a protective role of [Ca2+]i elevation. Desipramine 0-11 mitogen-activated protein kinase 8 Homo sapiens 80-83 18606486-0 2008 Desipramine-induced apoptosis in human PC3 prostate cancer cells: activation of JNK kinase and caspase-3 pathways and a protective role of [Ca2+]i elevation. Desipramine 0-11 caspase 3 Homo sapiens 95-104 18606486-1 2008 The antidepressant desipramine has been shown to induce a rise in cytosolic Ca2+ levels ([Ca2+]i) and cytotoxicity in human PC3 prostate cancer cells, but the mechanisms underlying its cytotoxic effect is unclear. Desipramine 19-30 chromobox 8 Homo sapiens 124-127 18606486-6 2008 Immunoblotting data revealed that desipramine activated the phosphorylation of c-Jun NH2-terminal kinase (JNK), but not extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 34-45 mitogen-activated protein kinase 8 Homo sapiens 79-104 18606486-6 2008 Immunoblotting data revealed that desipramine activated the phosphorylation of c-Jun NH2-terminal kinase (JNK), but not extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 34-45 mitogen-activated protein kinase 8 Homo sapiens 106-109 18606486-6 2008 Immunoblotting data revealed that desipramine activated the phosphorylation of c-Jun NH2-terminal kinase (JNK), but not extracellular signal-regulated kinase (ERK) and p38 mitogen-activated protein kinase (MAPK). Desipramine 34-45 mitogen-activated protein kinase 1 Homo sapiens 206-210 18606486-9 2008 Immunoblotting data suggest that BAPTA/AM pretreatment enhanced desipramine-evoked JNK phosphorylation and caspase-3 cleavage. Desipramine 64-75 mitogen-activated protein kinase 8 Homo sapiens 83-86 18606486-10 2008 The results suggest that in PC3 cells, desipramine caused apoptosis via inducing JNK-associated caspase-3 activation, and [Ca2+]i rises may slow down or alleviate desipramine-induced cytotoxicity. Desipramine 39-50 chromobox 8 Homo sapiens 28-31 18606486-10 2008 The results suggest that in PC3 cells, desipramine caused apoptosis via inducing JNK-associated caspase-3 activation, and [Ca2+]i rises may slow down or alleviate desipramine-induced cytotoxicity. Desipramine 39-50 mitogen-activated protein kinase 8 Homo sapiens 81-84 18606486-10 2008 The results suggest that in PC3 cells, desipramine caused apoptosis via inducing JNK-associated caspase-3 activation, and [Ca2+]i rises may slow down or alleviate desipramine-induced cytotoxicity. Desipramine 39-50 caspase 3 Homo sapiens 96-105 18606486-10 2008 The results suggest that in PC3 cells, desipramine caused apoptosis via inducing JNK-associated caspase-3 activation, and [Ca2+]i rises may slow down or alleviate desipramine-induced cytotoxicity. Desipramine 163-174 chromobox 8 Homo sapiens 28-31 18478316-1 2008 PURPOSE: It has been reported that telithromycin is primarily metabolized via hepatic CYP3A1/2 in rats, the expression and/or mRNA level of hepatic CYP3A1/2 increase in rat model of diabetes mellitus induced by alloxan (DMIA) or streptozotocin (DMIS), and intestinal CYP3A1/2 enzyme activity decreases in rat model of DMIS. Desipramine 245-249 cytochrome P450, family 3, subfamily a, polypeptide 23-polypeptide 1 Rattus norvegicus 148-154 18601743-6 2008 Short-term treatment (6 hours) with reboxetine or desipramine reduced total BDNF, whereas long-term treatment (48 hours) significantly increased total BDNF mRNA levels. Desipramine 50-61 brain derived neurotrophic factor Homo sapiens 76-80 18509851-4 2008 The influence of distinct classes of antidepressants, namely, electroconvulsive seizure, fluoxetine, tranylcypromine, and desipramine, on the gene expression of MMP-2, MMP-9, and TIMPs 1-4 in the hippocampus was determined using radioactive in situ hybridization. Desipramine 122-133 matrix metallopeptidase 2 Rattus norvegicus 161-166 18509851-4 2008 The influence of distinct classes of antidepressants, namely, electroconvulsive seizure, fluoxetine, tranylcypromine, and desipramine, on the gene expression of MMP-2, MMP-9, and TIMPs 1-4 in the hippocampus was determined using radioactive in situ hybridization. Desipramine 122-133 matrix metallopeptidase 9 Rattus norvegicus 168-173 18509851-4 2008 The influence of distinct classes of antidepressants, namely, electroconvulsive seizure, fluoxetine, tranylcypromine, and desipramine, on the gene expression of MMP-2, MMP-9, and TIMPs 1-4 in the hippocampus was determined using radioactive in situ hybridization. Desipramine 122-133 TIMP metallopeptidase inhibitor 1 Rattus norvegicus 179-188 17667959-2 2008 First, CYP2C19 converts imipramine into the active metabolite desipramine, which is then inactivated by CYP2D6. Desipramine 62-73 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 7-14 18420781-1 2008 There are documented clinical drug-drug interactions between bupropion and the CYP2D6-metabolized drug desipramine resulting in marked (5-fold) increases in desipramine exposure. Desipramine 103-114 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 79-85 18420781-1 2008 There are documented clinical drug-drug interactions between bupropion and the CYP2D6-metabolized drug desipramine resulting in marked (5-fold) increases in desipramine exposure. Desipramine 157-168 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 79-85 18420781-7 2008 This work indicates that the reductive metabolites of bupropion are potent competitive CYP2D6 inhibitors in vivo and provides a mechanistic explanation for the clinical drug-drug interaction between bupropion and desipramine. Desipramine 213-224 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 87-93 18266934-5 2008 drSERT has a higher affinity towards compounds of the imipramine class, desipramine in particular, exhibiting a 35-fold increased affinity compared to hSERT. Desipramine 72-83 solute carrier family 6 member 4 Homo sapiens 151-156 17727882-0 2008 Enhanced norepinephrine output during long-term desipramine treatment: a possible role for the extraneuronal monoamine transporter (SLC22A3). Desipramine 48-59 solute carrier family 22 member 3 Homo sapiens 132-139 18438645-6 2008 Moreover, at the neural level, DMI treatment led to a decrease in FST-induced c-fos messenger RNA levels in medial prefrontal cortex (PFC) and paraventricular nucleus of the hypothalamus (PVN) in LR but not HR animals. Desipramine 31-34 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 78-83 17667959-2 2008 First, CYP2C19 converts imipramine into the active metabolite desipramine, which is then inactivated by CYP2D6. Desipramine 62-73 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 104-110 17667959-3 2008 This retrospective single center study aimed to prove whether CYP2C19 and ample CYP2D6 genotyping (taking into consideration four null alleles and three decreased-activity alleles) could be used to predict imipramine and desipramine plasma concentrations in depressed patients, and whether genotype-based drug dose recommendations might assist in the early management of imipramine pharmacotherapy. Desipramine 221-232 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 62-69 17667959-3 2008 This retrospective single center study aimed to prove whether CYP2C19 and ample CYP2D6 genotyping (taking into consideration four null alleles and three decreased-activity alleles) could be used to predict imipramine and desipramine plasma concentrations in depressed patients, and whether genotype-based drug dose recommendations might assist in the early management of imipramine pharmacotherapy. Desipramine 221-232 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 80-86 17667959-5 2008 Desipramine and imipramine+desipramine plasma concentration per drug dose unit, imipramine dose at steady state, and imipramine dose requirement significantly depended on CYP2D6 genotype (Kruskal-Wallis test, P<0.0001). Desipramine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 171-177 17667959-5 2008 Desipramine and imipramine+desipramine plasma concentration per drug dose unit, imipramine dose at steady state, and imipramine dose requirement significantly depended on CYP2D6 genotype (Kruskal-Wallis test, P<0.0001). Desipramine 27-38 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 171-177 17667959-8 2008 Our protocol for CYP2D6 genotyping will thus importantly aid in the prediction of imipramine metabolism, allowing for the use of an adjusted starting dose and faster achievement of predefined imipramine+desipramine plasma levels in all genetic patient subgroups. Desipramine 203-214 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 17-23 18342622-4 2008 Dopamine transporter (DAT) mutant flies or flies treated with the DAT inhibitor desipramine exhibited poor memory retention. Desipramine 80-91 Dopamine transporter Drosophila melanogaster 66-69 18420612-7 2008 The level of DMI had less effect on plasma concentrations of IGF-I at d 14 (72.2 vs. 57.1 ng/mL). Desipramine 13-16 insulin like growth factor 1 Bos taurus 61-66 18420612-14 2008 Altering the ME or DMI components of the pasture-based diets produced changes in plasma IGF-I concentrations that did not become stabilized for 3 wk, but were then highly repeatable for individual cows within each dietary group. Desipramine 19-22 insulin like growth factor 1 Bos taurus 88-93 17981365-2 2008 The co-administration of local anesthetics with desipramine reversed the behavioral sensitization and down-regulation of NET function induced by desipramine. Desipramine 48-59 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 121-124 18243568-7 2008 Fluoxetine and desipramine administration significantly attenuated plasma ACTH and CORT levels in male and female Avpr1b KO mice when compared to their wild-type counterparts. Desipramine 15-26 cortistatin Mus musculus 83-87 18243568-7 2008 Fluoxetine and desipramine administration significantly attenuated plasma ACTH and CORT levels in male and female Avpr1b KO mice when compared to their wild-type counterparts. Desipramine 15-26 arginine vasopressin receptor 1B Mus musculus 114-120 18310890-5 2008 Imipramine, desipramine, mazindol, and GBR12909 competitively inhibited PROG 21-hydroxylation mediated by CYP2D4 and/or CYP2D6, and all psychotropic drugs inhibited ALLO 21-hydroxylation mediated by CYP2D4 and/or CYP2D6. Desipramine 12-23 cytochrome P450, family 2, subfamily d, polypeptide 4 Rattus norvegicus 106-112 18310890-5 2008 Imipramine, desipramine, mazindol, and GBR12909 competitively inhibited PROG 21-hydroxylation mediated by CYP2D4 and/or CYP2D6, and all psychotropic drugs inhibited ALLO 21-hydroxylation mediated by CYP2D4 and/or CYP2D6. Desipramine 12-23 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 120-126 18310890-5 2008 Imipramine, desipramine, mazindol, and GBR12909 competitively inhibited PROG 21-hydroxylation mediated by CYP2D4 and/or CYP2D6, and all psychotropic drugs inhibited ALLO 21-hydroxylation mediated by CYP2D4 and/or CYP2D6. Desipramine 12-23 cytochrome P450, family 2, subfamily d, polypeptide 4 Rattus norvegicus 199-205 18310890-5 2008 Imipramine, desipramine, mazindol, and GBR12909 competitively inhibited PROG 21-hydroxylation mediated by CYP2D4 and/or CYP2D6, and all psychotropic drugs inhibited ALLO 21-hydroxylation mediated by CYP2D4 and/or CYP2D6. Desipramine 12-23 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 213-219 18310890-6 2008 The inhibition constants (Ki values) of imipramine, desipramine, and mazindol against the 21-hydroxylation of PROG and ALLO by CYP2D6 were lower than those by CYP2D4. Desipramine 52-63 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 127-133 18310890-6 2008 The inhibition constants (Ki values) of imipramine, desipramine, and mazindol against the 21-hydroxylation of PROG and ALLO by CYP2D6 were lower than those by CYP2D4. Desipramine 52-63 cytochrome P450, family 2, subfamily d, polypeptide 4 Rattus norvegicus 159-165 17981365-0 2008 Down-regulation of norepinephrine transporter expression on membrane surface induced by chronic administration of desipramine and the antagonism by co-administration of local anesthetics in mice. Desipramine 114-125 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 19-45 17981365-1 2008 We have previously shown that chronic administration of the antidepressant desipramine, a norepinephrine transporter (NET) inhibitor to mice markedly enhanced convulsions induced by local anesthetics and that behavioral sensitization may be relevant to decreased [(3)H]norepinephrine uptake by the isolated hippocampus. Desipramine 75-86 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 90-116 17981365-1 2008 We have previously shown that chronic administration of the antidepressant desipramine, a norepinephrine transporter (NET) inhibitor to mice markedly enhanced convulsions induced by local anesthetics and that behavioral sensitization may be relevant to decreased [(3)H]norepinephrine uptake by the isolated hippocampus. Desipramine 75-86 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 118-121 17981365-2 2008 The co-administration of local anesthetics with desipramine reversed the behavioral sensitization and down-regulation of NET function induced by desipramine. Desipramine 145-156 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 121-124 17981365-3 2008 The present study aimed to elucidate whether chronic treatment with desipramine regulates the expression of NET protein examined in membrane fractions in various brain regions and whether co-administration of local anesthetics affects the desipramine-induced alteration of NET expression. Desipramine 68-79 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 108-111 17981365-3 2008 The present study aimed to elucidate whether chronic treatment with desipramine regulates the expression of NET protein examined in membrane fractions in various brain regions and whether co-administration of local anesthetics affects the desipramine-induced alteration of NET expression. Desipramine 239-250 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 273-276 17981365-9 2008 However, administration of desipramine significantly reduced the amount of immunoreactive NET in the cell-surface protein fraction. Desipramine 27-38 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 90-93 17981365-11 2008 These results indicate that the NET down-regulation indicated by the reduction of [(3)H]nisoxetine binding was induced by administration of desipramine via decrease of NET localization on the cell surface. Desipramine 140-151 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 32-35 17981365-11 2008 These results indicate that the NET down-regulation indicated by the reduction of [(3)H]nisoxetine binding was induced by administration of desipramine via decrease of NET localization on the cell surface. Desipramine 140-151 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 168-171 17981365-12 2008 The antagonistic actions of local anesthetics against NET down-regulation by desipramine were related to alterations of the cell-surface localization of NET. Desipramine 77-88 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 54-57 17981365-12 2008 The antagonistic actions of local anesthetics against NET down-regulation by desipramine were related to alterations of the cell-surface localization of NET. Desipramine 77-88 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 153-156 18171937-6 2008 Similar treatment with the norepinephrine transporter (NET) inhibitor, desipramine, did not adversely affect adult behavior, suggesting that 5-HT and norepinephrine (NE) do not share the same effects on brain development. Desipramine 71-82 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 27-53 18028907-9 2008 GRK2/3 levels remained unchanged and increased, respectively, in the cortex and the hippocampus, after chronic exposure to desipramine. Desipramine 123-134 beta-adrenergic receptor kinase 1 Cavia porcellus 0-6 18028907-11 2008 On the whole, the present data suggest that alpha(2)-adrenoceptor and mu-opioid receptor expression and transcription are similarly influenced by chronic treatment with desipramine, in the guinea pig cortex and hippocampus. Desipramine 169-180 mu-type opioid receptor Cavia porcellus 70-88 18048068-0 2008 The differential regulation of BDNF and TrkB levels in juvenile rats after four days of escitalopram and desipramine treatment. Desipramine 105-116 brain-derived neurotrophic factor Rattus norvegicus 31-35 18048068-0 2008 The differential regulation of BDNF and TrkB levels in juvenile rats after four days of escitalopram and desipramine treatment. Desipramine 105-116 neurotrophic receptor tyrosine kinase 2 Rattus norvegicus 40-44 18974610-6 2008 In addition, the method was applied to the CYP2D6-mediated desipramine-quinidine interaction: the predicted 6.4-fold increase of desipramine AUC by quinidine was consistent with the observed 6.7-fold increase of AUC in the clinical drug-drug interaction study. Desipramine 59-70 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 43-49 17356567-0 2007 The antidepressant desipramine requires the ABCB1 (Mdr1)-type p-glycoprotein to upregulate the glucocorticoid receptor in mice. Desipramine 19-30 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 44-49 17356567-0 2007 The antidepressant desipramine requires the ABCB1 (Mdr1)-type p-glycoprotein to upregulate the glucocorticoid receptor in mice. Desipramine 19-30 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 51-55 17356567-0 2007 The antidepressant desipramine requires the ABCB1 (Mdr1)-type p-glycoprotein to upregulate the glucocorticoid receptor in mice. Desipramine 19-30 phosphoglycolate phosphatase Mus musculus 62-76 17356567-0 2007 The antidepressant desipramine requires the ABCB1 (Mdr1)-type p-glycoprotein to upregulate the glucocorticoid receptor in mice. Desipramine 19-30 nuclear receptor subfamily 3, group C, member 1 Mus musculus 95-118 17356567-4 2007 Here, the tricyclic antidepressant, desipramine (20 mg/kg/day, i.p., for seven days), was administered to abcb1ab MDR PGP knockout mice (congenic on the FVB/N background strain) and to FVB/N controls. Desipramine 36-47 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 106-111 17356567-4 2007 Here, the tricyclic antidepressant, desipramine (20 mg/kg/day, i.p., for seven days), was administered to abcb1ab MDR PGP knockout mice (congenic on the FVB/N background strain) and to FVB/N controls. Desipramine 36-47 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 114-117 17356567-4 2007 Here, the tricyclic antidepressant, desipramine (20 mg/kg/day, i.p., for seven days), was administered to abcb1ab MDR PGP knockout mice (congenic on the FVB/N background strain) and to FVB/N controls. Desipramine 36-47 phosphoglycolate phosphatase Mus musculus 118-121 17356567-6 2007 In FVB/N controls, desipramine induced a significant upregulation of GR mRNA in the CA1 region (+31%; p=0.045); in contrast, in abcb1ab (-/-) mice, desipramine induced a significant downregulation of GR mRNA in the CA1 region (-45%; p=0.004). Desipramine 19-30 nuclear receptor subfamily 3, group C, member 1 Mus musculus 69-71 17356567-6 2007 In FVB/N controls, desipramine induced a significant upregulation of GR mRNA in the CA1 region (+31%; p=0.045); in contrast, in abcb1ab (-/-) mice, desipramine induced a significant downregulation of GR mRNA in the CA1 region (-45%; p=0.004). Desipramine 19-30 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 128-133 17356567-6 2007 In FVB/N controls, desipramine induced a significant upregulation of GR mRNA in the CA1 region (+31%; p=0.045); in contrast, in abcb1ab (-/-) mice, desipramine induced a significant downregulation of GR mRNA in the CA1 region (-45%; p=0.004). Desipramine 19-30 nuclear receptor subfamily 3, group C, member 1 Mus musculus 200-202 17356567-8 2007 Desipramine decreased corticosterone levels in both FVB/N controls and in abcb1ab (-/-) mice, but in abcb1ab (-/-) mice the effects were smaller. Desipramine 0-11 ATP-binding cassette, sub-family B (MDR/TAP), member 1B Mus musculus 74-79 17356567-10 2007 Finally, desipramine reduced Mdr1a mRNA expression across all hippocampus areas (-9 to -23%), but had no effect on 11beta-HSD1 mRNA expression. Desipramine 9-20 ATP-binding cassette, sub-family B (MDR/TAP), member 1A Mus musculus 29-34 17979223-5 2007 The NET blocker desipramine reduced the uptake of [ (131)I] 12a only to a small extent, further suggesting a limited role of NET in its binding and accumulation. Desipramine 16-27 solute carrier family 6 member 2 Homo sapiens 4-7 18324519-5 2008 Pre-treatment with ROS scavenger NAC, ASMase inhibitor desipramine or JNK inhibitor SP600125 was found to effectively prohibit UVA-induced apoptosis and desipramine markedly blocked phosphorylation of JNK. Desipramine 55-66 sphingomyelin phosphodiesterase 1 Homo sapiens 38-44 18324519-5 2008 Pre-treatment with ROS scavenger NAC, ASMase inhibitor desipramine or JNK inhibitor SP600125 was found to effectively prohibit UVA-induced apoptosis and desipramine markedly blocked phosphorylation of JNK. Desipramine 55-66 mitogen-activated protein kinase 8 Homo sapiens 201-204 18324519-5 2008 Pre-treatment with ROS scavenger NAC, ASMase inhibitor desipramine or JNK inhibitor SP600125 was found to effectively prohibit UVA-induced apoptosis and desipramine markedly blocked phosphorylation of JNK. Desipramine 153-164 synuclein alpha Homo sapiens 33-36 18324519-5 2008 Pre-treatment with ROS scavenger NAC, ASMase inhibitor desipramine or JNK inhibitor SP600125 was found to effectively prohibit UVA-induced apoptosis and desipramine markedly blocked phosphorylation of JNK. Desipramine 153-164 sphingomyelin phosphodiesterase 1 Homo sapiens 38-44 18324519-5 2008 Pre-treatment with ROS scavenger NAC, ASMase inhibitor desipramine or JNK inhibitor SP600125 was found to effectively prohibit UVA-induced apoptosis and desipramine markedly blocked phosphorylation of JNK. Desipramine 153-164 mitogen-activated protein kinase 8 Homo sapiens 70-73 18324519-5 2008 Pre-treatment with ROS scavenger NAC, ASMase inhibitor desipramine or JNK inhibitor SP600125 was found to effectively prohibit UVA-induced apoptosis and desipramine markedly blocked phosphorylation of JNK. Desipramine 153-164 mitogen-activated protein kinase 8 Homo sapiens 201-204 17166518-5 2008 They received desipramine for 6 weeks in order to simulate the length of time and dosing used previously to obtain an IR-1 down-regulation and a therapeutic response in depressed patients. Desipramine 14-25 nischarin Homo sapiens 118-122 17976922-9 2008 We conclude that pharmacological blockade of CB1 signaling shares antidepressant-like behavioral effects with desipramine, but reveals opposite effects on HPA axis activity. Desipramine 110-121 cannabinoid receptor 1 (brain) Mus musculus 45-48 17850785-5 2007 Competition binding studies using [(3)H]nisoxetine and [(3)H]citalopram showed desipramine to be 25 times more selective for the rat norepinephrine as compared to serotonin transporter (6.2 nM vs. 158 nM) whereas desmethyldesipramine is 12 times more selective for the serotonin over the norepinephrine transporter (12.8 nM vs. 153 nM). Desipramine 79-90 solute carrier family 6 member 2 Rattus norvegicus 288-314 17850785-6 2007 Interestingly, the affinity of desmethyldesipramine for the serotonin transporter is similar to the affinity of desipramine for the norepinephrine transporter. Desipramine 40-51 solute carrier family 6 member 4 Rattus norvegicus 60-81 17850785-8 2007 Thus, the pharmacological effects of desipramine in rats may be attributed not only to the inhibition of the norepinephrine transporter by desipramine but also to the inhibition of serotonin transporter by the active metabolite desmethyldesipramine. Desipramine 37-48 solute carrier family 6 member 2 Rattus norvegicus 109-135 17850785-8 2007 Thus, the pharmacological effects of desipramine in rats may be attributed not only to the inhibition of the norepinephrine transporter by desipramine but also to the inhibition of serotonin transporter by the active metabolite desmethyldesipramine. Desipramine 37-48 solute carrier family 6 member 4 Rattus norvegicus 181-202 17850785-8 2007 Thus, the pharmacological effects of desipramine in rats may be attributed not only to the inhibition of the norepinephrine transporter by desipramine but also to the inhibition of serotonin transporter by the active metabolite desmethyldesipramine. Desipramine 139-150 solute carrier family 6 member 2 Rattus norvegicus 109-135 18974610-6 2008 In addition, the method was applied to the CYP2D6-mediated desipramine-quinidine interaction: the predicted 6.4-fold increase of desipramine AUC by quinidine was consistent with the observed 6.7-fold increase of AUC in the clinical drug-drug interaction study. Desipramine 129-140 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 43-49 17584982-5 2007 We demonstrated that the LPS induced TNF-alpha response is in direct correlation with the biophase level of NE, as it is significantly higher when the release of NE of vesicular origin was completely inhibited in an animal model of depression (reserpine treatment) and it is significantly lower in the case of increasing biophase levels of NE by genetic (NET-KO) or chemical (desipramine) disruption of NE reuptake. Desipramine 376-387 toll-like receptor 4 Mus musculus 25-28 17584982-5 2007 We demonstrated that the LPS induced TNF-alpha response is in direct correlation with the biophase level of NE, as it is significantly higher when the release of NE of vesicular origin was completely inhibited in an animal model of depression (reserpine treatment) and it is significantly lower in the case of increasing biophase levels of NE by genetic (NET-KO) or chemical (desipramine) disruption of NE reuptake. Desipramine 376-387 tumor necrosis factor Mus musculus 37-46 17584982-6 2007 IL-10 was changed inversely to TNF-alpha levels only in the desipramine-treated animals. Desipramine 60-71 interleukin 10 Mus musculus 0-5 17584982-6 2007 IL-10 was changed inversely to TNF-alpha levels only in the desipramine-treated animals. Desipramine 60-71 tumor necrosis factor Mus musculus 31-40 17690258-3 2007 We determined the crystal structure at 2.9 angstroms of the bacterial leucine transporter (LeuT), a homolog of SERT, NET, and DAT, in complex with leucine and the antidepressant desipramine. Desipramine 178-189 Leucine transport, high Homo sapiens 70-89 17690258-3 2007 We determined the crystal structure at 2.9 angstroms of the bacterial leucine transporter (LeuT), a homolog of SERT, NET, and DAT, in complex with leucine and the antidepressant desipramine. Desipramine 178-189 Leucine transport, high Homo sapiens 91-95 17690258-3 2007 We determined the crystal structure at 2.9 angstroms of the bacterial leucine transporter (LeuT), a homolog of SERT, NET, and DAT, in complex with leucine and the antidepressant desipramine. Desipramine 178-189 solute carrier family 6 member 4 Homo sapiens 111-115 17690258-3 2007 We determined the crystal structure at 2.9 angstroms of the bacterial leucine transporter (LeuT), a homolog of SERT, NET, and DAT, in complex with leucine and the antidepressant desipramine. Desipramine 178-189 solute carrier family 6 member 3 Homo sapiens 126-129 17690258-7 2007 Mutagenesis experiments on human SERT and DAT indicate that both the desipramine-binding site and its inhibition mechanism are probably conserved in the human neurotransmitter transporters. Desipramine 69-80 solute carrier family 6 member 4 Homo sapiens 33-37 17690258-7 2007 Mutagenesis experiments on human SERT and DAT indicate that both the desipramine-binding site and its inhibition mechanism are probably conserved in the human neurotransmitter transporters. Desipramine 69-80 solute carrier family 6 member 3 Homo sapiens 42-45 17477857-16 2007 In vitro, desipramine and fluoxetine dose-dependently inhibited the release of TNF-alpha from LPS-treated monocytes. Desipramine 10-21 tumor necrosis factor Rattus norvegicus 79-88 17634380-3 2007 Here, we show that, at baseline, CREB(alpha delta) mutant mice exhibited increased hippocampal cell proliferation and neurogenesis compared with wild-type (WT) controls, effects similar to those observed in WT mice after chronic desipramine (DMI) administration. Desipramine 229-240 cAMP responsive element binding protein 1 Mus musculus 33-37 17634380-3 2007 Here, we show that, at baseline, CREB(alpha delta) mutant mice exhibited increased hippocampal cell proliferation and neurogenesis compared with wild-type (WT) controls, effects similar to those observed in WT mice after chronic desipramine (DMI) administration. Desipramine 242-245 cAMP responsive element binding protein 1 Mus musculus 33-37 17382299-3 2007 At nanomolar concentrations, CLOM and DMI increased expression of phospho-CREB (cyclic AMP response element binding protein). Desipramine 38-41 cAMP responsive element binding protein 1 Homo sapiens 74-78 17540003-7 2007 We found that acidic sphingomyelinase activity was induced by acidic extracellular pH and that the specific acidic sphingomyelinase inhibitors (perhexiline and desipramine) and siRNA targeting aSMase/smpd1 could inhibit acidic extracellular pH-induced matrix metalloproteinase-9 expression. Desipramine 160-171 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 193-199 17540003-7 2007 We found that acidic sphingomyelinase activity was induced by acidic extracellular pH and that the specific acidic sphingomyelinase inhibitors (perhexiline and desipramine) and siRNA targeting aSMase/smpd1 could inhibit acidic extracellular pH-induced matrix metalloproteinase-9 expression. Desipramine 160-171 sphingomyelin phosphodiesterase 1, acid lysosomal Mus musculus 200-205 17540003-7 2007 We found that acidic sphingomyelinase activity was induced by acidic extracellular pH and that the specific acidic sphingomyelinase inhibitors (perhexiline and desipramine) and siRNA targeting aSMase/smpd1 could inhibit acidic extracellular pH-induced matrix metalloproteinase-9 expression. Desipramine 160-171 matrix metallopeptidase 9 Mus musculus 252-278 17510525-0 2007 Desipramine activated Bcl-2 expression and inhibited lipopolysaccharide-induced apoptosis in hippocampus-derived adult neural stem cells. Desipramine 0-11 BCL2, apoptosis regulator Rattus norvegicus 22-27 17417877-4 2007 FCS-binding studies with the fluorescently labeled ligand Alexa532-noradrenaline (Alexa532-NA) binding to beta2-AR of C6 cells showed a significant reduction in total beta2-AR binding after preincubation with hyperforin and hyperoside for 3 days, respectively, which was also found for DMI. Desipramine 286-289 adrenoceptor beta 2 Homo sapiens 106-114 17417877-4 2007 FCS-binding studies with the fluorescently labeled ligand Alexa532-noradrenaline (Alexa532-NA) binding to beta2-AR of C6 cells showed a significant reduction in total beta2-AR binding after preincubation with hyperforin and hyperoside for 3 days, respectively, which was also found for DMI. Desipramine 286-289 adrenoceptor beta 2 Homo sapiens 167-175 17267168-0 2007 Desipramine-induced Ca2+ movement and cytotoxicity in PC3 human prostate cancer cells. Desipramine 0-11 carbonic anhydrase 2 Homo sapiens 20-23 17267168-0 2007 Desipramine-induced Ca2+ movement and cytotoxicity in PC3 human prostate cancer cells. Desipramine 0-11 chromobox 8 Homo sapiens 54-57 17267168-12 2007 Collectively, the data suggest that in PC3 cells, desipramine induced a [Ca(2+)](i) increase by causing Ca(2+) release from endoplasmic reticulum in a phospholipase C-independent fashion and by inducing Ca(2+) influx. Desipramine 50-61 chromobox 8 Homo sapiens 39-42 17352382-5 2007 Both, hypoosmotic ROS generation and p47(phox) serine phosphorylation were sensitive to the acidic sphingomyelinase inhibitors AY9944 and desipramine, the protein kinase C (PKC)zeta-inhibitory pseudosubstrate peptide, the NMDA receptor antagonist MK-801 and the intracellular Ca(2+) chelator BAPTA-AM. Desipramine 138-149 NSFL1 (p97) cofactor (p47) Mus musculus 37-40 17510525-5 2007 First, we demonstrated that the expression of Bcl-2 mRNA and nestin in 2 microM DP-treated NSCs were up-regulated and detected by real-time reverse transcriptase polymerase chain reaction (RT-PCR). Desipramine 80-82 BCL2, apoptosis regulator Rattus norvegicus 46-51 17510525-6 2007 The results of Western blotting and immunofluorescent study confirmed that Bcl-2 protein expression was significantly increased in Day 3 DP-treated NSCs. Desipramine 137-139 BCL2, apoptosis regulator Rattus norvegicus 75-80 17510525-7 2007 Using the Bcl-2 small interfering RNA (siRNA) method, our results further showed that DP protects the lipopolysaccharide (LPS)-induced apoptosis in NSCs, in part by activating the expression of Bcl-2. Desipramine 86-88 BCL2, apoptosis regulator Rattus norvegicus 10-15 17510525-7 2007 Using the Bcl-2 small interfering RNA (siRNA) method, our results further showed that DP protects the lipopolysaccharide (LPS)-induced apoptosis in NSCs, in part by activating the expression of Bcl-2. Desipramine 86-88 BCL2, apoptosis regulator Rattus norvegicus 194-199 17510525-8 2007 Furthermore, DP treatment significantly inhibited the induction of proinflammatory factor interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha in the culture medium of LPS-treated NSCs mediated by Bcl-2 modulation. Desipramine 13-15 interleukin 1 beta Rattus norvegicus 90-112 17510525-8 2007 Furthermore, DP treatment significantly inhibited the induction of proinflammatory factor interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha in the culture medium of LPS-treated NSCs mediated by Bcl-2 modulation. Desipramine 13-15 interleukin 6 Rattus norvegicus 114-151 17510525-8 2007 Furthermore, DP treatment significantly inhibited the induction of proinflammatory factor interleukin (IL)-1beta, IL-6, and tumor necrosis factor-alpha in the culture medium of LPS-treated NSCs mediated by Bcl-2 modulation. Desipramine 13-15 BCL2, apoptosis regulator Rattus norvegicus 206-211 17510525-9 2007 The results of high performance liquid chromatography coupled to electrochemical detection further confirmed that DP significantly increased the functional production of serotonin (26+/-3.5 microM, DP-treated 96 h) and noradrenaline (50+/-8.9 microM, DP-treated 96 h) in NSCs through activation of the MAPK/ERK pathway and partially mediated by Bcl-2. Desipramine 114-116 Eph receptor B1 Rattus norvegicus 307-310 17510525-9 2007 The results of high performance liquid chromatography coupled to electrochemical detection further confirmed that DP significantly increased the functional production of serotonin (26+/-3.5 microM, DP-treated 96 h) and noradrenaline (50+/-8.9 microM, DP-treated 96 h) in NSCs through activation of the MAPK/ERK pathway and partially mediated by Bcl-2. Desipramine 114-116 BCL2, apoptosis regulator Rattus norvegicus 345-350 17510525-10 2007 In conclusion, the present results indicate that DP can increase neuroprotection ability by inhibiting the LPS-induced inflammatory process in NSCs via the modulation of Bcl-2 expression, as confirmed by the siRNA method. Desipramine 49-51 BCL2, apoptosis regulator Rattus norvegicus 170-175 16680561-2 2007 The purpose of this study was to evaluate the effect of cinacalcet on CYP2D6 activity, using desipramine as a probe substrate, in healthy subjects. Desipramine 93-104 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 70-76 17121970-10 2007 Alpha-amylase supplementation increased DMI (P = 0.05) and ADG (P = 0.03) during the initial 28 d on feed and carcass-adjusted ADG (P = 0.04) across corn processing methods. Desipramine 40-43 alpha-amylase Zea mays 0-13 17194442-4 2007 Paroxetine increased the interaction of GluR1 with Rab4A, and desipramine markedly increased the interaction of GluR1 with SAP97. Desipramine 62-73 glutamate ionotropic receptor AMPA type subunit 1 Rattus norvegicus 112-117 17194442-4 2007 Paroxetine increased the interaction of GluR1 with Rab4A, and desipramine markedly increased the interaction of GluR1 with SAP97. Desipramine 62-73 discs large MAGUK scaffold protein 1 Rattus norvegicus 123-128 17055479-8 2006 Levels of G protein-coupled receptor kinase 2/3 and of inhibitory G(i/o) proteins were significantly reduced in the myenteric plexus after chronic treatment with desipramine. Desipramine 162-173 beta-adrenergic receptor kinase 1 Cavia porcellus 10-47 16697351-5 2007 We also demonstrate that loss of BDNF in both male and female mice attenuates the actions of the antidepressant desipramine in the forced swim test. Desipramine 112-123 brain derived neurotrophic factor Mus musculus 33-37 17071817-7 2007 Other TCAs, such as amitriptyline, desipramine, and imipramine, also inhibited Kir4.1 currents in a similar voltage-dependent fashion. Desipramine 35-46 potassium inwardly rectifying channel subfamily J member 10 Homo sapiens 79-85 19356015-2 2007 The CYP form with the lowest in vitro K(i) is evaluated first clinically, employing a suitable probe drug like midazolam (CYP3A4), theophylline (CYP1A2), (S)-warfarin (CYP2C9) and desipramine (CYP2D6), and the NCE is classified as a "none", "weak", "moderate", or "strong" inhibitor. Desipramine 180-191 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 4-7 17069850-11 2007 The survival signal was further confirmed by increased phosphorylation of AKT and enhanced induction of expression of Bcl-2 during adaptation and its reversal with desipramine. Desipramine 164-175 AKT serine/threonine kinase 1 Rattus norvegicus 74-77 17069850-11 2007 The survival signal was further confirmed by increased phosphorylation of AKT and enhanced induction of expression of Bcl-2 during adaptation and its reversal with desipramine. Desipramine 164-175 BCL2, apoptosis regulator Rattus norvegicus 118-123 16388933-7 2006 Interestingly, exposure to a lower concentration (1 microM) of the antidepressants tended to increase T-cell-derived IL-10 production, with significant effects elicited by the noradrenaline reuptake inhibitors reboxetine and desipramine. Desipramine 225-236 interleukin 10 Homo sapiens 117-122 16710317-4 2006 Furthermore, chronic desipramine treatment resulted in a reduction in both secretion of corticosterone and the induction of the immediate early gene c-fos in the medial dorsal parvocellular region of the paraventricular nucleus of the hypothalamus (PVN) following a 5 min exposure to swim stress. Desipramine 21-32 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 149-154 16710317-5 2006 Acute treatment with the CB(1) receptor antagonist, AM251 (1 mg/kg), before exposure to swim stress, completely occluded the ability of desipramine to reduce both corticosterone secretion and induction of c-fos expression in the PVN. Desipramine 136-147 Fos proto-oncogene, AP-1 transcription factor subunit Homo sapiens 205-210 16941481-0 2006 Desipramine prevents the sustained increase in corticotropin-releasing hormone-like immunoreactivity induced by repeated immobilization stress in the rat central extended amygdala. Desipramine 0-11 corticotropin releasing hormone Rattus norvegicus 47-78 16941481-6 2006 The concomitant administration of desipramine (DMI), a specific noradrenaline uptake inhibitor, fully prevented the RIS-induced increase in CRH expression. Desipramine 34-45 corticotropin releasing hormone Rattus norvegicus 140-143 16941481-6 2006 The concomitant administration of desipramine (DMI), a specific noradrenaline uptake inhibitor, fully prevented the RIS-induced increase in CRH expression. Desipramine 47-50 corticotropin releasing hormone Rattus norvegicus 140-143 16941481-7 2006 RIS also induced an increase of CRH-LI expression in the PVN that was prevented by the concomitant DMI administration. Desipramine 99-102 corticotropin releasing hormone Rattus norvegicus 32-35 16803890-7 2006 The amphiphilic compound desipramine, a frequently employed inhibitor of acid sphingomyelinase (ASMase), blocked PGE(2) production. Desipramine 25-36 sphingomyelin phosphodiesterase 1 Homo sapiens 73-94 16926120-6 2006 Cotreatment of Ad-GFPApoptin-infected cells with the ASMase inhibitor desipramine (2.5 muM) attenuated (30%; P<0.01) apoptin-induced cell death. Desipramine 70-81 sphingomyelin phosphodiesterase 1 Homo sapiens 53-59 16844100-12 2006 The highest potency at Ca(v)2.2 tricyclic antidepressant identified was desipramine. Desipramine 72-83 calcium voltage-gated channel subunit alpha1 B Homo sapiens 23-31 16842762-6 2006 Following an acute injection, all three drugs tested: fluoxetine, desipramine and TCP decreased total BDNF mRNA (exon V) as well as exon IV mRNA, while no significant effect was recorded for exons I and III mRNAs. Desipramine 66-77 brain-derived neurotrophic factor Rattus norvegicus 102-106 16803890-7 2006 The amphiphilic compound desipramine, a frequently employed inhibitor of acid sphingomyelinase (ASMase), blocked PGE(2) production. Desipramine 25-36 sphingomyelin phosphodiesterase 1 Homo sapiens 96-102 16803890-9 2006 Further investigations revealed that desipramine down-regulated acid ceramidase (AC), but not sphingosine kinase, at the protein level. Desipramine 37-48 N-acylsphingosine amidohydrolase 1 Homo sapiens 64-79 16803890-14 2006 In addition, the results of this study warrant careful reconsideration of desipramine as a specific inhibitor for ASMase. Desipramine 74-85 sphingomyelin phosphodiesterase 1 Homo sapiens 114-120 16725121-0 2006 Down-regulation of norepinephrine transporter function induced by chronic administration of desipramine linking to the alteration of sensitivity of local-anesthetics-induced convulsions and the counteraction by co-administration with local anesthetics. Desipramine 92-103 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 19-45 16901483-0 2006 New insights on the use of desipramine as an inhibitor for acid ceramidase. Desipramine 27-38 N-acylsphingosine amidohydrolase 1 Homo sapiens 59-74 16901483-1 2006 Treatment of different cancer cell lines with desipramine induced a time- and dose-dependent downregulation of acid ceramidase. Desipramine 46-57 N-acylsphingosine amidohydrolase 1 Homo sapiens 111-126 16901483-2 2006 Desipramine"s effect on acid ceramidase appeared specific for amphiphilic agents (desipramine, chlorpromazine, and chloroquine) but not other lysomotropic agents such as ammonium chloride and bafilomycin A1, and was not transcriptionally regulated. Desipramine 0-11 N-acylsphingosine amidohydrolase 1 Homo sapiens 24-39 16901483-2 2006 Desipramine"s effect on acid ceramidase appeared specific for amphiphilic agents (desipramine, chlorpromazine, and chloroquine) but not other lysomotropic agents such as ammonium chloride and bafilomycin A1, and was not transcriptionally regulated. Desipramine 82-93 N-acylsphingosine amidohydrolase 1 Homo sapiens 24-39 16901483-3 2006 The cathepsin B/L inhibitor, CA074ME, but not the cathepsin D inhibitor, pepstatin A, blocked desipramine"s effect on acid ceramidase. Desipramine 94-105 cathepsin B Homo sapiens 4-15 16901483-3 2006 The cathepsin B/L inhibitor, CA074ME, but not the cathepsin D inhibitor, pepstatin A, blocked desipramine"s effect on acid ceramidase. Desipramine 94-105 N-acylsphingosine amidohydrolase 1 Homo sapiens 118-133 16901483-4 2006 Desipramine led to a more pronounced downregulation of sphingosine compared to ceramide suggesting acid ceramidase inhibition is important to desipramine"s mechanism of action. Desipramine 0-11 N-acylsphingosine amidohydrolase 1 Homo sapiens 99-114 16901483-4 2006 Desipramine led to a more pronounced downregulation of sphingosine compared to ceramide suggesting acid ceramidase inhibition is important to desipramine"s mechanism of action. Desipramine 142-153 N-acylsphingosine amidohydrolase 1 Homo sapiens 99-114 16730340-3 2006 In mouse hippocampus and cerebral cortex, the effects of repeated treatment with the antidepressants desipramine and fluoxetine or the PDE4 inhibitor rolipram on the expression of PDE4D was compared to that of PDE4A and PDE4B, the other two subtypes expressed in the brain. Desipramine 101-112 phosphodiesterase 4D, cAMP specific Mus musculus 180-185 16278342-5 2005 We showed that 1) after 1 h of reoxygenation, fluorescence had risen and that ROS production was inhibited by desipramine, an inhibitor of sphingomyelinase, an enzyme responsible for ceramide production (126 +/- 7% vs. 48 +/- 12%, P < 0.05); 2) administration of ceramide (N-acetylsphingosine) per se (i.e., in the absence of H/R) induced ROS production (65 +/- 3%), which was inhibited by complex III inhibitor: antimycin A (24 +/- 3%, P < 0.0001), or stigmatellin (31 +/- 2%, P < 0.0001); 3) hypoxia/reoxygenation-induced ROS production was not affected by either ceramide-activated protein kinase inhibitor dimethyl aminopurine or mitochondrial permeability transition inhibitor cyclosporin A but was significantly inhibited by the antiapoptotic protein Bcl-2 (82 +/- 8%, P < 0.05); 4) ceramide-induced ROS production was also inhibited by Bcl-2 (41 +/- 4%, P < 0.0001). Desipramine 110-121 BCL2 apoptosis regulator Homo sapiens 766-771 16845218-4 2006 Desipramine, a classic antidepressant, increased BDNF mRNA level in both examined brain regions. Desipramine 0-11 brain derived neurotrophic factor Homo sapiens 49-53 16460696-5 2006 The most consistent effect, observed with pronoradrenergic drugs (desipramine and reboxetine), was a decrease of GluR3 expression both in P/FC and HI. Desipramine 66-77 glutamate ionotropic receptor AMPA type subunit 3 Rattus norvegicus 113-118 16460696-6 2006 Interestingly, in HI, the same drugs also decreased the editing levels of either the flip (desipramine) or flop (reboxetine) form of GluR3. Desipramine 91-102 glutamate ionotropic receptor AMPA type subunit 3 Rattus norvegicus 133-138 16478969-14 2006 The RFIM (DMI - DMR) was moderately correlated with DMI and ADG (0.37 and -0.38; respectively), suggesting that selecting for low RFI(M) would decrease DMI and increase ADG in this database. Desipramine 10-13 RFI Bos taurus 4-7 16478969-14 2006 The RFIM (DMI - DMR) was moderately correlated with DMI and ADG (0.37 and -0.38; respectively), suggesting that selecting for low RFI(M) would decrease DMI and increase ADG in this database. Desipramine 52-55 RFI Bos taurus 4-7 16478969-14 2006 The RFIM (DMI - DMR) was moderately correlated with DMI and ADG (0.37 and -0.38; respectively), suggesting that selecting for low RFI(M) would decrease DMI and increase ADG in this database. Desipramine 52-55 RFI Bos taurus 4-7 16293366-10 2006 Chronic citalopram and desipramine treatments also increased the somatostatin levels by 83+/-32% and 40+/-6% of basal, respectively. Desipramine 23-34 somatostatin Rattus norvegicus 65-77 16753142-5 2006 Further studies showed that desipramine caused the apoptotic cell death, which could be prevented by neither catalase, reduced-form glutathione (GSH), nor N-acetylcysteine (NAC), without accompanying the disruption of mitochondrial membrane potential within the cells and the release of cytochrome c into the cell cytoplasm. Desipramine 28-39 cytochrome c, somatic Homo sapiens 287-299 16386803-6 2006 We found that propranolol blocked the IL-1beta response to footshock in both the hypothalamus and the spleen, while the noradrenergic reuptake inhibitor desipramine significantly augmented the footshock-induced IL-1beta response in both of these sites. Desipramine 153-164 interleukin 1 beta Rattus norvegicus 211-219 16140280-9 2006 Also, rats treated simultaneously with sertraline plus desipramine exhibited reductions in both SERT and NET binding. Desipramine 55-66 solute carrier family 6 member 4 Rattus norvegicus 96-100 16500030-8 2006 Given to normal rats, fluoxetine increased total brain-derived neurotrophic factor mRNA only in hippocampus, whereas desipramine or phenelzine increased brain-derived neurotrophic factor mRNA in both frontal cortex and hippocampus. Desipramine 117-128 brain-derived neurotrophic factor Rattus norvegicus 153-186 16500030-12 2006 Treatment with desipramine or phenelzine of corticosterone pellet-implanted rats reversed the corticosterone-induced decrease in total brain-derived neurotrophic factor expression in both brain areas; however, fluoxetine reversed the decrease only partially in hippocampus. Desipramine 15-26 brain-derived neurotrophic factor Rattus norvegicus 135-168 16278342-5 2005 We showed that 1) after 1 h of reoxygenation, fluorescence had risen and that ROS production was inhibited by desipramine, an inhibitor of sphingomyelinase, an enzyme responsible for ceramide production (126 +/- 7% vs. 48 +/- 12%, P < 0.05); 2) administration of ceramide (N-acetylsphingosine) per se (i.e., in the absence of H/R) induced ROS production (65 +/- 3%), which was inhibited by complex III inhibitor: antimycin A (24 +/- 3%, P < 0.0001), or stigmatellin (31 +/- 2%, P < 0.0001); 3) hypoxia/reoxygenation-induced ROS production was not affected by either ceramide-activated protein kinase inhibitor dimethyl aminopurine or mitochondrial permeability transition inhibitor cyclosporin A but was significantly inhibited by the antiapoptotic protein Bcl-2 (82 +/- 8%, P < 0.05); 4) ceramide-induced ROS production was also inhibited by Bcl-2 (41 +/- 4%, P < 0.0001). Desipramine 110-121 BCL2 apoptosis regulator Homo sapiens 855-860 16330572-11 2005 11C-mHED accumulation could be inhibited by desipramine, a potent inhibitor of hNET. Desipramine 44-55 solute carrier family 6 member 2 Homo sapiens 79-83 16338282-9 2005 CONCLUSIONS: Low-dose ritonavir (100 mg twice daily) exerts a modest inhibitory effect on the activity of CYP2D6 in extensive metabolizers, as assessed with desipramine as the index substrate. Desipramine 157-168 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 106-112 16143352-7 2005 On the contrary, the effect of DMI on BDNF mRNA was neither dose- nor time-dependent. Desipramine 31-34 brain-derived neurotrophic factor Rattus norvegicus 38-42 16143352-3 2005 Here we looked for the time course of the effect of different doses of two antidepressants, desipramine (DMI) and paroxetine (PAR), which differentially affect monoamine reuptake, on BDNF mRNA expression in hippocampal subfields (CA1, CA3 and dentate gyrus) and levels of AMPAR subunits in total and membrane-enriched extracts from rat hippocampus. Desipramine 92-103 brain-derived neurotrophic factor Rattus norvegicus 183-187 16143352-3 2005 Here we looked for the time course of the effect of different doses of two antidepressants, desipramine (DMI) and paroxetine (PAR), which differentially affect monoamine reuptake, on BDNF mRNA expression in hippocampal subfields (CA1, CA3 and dentate gyrus) and levels of AMPAR subunits in total and membrane-enriched extracts from rat hippocampus. Desipramine 105-108 brain-derived neurotrophic factor Rattus norvegicus 183-187 16359723-6 2005 Furthermore, as with selective serotonin reuptake inhibitors such as fluvoxamine and paroxetine, SIV was also reduced by the serotonin and noradrenaline reuptake inhibitor milnacipran and the metabotropic glutamate receptor 5 antagonist MPEP, while desipramine was without effect. Desipramine 249-260 glutamate metabotropic receptor 5 Rattus norvegicus 192-225 16337960-10 2006 Ceramide plays a crucial role in this process, because prevention of ceramide formation either by myocardial adaptation to ischemia or with desipramine results in the inhibition of eNOS association with caveolin-1 thereby reducing myocardial ischemic reperfusion injury. Desipramine 140-151 nitric oxide synthase 3 Rattus norvegicus 181-185 16337960-10 2006 Ceramide plays a crucial role in this process, because prevention of ceramide formation either by myocardial adaptation to ischemia or with desipramine results in the inhibition of eNOS association with caveolin-1 thereby reducing myocardial ischemic reperfusion injury. Desipramine 140-151 caveolin 1 Rattus norvegicus 203-213 15894065-5 2005 On the other hand, desipramine and mazindol inhibited the increase in wet-dog shakes in ACTH-treated rats. Desipramine 19-30 proopiomelanocortin Canis lupus familiaris 88-92 16111724-5 2005 We show that agents with antidepressant activity, namely the antidepressants fluoxetine and desipramine, the atypical antipsychotic clozapine, and the mood stabilizer lithium increased VGLUT1 mRNA expression in neurons of the cerebral cortex and the hippocampus and in concert enhanced VGLUT1 protein expression in their projection fields. Desipramine 92-103 solute carrier family 17 member 7 Homo sapiens 185-191 16111724-5 2005 We show that agents with antidepressant activity, namely the antidepressants fluoxetine and desipramine, the atypical antipsychotic clozapine, and the mood stabilizer lithium increased VGLUT1 mRNA expression in neurons of the cerebral cortex and the hippocampus and in concert enhanced VGLUT1 protein expression in their projection fields. Desipramine 92-103 solute carrier family 17 member 7 Homo sapiens 286-292 15983795-7 2005 In contrast, complexin II was significantly induced by desipramine and tranylcypromine, but not fluoxetine, in several brain regions. Desipramine 55-66 complexin 2 Rattus norvegicus 13-25 15922009-3 2005 The different CYPs involved in IMI demethylation to the pharmacologically active metabolite desipramine (DES) were CYP2C19 > CYP1A2 > CYP3A4. Desipramine 92-103 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 115-122 15922009-3 2005 The different CYPs involved in IMI demethylation to the pharmacologically active metabolite desipramine (DES) were CYP2C19 > CYP1A2 > CYP3A4. Desipramine 92-103 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 128-134 15922009-3 2005 The different CYPs involved in IMI demethylation to the pharmacologically active metabolite desipramine (DES) were CYP2C19 > CYP1A2 > CYP3A4. Desipramine 92-103 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 15922009-3 2005 The different CYPs involved in IMI demethylation to the pharmacologically active metabolite desipramine (DES) were CYP2C19 > CYP1A2 > CYP3A4. Desipramine 105-108 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 115-122 15922009-3 2005 The different CYPs involved in IMI demethylation to the pharmacologically active metabolite desipramine (DES) were CYP2C19 > CYP1A2 > CYP3A4. Desipramine 105-108 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 128-134 15922009-3 2005 The different CYPs involved in IMI demethylation to the pharmacologically active metabolite desipramine (DES) were CYP2C19 > CYP1A2 > CYP3A4. Desipramine 105-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 140-146 15705795-4 2005 Desipramine and imipramine, inhibitors of acid sphingomyelinase (ASMase), suppressed RGDfV-induced ceramide increase. Desipramine 0-11 sphingomyelin phosphodiesterase 1 Homo sapiens 42-63 15705795-4 2005 Desipramine and imipramine, inhibitors of acid sphingomyelinase (ASMase), suppressed RGDfV-induced ceramide increase. Desipramine 0-11 sphingomyelin phosphodiesterase 1 Homo sapiens 65-71 15705795-5 2005 Importantly, desipramine, imipramine, and a third ASMase inhibitor, SR33557, but not inhibitors of neutral sphingomyelinase, suppressed RGDfV-induced apoptosis, suggesting that ASMase was required for integrin-mediated apoptosis. Desipramine 13-24 sphingomyelin phosphodiesterase 1 Homo sapiens 177-183 15885359-8 2005 Co-incubation with tricyclic antidepressants (desipramine) or serotonin reuptake inhibitors (fluoxetine) attenuated the effect of IFN-alpha on GR or 5-HTR1A. Desipramine 46-57 interferon alpha 1 Homo sapiens 130-139 15885359-8 2005 Co-incubation with tricyclic antidepressants (desipramine) or serotonin reuptake inhibitors (fluoxetine) attenuated the effect of IFN-alpha on GR or 5-HTR1A. Desipramine 46-57 nuclear receptor subfamily 3 group C member 1 Homo sapiens 143-145 15885359-8 2005 Co-incubation with tricyclic antidepressants (desipramine) or serotonin reuptake inhibitors (fluoxetine) attenuated the effect of IFN-alpha on GR or 5-HTR1A. Desipramine 46-57 5-hydroxytryptamine receptor 1A Homo sapiens 151-156 15885359-12 2005 These levels of GR and 5-HTR1A, following IFN-alpha-induced downregulation, recovered after withdrawal of IFN-alpha or addition of desipramine or fluoxetine. Desipramine 131-142 5-hydroxytryptamine receptor 1A Homo sapiens 25-30 15901772-5 2005 Chronic fluoxetine did not influence activin betaA expression, but fluoxetine as well as desipramine did increase Smad2 phosphorylation in the frontal cortex. Desipramine 89-100 SMAD family member 2 Rattus norvegicus 114-119 15814154-1 2005 Previous research has shown that exposure of norepinephrine transporter (NET)-expressing cells to desipramine (DMI) downregulates the norepinephrine transporter, although changes in the several transporter parameters do not demonstrate the same time course. Desipramine 98-109 solute carrier family 6 member 2 Rattus norvegicus 45-71 15814154-1 2005 Previous research has shown that exposure of norepinephrine transporter (NET)-expressing cells to desipramine (DMI) downregulates the norepinephrine transporter, although changes in the several transporter parameters do not demonstrate the same time course. Desipramine 98-109 solute carrier family 6 member 2 Rattus norvegicus 73-76 15814154-1 2005 Previous research has shown that exposure of norepinephrine transporter (NET)-expressing cells to desipramine (DMI) downregulates the norepinephrine transporter, although changes in the several transporter parameters do not demonstrate the same time course. Desipramine 98-109 solute carrier family 6 member 2 Rattus norvegicus 134-160 15814154-1 2005 Previous research has shown that exposure of norepinephrine transporter (NET)-expressing cells to desipramine (DMI) downregulates the norepinephrine transporter, although changes in the several transporter parameters do not demonstrate the same time course. Desipramine 111-114 solute carrier family 6 member 2 Rattus norvegicus 45-71 15814154-1 2005 Previous research has shown that exposure of norepinephrine transporter (NET)-expressing cells to desipramine (DMI) downregulates the norepinephrine transporter, although changes in the several transporter parameters do not demonstrate the same time course. Desipramine 111-114 solute carrier family 6 member 2 Rattus norvegicus 73-76 15814154-1 2005 Previous research has shown that exposure of norepinephrine transporter (NET)-expressing cells to desipramine (DMI) downregulates the norepinephrine transporter, although changes in the several transporter parameters do not demonstrate the same time course. Desipramine 111-114 solute carrier family 6 member 2 Rattus norvegicus 134-160 15814154-3 2005 Recent demonstration of persistent drug retention in cells following desipramine exposures raises the possibility that previous reported changes in the norepinephrine transporter may be partly accountable by residual drug. Desipramine 69-80 solute carrier family 6 member 2 Rattus norvegicus 152-178 16220339-6 2005 Chronic bupropion and desipramine significantly decreased BDNF expression in the dentate gyrus of the hippocampus, while fluoxetine had no effect in any brain region. Desipramine 22-33 brain-derived neurotrophic factor Rattus norvegicus 58-62 21180171-6 2005 Norepinephrine and dopamine could not induce the transporter current while the 5-HT induced current could be specifically inhibited by 5-HTT blocker, desipramine. Desipramine 150-161 solute carrier family 6 member 4 Homo sapiens 135-140 16036225-4 2005 This effect of DMI was not dependent upon activity at the levels of the beta2 receptor, cAMP accumulation or phosphorylation of the transcription factor, cAMP response element binding protein (CREB). Desipramine 15-18 cAMP responsive element binding protein 1 Homo sapiens 193-197 16036225-8 2005 The results suggest a molecular target for DMI that lies downstream of CREB phosphorylation. Desipramine 43-46 cAMP responsive element binding protein 1 Homo sapiens 71-75 15992385-4 2005 Repeated treatment with fluoxetine, paroxetine or desipramine increased VGLUT1 mRNA abundance in frontal, orbital, cingulate and parietal cortices, and regions of the hippocampus. Desipramine 50-61 solute carrier family 17 member 7 Rattus norvegicus 72-78 15946644-4 2005 Here, intrinsic tryptophan fluorescence spectroscopy is used to document the binding of desipramine, a member of the tricyclic antidepressant family, to a well-defined extracellular glutamate binding domain (S1S2) of the GluR2 subunit of the amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor. Desipramine 88-99 glutamate ionotropic receptor AMPA type subunit 2 Homo sapiens 221-226 15726116-1 2005 Previous studies demonstrated that Gsalpha migrates from a Triton X-100 (TTX-100) insoluble membrane domain to a TTX-100 soluble membrane domain in response to chronic treatment with the antidepressants desipramine and fluoxetine. Desipramine 203-214 GNAS complex locus Homo sapiens 35-42 15726116-4 2005 Chronic treatment of C6 cells with two fuctionally and structurally distinct antidepressants, desipramine and fluoxetine, decreased the Gsalpha content of TTX-100 insoluble membrane domains by as much as 60%, while the inactive fluoxetine analog LY368514 had no effect. Desipramine 94-105 GNAS complex locus Homo sapiens 136-143 15617725-7 2005 Based on a possible correlation between the onset of 5HT2C receptor-mediated inhibition and the behavioral effects of desipramine and nefazodone treatment that was described in our previous studies, we suggest that 5HT2C receptor activation may be important for the onset of the behavioral effects of antidepressant treatment. Desipramine 118-129 5-hydroxytryptamine receptor 2C Rattus norvegicus 215-220 15802801-3 2005 When either rat liver microsomes or recombinant CYP2D2 was employed, the preincubation with DMI (0.3 microM) caused a greater inhibition of BF 1""-hydroxylation than the co-incubation did, whereas BF 1""-hydroxylation by rat CYP2D1 was not markedly affected under the same conditions. Desipramine 92-95 cytochrome P450, family 2, subfamily d, polypeptide 2 Rattus norvegicus 48-54 15802801-3 2005 When either rat liver microsomes or recombinant CYP2D2 was employed, the preincubation with DMI (0.3 microM) caused a greater inhibition of BF 1""-hydroxylation than the co-incubation did, whereas BF 1""-hydroxylation by rat CYP2D1 was not markedly affected under the same conditions. Desipramine 92-95 forkhead box G1 Rattus norvegicus 140-144 15802801-3 2005 When either rat liver microsomes or recombinant CYP2D2 was employed, the preincubation with DMI (0.3 microM) caused a greater inhibition of BF 1""-hydroxylation than the co-incubation did, whereas BF 1""-hydroxylation by rat CYP2D1 was not markedly affected under the same conditions. Desipramine 92-95 forkhead box G1 Rattus norvegicus 197-201 15802801-3 2005 When either rat liver microsomes or recombinant CYP2D2 was employed, the preincubation with DMI (0.3 microM) caused a greater inhibition of BF 1""-hydroxylation than the co-incubation did, whereas BF 1""-hydroxylation by rat CYP2D1 was not markedly affected under the same conditions. Desipramine 92-95 cytochrome P450, family 2, subfamily d, polypeptide 1 Rattus norvegicus 225-231 15802801-4 2005 The inhibitory effect of DMI on BF 1""-hydroxylation by human liver microsomal fractions or recombinant CYP2D6 was much lower than that on the hydroxylation by rat liver microsomes or CYP2D2. Desipramine 25-28 forkhead box G1 Rattus norvegicus 32-36 15802801-4 2005 The inhibitory effect of DMI on BF 1""-hydroxylation by human liver microsomal fractions or recombinant CYP2D6 was much lower than that on the hydroxylation by rat liver microsomes or CYP2D2. Desipramine 25-28 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 104-110 15802801-7 2005 Binding experiments employing the recombinant enzymes and [(3)H]-DMI revealed that CYP2D2 and CYP2D6 were the only prominent proteins to which considerable radioactive DMI metabolite(s) bound. Desipramine 65-68 cytochrome P450, family 2, subfamily d, polypeptide 2 Rattus norvegicus 83-89 15802801-7 2005 Binding experiments employing the recombinant enzymes and [(3)H]-DMI revealed that CYP2D2 and CYP2D6 were the only prominent proteins to which considerable radioactive DMI metabolite(s) bound. Desipramine 65-68 cytochrome P450, family 2, subfamily d, polypeptide 3 Rattus norvegicus 94-100 15802801-8 2005 These results indicate that rat CYP2D2 biotransforms DMI into reactive metabolite(s), which covalently bind to CYP2D2, resulting in inactivation of the enzyme. Desipramine 53-56 cytochrome P450, family 2, subfamily d, polypeptide 2 Rattus norvegicus 32-38 15802801-8 2005 These results indicate that rat CYP2D2 biotransforms DMI into reactive metabolite(s), which covalently bind to CYP2D2, resulting in inactivation of the enzyme. Desipramine 53-56 cytochrome P450, family 2, subfamily d, polypeptide 2 Rattus norvegicus 111-117 15802801-9 2005 In contrast, human CYP2D6 may also biotransform DMI into some metabolite(s) that covalently bind to CYP2D6, but that do not inactivate the enzyme. Desipramine 48-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 19-25 15802801-9 2005 In contrast, human CYP2D6 may also biotransform DMI into some metabolite(s) that covalently bind to CYP2D6, but that do not inactivate the enzyme. Desipramine 48-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 100-106 15814105-9 2005 Furthermore, with the exception of "grooming", the effects of desipramine on behaviour of NK1-/- mice could be explained by the effects of this antidepressant on locomotor activity. Desipramine 62-73 tachykinin 1 Mus musculus 90-93 15652409-6 2005 Consistent with expression of a functional SERT, specific uptake of (3)H-serotonin in macrophages was sodium dependent and inhibited by fluoxetine (IC(50) 6.9 nM) and desipramine (IC(50) 32 nM) but not by nisoxetine or reserpine. Desipramine 167-178 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 43-47 15617725-6 2005 However, only 14-day treatment with desipramine or 7-day treatment with nefazodone, but not 7-day treatment with desipramine, decreased 5HT2C receptor-mediated inhibition of accumbal dopamine release. Desipramine 36-47 5-hydroxytryptamine receptor 2C Rattus norvegicus 136-141 15617725-7 2005 Based on a possible correlation between the onset of 5HT2C receptor-mediated inhibition and the behavioral effects of desipramine and nefazodone treatment that was described in our previous studies, we suggest that 5HT2C receptor activation may be important for the onset of the behavioral effects of antidepressant treatment. Desipramine 118-129 5-hydroxytryptamine receptor 2C Rattus norvegicus 53-58 15802801-2 2005 Inhibition was examined under the following two conditions: 1) DMI was co-incubated with BF and NADPH in the reaction mixture containing rat or human liver microsomes or yeast cell microsomes expressing rat CYP2D1, CYP2D2 or human CYP2D6 (co-incubation); 2) DMI was preincubated with NADPH and the same enzyme sources prior to adding the substrate (preincubation). Desipramine 63-66 cytochrome P450, family 2, subfamily d, polypeptide 2 Rattus norvegicus 215-221 15802801-2 2005 Inhibition was examined under the following two conditions: 1) DMI was co-incubated with BF and NADPH in the reaction mixture containing rat or human liver microsomes or yeast cell microsomes expressing rat CYP2D1, CYP2D2 or human CYP2D6 (co-incubation); 2) DMI was preincubated with NADPH and the same enzyme sources prior to adding the substrate (preincubation). Desipramine 63-66 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 231-237 15710233-0 2005 Effects of desipramine treatment on tyrosine hydroxylase gene expression in cultured neuroblastoma cells and rat brain tissue. Desipramine 11-22 tyrosine hydroxylase Rattus norvegicus 36-56 15710233-5 2005 In contrast, 500 nM DMI led to an initial slight increase, followed by a continuous and sustained decrease in TH protein level by up to 53%, from day 3 to day 14. Desipramine 20-23 tyrosine hydroxylase Rattus norvegicus 110-112 15710233-9 2005 These findings indicate that DMI exerts complex, typically opposite and perhaps compensatory, gradually evolving effects on the expression of TH protein (decreases) and its message (increases), possibly in response to increased synaptic availability of NE. Desipramine 29-32 tyrosine hydroxylase Rattus norvegicus 142-144 15878644-0 2005 An antidepressant mechanism of desipramine is to decrease tumor necrosis factor-alpha production culminating in increases in noradrenergic neurotransmission. Desipramine 31-42 tumor necrosis factor Homo sapiens 58-85 15878644-7 2005 Desipramine administration results in a virtual ablation of neuron-derived tumor necrosis factor-alpha (TNF), thus implicating an essential role of TNF in the therapeutic efficacy of this antidepressant. Desipramine 0-11 tumor necrosis factor Homo sapiens 75-102 15878644-7 2005 Desipramine administration results in a virtual ablation of neuron-derived tumor necrosis factor-alpha (TNF), thus implicating an essential role of TNF in the therapeutic efficacy of this antidepressant. Desipramine 0-11 tumor necrosis factor Homo sapiens 104-107 15878644-7 2005 Desipramine administration results in a virtual ablation of neuron-derived tumor necrosis factor-alpha (TNF), thus implicating an essential role of TNF in the therapeutic efficacy of this antidepressant. Desipramine 0-11 tumor necrosis factor Homo sapiens 148-151 15878644-8 2005 Additionally, following chronic administration of desipramine, TNF-regulation of NE release is transformed, from inhibition to facilitation. Desipramine 50-61 tumor necrosis factor Homo sapiens 63-66 15878644-11 2005 Thus, the efficacy of desipramine is due to decreased levels of TNF in the brain induced by this drug, ultimately modifying noradrenergic neurotransmission. Desipramine 22-33 tumor necrosis factor Homo sapiens 64-67 15150531-8 2004 Furthermore, GIRK current responses activated by the cloned A1 adenosine receptor were similarly inhibited by the tricyclic antidepressant desipramine. Desipramine 139-150 potassium inwardly rectifying channel subfamily J member 3 S homeolog Xenopus laevis 13-17 15451381-3 2004 In the present study we performed parallel measurements of BDNF mRNA and protein expression in the frontal cortex and hippocampus of the rat after chronic treatment with electroconvulsive seizures (ECS), lithium, desipramine or escitalopram. Desipramine 213-224 brain-derived neurotrophic factor Rattus norvegicus 59-63 15451381-5 2004 Desipramine moderately increased BDNF mRNA expression in the dentate gyrus but did not change BDNF protein in neither region. Desipramine 0-11 brain-derived neurotrophic factor Rattus norvegicus 33-37 15138445-4 2004 We investigated how different drugs (fluoxetine (FLX), desipramine (DMI), reboxetine (RBX)) affect CREB expression and phosphorylation of Ser(133) in the hippocampus and prefrontal/frontal cortex (PFCX). Desipramine 55-66 cAMP responsive element binding protein 1 Homo sapiens 99-103 15138445-4 2004 We investigated how different drugs (fluoxetine (FLX), desipramine (DMI), reboxetine (RBX)) affect CREB expression and phosphorylation of Ser(133) in the hippocampus and prefrontal/frontal cortex (PFCX). Desipramine 68-71 cAMP responsive element binding protein 1 Homo sapiens 99-103 15150531-10 2004 The GIRK currents induced by ethanol were also attenuated in the presence of desipramine. Desipramine 77-88 potassium inwardly rectifying channel subfamily J member 3 S homeolog Xenopus laevis 4-8 15150531-4 2004 Here, we report the inhibitory effects of various antidepressants: imipramine, desipramine, amitriptyline, nortriptyline, clomipramine, maprotiline, and citalopram, on GIRK channels. Desipramine 79-90 potassium inwardly rectifying channel subfamily J member 3 S homeolog Xenopus laevis 168-172 15337319-4 2004 The obtained results indicate that transcriptional activity of dopamine D2 receptor gene promoter was dose-dependently increased by retinoic acid, forskolin, rolipram and phorbol 12 myristate 13-acetate, as well as by DMI, CIT and MIA. Desipramine 218-221 dopamine receptor D2 Mus musculus 63-83 15262904-6 2004 Blockade of norepinephrine transporter or alpha2-adrenoceptors using desipramine or rauwolscine reduced the losartan-induced shifts in the ED50 values of noradrenaline by 63% and 21%, respectively. Desipramine 69-80 solute carrier family 6 member 2 Rattus norvegicus 12-38 15082752-1 2004 The present study documents a role for brain-derived tumor necrosis factor-alpha (TNF) in the mechanism of action of the antidepressant drug desmethylimipramine (desipramine). Desipramine 141-160 tumor necrosis factor Rattus norvegicus 82-85 15082752-1 2004 The present study documents a role for brain-derived tumor necrosis factor-alpha (TNF) in the mechanism of action of the antidepressant drug desmethylimipramine (desipramine). Desipramine 162-173 tumor necrosis factor Rattus norvegicus 82-85 15082752-3 2004 Chronic desipramine administration transforms TNF-mediated inhibition of NE release to facilitation, dependent upon alpha2-adrenergic receptor activation. Desipramine 8-19 tumor necrosis factor Rattus norvegicus 46-49 15082752-6 2004 To determine whether this transformation is due to desipramine-induced inhibition of TNF bioactivity in the brain, rats were i.c.v. Desipramine 51-62 tumor necrosis factor Rattus norvegicus 85-88 15082752-10 2004 Although simultaneous microinfusion of rrTNF with chronic desipramine administration prevents the transformation induced by desipramine, microinfusion of rrTNF enhances TNF inhibition of NE release. Desipramine 124-135 tumor necrosis factor Rattus norvegicus 41-44 15082752-17 2004 microinfusion of rrTNF with concomitant desipramine administration opposes decreases in neuron-associated TNF levels, required to transform presynaptic sensitivity to TNF, which is necessary for the drug to be efficacious. Desipramine 40-51 tumor necrosis factor Rattus norvegicus 106-109 15219272-5 2004 Fluoxetine and desipramine, both are psychotropics and specific SERT ligands and decreased the affinity of [(123)I]ADAM, while p-chloroamphetamine (PCA), a narcotic, destroyed most of serotonergic neurons, as well as reducing the concentration of serotonin and the number of SERT in the brain as shown by the biodistribution of [(123)I]ADAM. Desipramine 15-26 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 64-68 15261759-1 2004 High concentration of corticosterone (Cort) 0.2 mM was incubated with PC12 cells to simulate the lesion state of brain neurons in depressive illness, it was found that the inulin-type oligosaccharides extracted from Morinda officinalis, inulin-type hexasaccharide (IHS) at the doses of 0.625, 1.25 microM or desipramine (DIM) 0.25, 1 microM protected the PC12 cells from the lesion induced by Cort. Desipramine 308-319 cortistatin Rattus norvegicus 38-42 15249684-5 2004 We also demonstrate that the loss of forebrain BDNF attenuates the actions of desipramine, an antidepressant, in the forced swim test, suggesting the involvement of BDNF in antidepressant efficacy. Desipramine 78-89 brain derived neurotrophic factor Mus musculus 47-51 15249684-5 2004 We also demonstrate that the loss of forebrain BDNF attenuates the actions of desipramine, an antidepressant, in the forced swim test, suggesting the involvement of BDNF in antidepressant efficacy. Desipramine 78-89 brain derived neurotrophic factor Mus musculus 165-169 15219272-5 2004 Fluoxetine and desipramine, both are psychotropics and specific SERT ligands and decreased the affinity of [(123)I]ADAM, while p-chloroamphetamine (PCA), a narcotic, destroyed most of serotonergic neurons, as well as reducing the concentration of serotonin and the number of SERT in the brain as shown by the biodistribution of [(123)I]ADAM. Desipramine 15-26 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 275-279 15219272-8 2004 In addition, the inhibition effect on binding ability of [(123)I]ADAM to SERT by the psychotropics and the narcotic was found to have the order of: PCA > fluoxetine > desipramine. Desipramine 173-184 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 73-77 14735130-6 2004 It was found that imipramine, amitryptyline, desipramine, fluoxetine, and mianserin, present in the culture medium for 5 days, in a concentration-dependent manner inhibited basal hCRH gene promoter activity in undifferentiated Neuro-2A cells, while other drugs under study (citalopram, tianeptine, moclobemide, venlafaxine, reboxetine, mirtazapine, and milnacipram) were inactive. Desipramine 45-56 corticotropin releasing hormone Homo sapiens 179-183 15110811-7 2004 The cardioprotective abilities of PC were abolished with desipramine, which also downregulated a PC-mediated increase in antiapoptotic protein Bcl-2. Desipramine 57-68 BCL2 apoptosis regulator Homo sapiens 143-148 15115913-0 2004 Impact of CYP2D6 intermediate metabolizer alleles on single-dose desipramine pharmacokinetics. Desipramine 65-76 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 10-16 15115913-1 2004 This study utilized cytochrome P450 2D6 (CYP2D6) genotypes to explain variability of desipramine pharmacokinetics in a cohort of non-poor metabolizer individuals. Desipramine 85-96 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 20-39 15115913-1 2004 This study utilized cytochrome P450 2D6 (CYP2D6) genotypes to explain variability of desipramine pharmacokinetics in a cohort of non-poor metabolizer individuals. Desipramine 85-96 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 41-47 15132707-1 2004 Interactions of two amphiphilic antidepressant drugs, imipramine and desipramine hydrochlorides, with the blood protein human serum albumin (HSA) were investigated to gain an understanding of the effects of drug molecular structure on the complex formation of drug-protein molecules. Desipramine 69-95 albumin Homo sapiens 126-139 16680870-1 2004 Clinically, cimetidine therapy impairs the clearance of various drugs metabolized by CYP2D6, such as desipramine and sparteine. Desipramine 101-112 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 85-91 15033385-5 2004 These results indicate that the blockade of 5-HT(1B) rather than 5-HT(1A) receptors may facilitate the anti-immobility effect of imipramine, desipramine or moclobemide in the forced swimming test. Desipramine 141-152 5-hydroxytryptamine receptor 1B Rattus norvegicus 44-51 14985439-5 2004 Here we establish that both subchronic and chronic desipramine (DMI) treatments upregulate hypothalamic ICER expression in wild-type mice. Desipramine 51-62 cAMP responsive element modulator Mus musculus 104-108 14730412-0 2004 Effect of the novel anxiolytic drug deramciclane on cytochrome P(450) 2D6 activity as measured by desipramine pharmacokinetics. Desipramine 98-109 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 52-73 14730412-1 2004 BACKGROUND: In vitro findings have indicated that the novel anxiolytic drug, deramciclane, is an inhibitor of the cytochrome P(450) (CYP) 2D6 enzyme and co-administration of deramciclane and the CYP2D6 probe drug desipramine is possible in clinical practice. Desipramine 213-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 114-131 14730412-1 2004 BACKGROUND: In vitro findings have indicated that the novel anxiolytic drug, deramciclane, is an inhibitor of the cytochrome P(450) (CYP) 2D6 enzyme and co-administration of deramciclane and the CYP2D6 probe drug desipramine is possible in clinical practice. Desipramine 213-224 cytochrome P450 family 4 subfamily F member 3 Homo sapiens 133-136 14960314-0 2004 Interactions of acid sphingomyelinase and lipid bilayers in the presence of the tricyclic antidepressant desipramine. Desipramine 105-116 sphingomyelin phosphodiesterase 1 Homo sapiens 16-37 14960314-1 2004 The tricyclic antidepressant desipramine causes a decrease in cellular acid sphingomyelinase (A-SMase, EC 3.1.4.12) activity when added to culture medium of human fibroblasts. Desipramine 29-40 sphingomyelin phosphodiesterase 1 Homo sapiens 71-92 14960314-1 2004 The tricyclic antidepressant desipramine causes a decrease in cellular acid sphingomyelinase (A-SMase, EC 3.1.4.12) activity when added to culture medium of human fibroblasts. Desipramine 29-40 sphingomyelin phosphodiesterase 1 Homo sapiens 94-101 14960314-6 2004 Our findings indicate that desipramine interferes with the binding of A-SMase to the lipid bilayers and thereby displaces the enzyme from its membrane-bound substrate. Desipramine 27-38 sphingomyelin phosphodiesterase 1 Homo sapiens 70-77 14960314-8 2004 We hypothesize that the displacement of the glycoprotein A-SMase from the inner membranes of late endosomes and lysosomes by desipramine renders it susceptible to proteolytic cleavage by lysosomal proteases. Desipramine 125-136 sphingomyelin phosphodiesterase 1 Homo sapiens 57-64 14985439-5 2004 Here we establish that both subchronic and chronic desipramine (DMI) treatments upregulate hypothalamic ICER expression in wild-type mice. Desipramine 64-67 cAMP responsive element modulator Mus musculus 104-108 14985439-6 2004 Behavioral responses to DMI in the forced swim and tail suspension tests are unchanged in mice lacking ICER. Desipramine 24-27 cAMP responsive element modulator Mus musculus 103-107 14610241-5 2004 Based on the potential of interaction from the in vitro experiments, drug interaction studies in healthy subjects were conducted using probe substrates for CYP2D6 (desipramine) in CYP2D6 extensive metabolizer subjects and CYP3A (midazolam) in CYP2D6 poor metabolizer subjects. Desipramine 164-175 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 156-162 14985439-7 2004 However, the ability of DMI to suppress an acute corticosterone response after swim stress is compromised in ICER-deficient mice, suggesting that increased hypothalamic ICER mRNA after DMI treatment may be required for suppression of corticosterone release. Desipramine 185-188 cAMP responsive element modulator Mus musculus 109-113 14610241-5 2004 Based on the potential of interaction from the in vitro experiments, drug interaction studies in healthy subjects were conducted using probe substrates for CYP2D6 (desipramine) in CYP2D6 extensive metabolizer subjects and CYP3A (midazolam) in CYP2D6 poor metabolizer subjects. Desipramine 164-175 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 180-186 14985439-7 2004 However, the ability of DMI to suppress an acute corticosterone response after swim stress is compromised in ICER-deficient mice, suggesting that increased hypothalamic ICER mRNA after DMI treatment may be required for suppression of corticosterone release. Desipramine 185-188 cAMP responsive element modulator Mus musculus 169-173 14610241-5 2004 Based on the potential of interaction from the in vitro experiments, drug interaction studies in healthy subjects were conducted using probe substrates for CYP2D6 (desipramine) in CYP2D6 extensive metabolizer subjects and CYP3A (midazolam) in CYP2D6 poor metabolizer subjects. Desipramine 164-175 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 180-186 14985439-11 2004 These data indicate that ICER is required for DMI to reduce stress-induced corticosterone release through regulation of hypothalamic CRF expression, revealing a novel role for ICER in antidepressant regulation of the hypothalamic-pituitary adrenal axis. Desipramine 46-49 cAMP responsive element modulator Mus musculus 25-29 14985439-11 2004 These data indicate that ICER is required for DMI to reduce stress-induced corticosterone release through regulation of hypothalamic CRF expression, revealing a novel role for ICER in antidepressant regulation of the hypothalamic-pituitary adrenal axis. Desipramine 46-49 cAMP responsive element modulator Mus musculus 176-180 14515337-9 2003 After inhibition of the DA and NA transporter, by perfusing 100 microM desmethyl-imipramine into the mPFC, clonidine perfusion into the LC reduced extracellular NA and DA in the mPFC by about 50%. Desipramine 71-91 complement factor properdin Mus musculus 101-105 14716709-6 2004 A study on the relative influence of CYP1A2 and 3A4 only revealed minor changes in the presence of desipramine. Desipramine 99-110 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 37-43 14640711-5 2003 The system was used both to yield the Michaelis constant (K(m)) of the P450 biotransformation of imipramine into desipramine and to determine the IC50 value of a chemical inhibitor (tranylcypromine) for this CYP2C19-mediated reaction. Desipramine 113-124 cytochrome P450 family 2 subfamily B member 6 Homo sapiens 71-75 14640711-5 2003 The system was used both to yield the Michaelis constant (K(m)) of the P450 biotransformation of imipramine into desipramine and to determine the IC50 value of a chemical inhibitor (tranylcypromine) for this CYP2C19-mediated reaction. Desipramine 113-124 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 208-215 14515337-9 2003 After inhibition of the DA and NA transporter, by perfusing 100 microM desmethyl-imipramine into the mPFC, clonidine perfusion into the LC reduced extracellular NA and DA in the mPFC by about 50%. Desipramine 71-91 complement factor properdin Mus musculus 178-182 14623141-6 2003 This result suggests that MAP-4 and drebrin may be involved in the antidepressant like effects of desipramine and fluoxetine. Desipramine 98-109 microtubule-associated protein 4 Rattus norvegicus 26-31 14623141-6 2003 This result suggests that MAP-4 and drebrin may be involved in the antidepressant like effects of desipramine and fluoxetine. Desipramine 98-109 drebrin 1 Rattus norvegicus 36-43 14623141-5 2003 cDNA homology analysis revealed that desipramine up-regulated the expression of microtubule-associated protein 4 (MAP-4) mRNA and fluoxetine up-regulated the expression of drebrin A mRNA in the rat hippocampus compared with the chronically stressed group. Desipramine 37-48 microtubule-associated protein 4 Rattus norvegicus 80-112 14623141-5 2003 cDNA homology analysis revealed that desipramine up-regulated the expression of microtubule-associated protein 4 (MAP-4) mRNA and fluoxetine up-regulated the expression of drebrin A mRNA in the rat hippocampus compared with the chronically stressed group. Desipramine 37-48 microtubule-associated protein 4 Rattus norvegicus 114-119 12837768-0 2003 chi-Conopeptide MrIA partially overlaps desipramine and cocaine binding sites on the human norepinephrine transporter. Desipramine 40-51 solute carrier family 6 member 2 Homo sapiens 91-117 12750385-12 2003 Desipramine (a sphingomyelinase inhibitor) prevented the increase in ceramide and inhibited apoptosis after M-CSF deprivation. Desipramine 0-11 colony stimulating factor 1 Homo sapiens 108-113 14499328-5 2003 Pretreatment with the NET ligand, desipramine, decreased the specific binding of (S,S)-[(11)C]MeNER. Desipramine 34-45 solute carrier family 6 member 2 Homo sapiens 22-25 14555335-9 2003 The secondary amines nisoxetine (NIS), and especially, desipramine (DES) and nortriptyline (NOR), also inhibited CYP2C11 and generated MI complexes with microsomal CYP. Desipramine 55-66 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 113-120 14555335-9 2003 The secondary amines nisoxetine (NIS), and especially, desipramine (DES) and nortriptyline (NOR), also inhibited CYP2C11 and generated MI complexes with microsomal CYP. Desipramine 55-66 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 113-116 14555335-9 2003 The secondary amines nisoxetine (NIS), and especially, desipramine (DES) and nortriptyline (NOR), also inhibited CYP2C11 and generated MI complexes with microsomal CYP. Desipramine 68-71 cytochrome P450, subfamily 2, polypeptide 11 Rattus norvegicus 113-120 14555335-9 2003 The secondary amines nisoxetine (NIS), and especially, desipramine (DES) and nortriptyline (NOR), also inhibited CYP2C11 and generated MI complexes with microsomal CYP. Desipramine 68-71 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 113-116 12782200-6 2003 Acute treatment of both wild-type and NET-KO mice with desipramine significantly decreased the TNF-alpha response and significantly increased interleukin-10 production, indicating the role of an intact noradrenaline transporter in anti-inflammatory responses. Desipramine 55-66 tumor necrosis factor Mus musculus 95-104 12849931-4 2003 Under the conditions of the assays, [(3)H]paroxetine binding in the LC was specific for the SERT, based on the rank order of affinity of compounds for inhibiting [(3)H]paroxetine binding in the LC, i.e. citalopram > imipramine > desipramine > mazindol. Desipramine 235-246 solute carrier family 6 member 4 Homo sapiens 92-96 12782200-6 2003 Acute treatment of both wild-type and NET-KO mice with desipramine significantly decreased the TNF-alpha response and significantly increased interleukin-10 production, indicating the role of an intact noradrenaline transporter in anti-inflammatory responses. Desipramine 55-66 interleukin 10 Mus musculus 142-156 12757900-3 2003 Desipramine, clomiplamine, fluoxetine, milnacipran and clorgyline all induced rapid and sustained translocation of GR into the nucleus of human lymphocytes. Desipramine 0-11 nuclear receptor subfamily 3 group C member 1 Homo sapiens 115-117 12700702-0 2003 Impact of maternal deprivation on brain corticotropin-releasing hormone circuits: prevention of CRH receptor-2 mRNA changes by desipramine treatment. Desipramine 127-138 corticotropin releasing hormone Homo sapiens 40-71 12694379-4 2003 We first showed that the nonspecific adrenergic agonist noradrenaline, the alpha- or beta-adrenergic agonists phenylephrine or dobutamine, or the noradrenergic uptake inhibitor desipramine, all significantly stimulated ACTH secretion by freely moving, nonanaesthetized rats. Desipramine 177-188 proopiomelanocortin Homo sapiens 219-223 12700702-0 2003 Impact of maternal deprivation on brain corticotropin-releasing hormone circuits: prevention of CRH receptor-2 mRNA changes by desipramine treatment. Desipramine 127-138 corticotropin releasing hormone Homo sapiens 96-99 12700702-11 2003 One week of desipramine (DES) administration preceding the maternal deprivation event prevented all the deprivation-induced changes in CRHr2 mRNA, regardless of the direction of the original change. Desipramine 12-23 corticotropin releasing hormone receptor 2 Homo sapiens 135-140 12700702-11 2003 One week of desipramine (DES) administration preceding the maternal deprivation event prevented all the deprivation-induced changes in CRHr2 mRNA, regardless of the direction of the original change. Desipramine 25-28 corticotropin releasing hormone receptor 2 Homo sapiens 135-140 12657510-5 2003 The SERT inhibitors desipramine and fluoxetine also inhibited (3)H-MPP(+) specific uptake (with IC(50)s of 189 and 0.92 microM, respectively). Desipramine 20-31 solute carrier family 6 member 4 Homo sapiens 4-8 12751629-6 2003 The hepatic metabolism of lidocaine was inhibited by a CYP2D6 substrate desipramine, not by a CYP3A4 inhibitor ketoconazole. Desipramine 72-83 cytochrome P450, family 2, subfamily d, polypeptide 3 Rattus norvegicus 55-61 12499385-7 2003 We show that ASP(+) (4-(4-(dimethylamino)styrl)-N-methylpyridinium) has micromolar potency for the human norepinephrine transporter, that ASP(+) accumulation is Na(+)-, Cl(-)-, cocaine-, and desipramine-sensitive and temperature-dependent, and that ASP(+) competes with norepinephrine uptake. Desipramine 191-202 solute carrier family 6 member 2 Homo sapiens 105-131 12644358-0 2003 Regulation of GAP-43 expression by chronic desipramine treatment in rat cultured hippocampal cells. Desipramine 43-54 growth associated protein 43 Rattus norvegicus 14-20 12644358-7 2003 In situ hybridization revealed that desipramine increased GAP-43 gene expression in dentate gyrus but not other brain regions. Desipramine 36-47 growth associated protein 43 Rattus norvegicus 58-64 12644358-8 2003 Northern and immunoblotting analysis revealed that desipramine increased GAP-43 mRNA and protein levels. Desipramine 51-62 growth associated protein 43 Rattus norvegicus 73-79 12493574-5 2003 In the presence of P(6), or tricyclic antidepressant desipramine (DIM), the apoptosis induced by Cort in the three measurements above was significantly inhibited. Desipramine 53-64 cortistatin Rattus norvegicus 97-101 12493574-5 2003 In the presence of P(6), or tricyclic antidepressant desipramine (DIM), the apoptosis induced by Cort in the three measurements above was significantly inhibited. Desipramine 66-69 cortistatin Rattus norvegicus 97-101 12464448-11 2002 In contrast, 5-HTT-/- mice retained sensitivity to the anti-immobility effects of the norepinephrine reuptake inhibitor, desipramine (20 mg/kg), and the mixed serotonin/norepinephrine reuptake inhibitor, imipramine (25 mg/kg). Desipramine 121-132 solute carrier family 6 (neurotransmitter transporter, serotonin), member 4 Mus musculus 13-18 12644358-10 2003 CONCLUSIONS: Because GAP-43 regulates growth of axons and modulates the formation of new connections, our findings suggest that desipramine may have an effect on neuronal plasticity in the central nervous system. Desipramine 128-139 growth associated protein 43 Rattus norvegicus 21-27 12464451-5 2002 Finally, rolipram potently augmented GR enhancement by the antidepressant, desipramine. Desipramine 75-86 nuclear receptor subfamily 3, group C, member 1 Mus musculus 37-39 12613867-4 2003 During the hot period, the THI 2 d earlier and mean air temperature 2 d earlier had the greatest impact on milk yield and DMI, respectively. Desipramine 122-125 alcohol dehydrogenase iron containing 1 Bos taurus 11-14 12228186-8 2002 The secondary amine TCAs, nortriptyline and desipramine, however, showed minimal inhibition of CYP2C19 (estimated IC(50) of 600 and 685 micro M, respectively). Desipramine 44-55 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 95-102 12527472-2 2002 Here we studied the acute and chronic effect of the antidepressants desipramine and paroxetine, which differentially affect monoamine reuptake, on the expression of the AMPAR subunits GluR1 and GluR2/3, analyzed by Western blot, both in total and in membrane-enriched extracts from rat hippocampus. Desipramine 68-79 glutamate ionotropic receptor AMPA type subunit 1 Rattus norvegicus 184-189 12527472-2 2002 Here we studied the acute and chronic effect of the antidepressants desipramine and paroxetine, which differentially affect monoamine reuptake, on the expression of the AMPAR subunits GluR1 and GluR2/3, analyzed by Western blot, both in total and in membrane-enriched extracts from rat hippocampus. Desipramine 68-79 glutamate ionotropic receptor AMPA type subunit 2 Rattus norvegicus 194-199 12527474-2 2002 This study was designed to assess the in vivo indirect activation of adrenoceptors or 5-HT receptors by the reuptake blocker desipramine or fluoxetine on the cellular distribution of GRK 2/3 in rat brain. Desipramine 125-136 G protein-coupled receptor kinase 2 Rattus norvegicus 183-190 12527474-11 2002 The results indicate that the in vivo activation of adrenoceptors by desipramine is associated with a time-dependent modulation of membrane-associated GRK 2/3 (i.e. an acute increase in the kinase content which is followed by a return to the basal expression upon repeated treatment). Desipramine 69-80 G protein-coupled receptor kinase 2 Rattus norvegicus 151-158 12412819-0 2002 Effect of terbinafine on the pharmacokinetics and pharmacodynamics of desipramine in healthy volunteers identified as cytochrome P450 2D6 (CYP2D6) extensive metabolizers. Desipramine 70-81 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 118-137 12412819-0 2002 Effect of terbinafine on the pharmacokinetics and pharmacodynamics of desipramine in healthy volunteers identified as cytochrome P450 2D6 (CYP2D6) extensive metabolizers. Desipramine 70-81 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 139-145 12412819-1 2002 Terbinafine-CYP2D6 inhibition was evaluated by assessing 48-hour concentration-time profiles of the tricyclic antidepressant desipramine in 12 healthy volunteers identified as extensive cytochrome P450 2D6 (CYP2D6) metabolizers by genotyping and phenotyping. Desipramine 125-136 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 12-18 12084721-6 2002 Heparin or heparan sulfate displaces FGF3 from binding sites on the cell surface inhibiting the growth of DMI cells and reverts the transformed phenotype (). Desipramine 106-109 fibroblast growth factor 3 Mus musculus 37-41 12230948-0 2002 Caspase 3 gene expression and [Ca2+]i homeostasis underlying desipramine-induced C6 glioma cell apoptosis. Desipramine 61-72 caspase 3 Rattus norvegicus 0-9 12230948-1 2002 AIM: To study desipramine (Des)-induced apoptosis and regulation of caspase 3 gene expression and [Ca2+]i homeostasis in rat glioma C6 cells. Desipramine 14-25 caspase 3 Rattus norvegicus 68-77 12230948-1 2002 AIM: To study desipramine (Des)-induced apoptosis and regulation of caspase 3 gene expression and [Ca2+]i homeostasis in rat glioma C6 cells. Desipramine 27-30 caspase 3 Rattus norvegicus 68-77 12230948-6 2002 Apoptotic DNA breaks were further confirmed by a typical "DNA ladder" on agarose gel electrophoresis after exposure to Des 40 micromol/L for 24 h. Meanwhile, expression of caspase 3 gene was observed following Des 20 micromol/L treatment. Desipramine 119-122 caspase 3 Rattus norvegicus 172-181 12162636-8 2002 In animals challenged with clonidine, there was a significant age by treatment interaction effect for the prepubertal group pretreated with high doses of desipramine (less growth hormone secretion) but not for the peri- or postpubertal groups. Desipramine 154-165 gonadotropin releasing hormone receptor Rattus norvegicus 172-186 12070759-0 2002 Inhibition of transport function and desipramine binding at the human noradrenaline transporter by N-ethylmaleimide and protection by substrate analogs. Desipramine 37-48 solute carrier family 6 member 2 Homo sapiens 70-95 12137927-0 2002 Regulation of norepinephrine transporter abundance by catecholamines and desipramine in vivo. Desipramine 73-84 solute carrier family 6 (neurotransmitter transporter, noradrenalin), member 2 Mus musculus 14-40 12144933-0 2002 Inhibition of SK3 channels in the TE671 human medulloblastoma cell line by desipramine and imipramine. Desipramine 75-86 potassium calcium-activated channel subfamily N member 3 Homo sapiens 14-17 12144933-4 2002 The interaction of desipramine with the selective SK3 blocker, apamin, was studied in more detail. Desipramine 19-30 potassium calcium-activated channel subfamily N member 3 Homo sapiens 50-53 12091475-0 2002 Effects of desipramine treatment on norepinephrine transporter gene expression in the cultured SK-N-BE(2)M17 cells and rat brain tissue. Desipramine 11-22 solute carrier family 6 member 2 Rattus norvegicus 36-62 12091475-1 2002 The antidepressant desipramine (DMI) is a selective inhibitor of norepinephrine (NE) transport that down-regulates the norepinephrine transporter (NET) protein in a concentration- and time-dependent manner in vitro. Desipramine 19-30 solute carrier family 6 member 2 Rattus norvegicus 119-145 12091475-1 2002 The antidepressant desipramine (DMI) is a selective inhibitor of norepinephrine (NE) transport that down-regulates the norepinephrine transporter (NET) protein in a concentration- and time-dependent manner in vitro. Desipramine 19-30 solute carrier family 6 member 2 Rattus norvegicus 147-150 12091475-1 2002 The antidepressant desipramine (DMI) is a selective inhibitor of norepinephrine (NE) transport that down-regulates the norepinephrine transporter (NET) protein in a concentration- and time-dependent manner in vitro. Desipramine 32-35 solute carrier family 6 member 2 Rattus norvegicus 119-145 12091475-1 2002 The antidepressant desipramine (DMI) is a selective inhibitor of norepinephrine (NE) transport that down-regulates the norepinephrine transporter (NET) protein in a concentration- and time-dependent manner in vitro. Desipramine 32-35 solute carrier family 6 member 2 Rattus norvegicus 147-150 12091475-2 2002 In this study, possible regulatory effects of DMI on NET mRNA and protein levels were investigated with the NET-expressing SK-N-BE(2)M17 cell line and rat brain tissue. Desipramine 46-49 solute carrier family 6 member 2 Rattus norvegicus 53-56 12091475-3 2002 Northern blot analysis showed that incubation of the cultured cells with DMI (5-500 nm) for 3 days reduced levels of NET mRNA in both its 5.8-kb (by up to 58%) and 3.6-kb forms (to 68%), whereas incubation for 14 days increased both levels (to 40% and 100%) in a concentration-dependent manner. Desipramine 73-76 solute carrier family 6 member 2 Rattus norvegicus 117-120 12091475-4 2002 In contrast, NET protein levels decreased after 3-14 days of exposure of the cells to DMI, as determined by western blotting. Desipramine 86-89 solute carrier family 6 member 2 Rattus norvegicus 13-16 12091475-6 2002 Thus, in situ hybridization demonstrated initially decreased, and later increased, NET mRNA levels in locus coeruleus (LC) tissue of rats treated with DMI; whereas NET protein levels in the LC were reduced after 14 days, but unchanged after three daily DMI treatments. Desipramine 151-154 solute carrier family 6 member 2 Rattus norvegicus 83-86 12091475-6 2002 Thus, in situ hybridization demonstrated initially decreased, and later increased, NET mRNA levels in locus coeruleus (LC) tissue of rats treated with DMI; whereas NET protein levels in the LC were reduced after 14 days, but unchanged after three daily DMI treatments. Desipramine 151-154 solute carrier family 6 member 2 Rattus norvegicus 164-167 12091475-7 2002 Thus, DMI had similar effects on NET expression in vitro and in vivo, with opposite changes in NET mRNA and protein levels, suggesting that the regulatory mechanisms involved are complex and non-congruent. Desipramine 6-9 solute carrier family 6 member 2 Rattus norvegicus 33-36 12070759-6 2002 These compounds as well as methamphetamine, methcathinone, and desipramine also protected the hNET from NEM inactivation of [(3)H]desipramine binding. Desipramine 63-74 solute carrier family 6 member 2 Homo sapiens 94-98 12070759-6 2002 These compounds as well as methamphetamine, methcathinone, and desipramine also protected the hNET from NEM inactivation of [(3)H]desipramine binding. Desipramine 130-141 solute carrier family 6 member 2 Homo sapiens 94-98 11996893-5 2002 Likewise, attenuation of antidepressant-induced NF-kappa B activity by elevation of the intracellular cAMP concentration or by retroviral driven expression of the non-degradable superrepressor I kappa B alpha S32A/S36A demonstrated that the elevation of NF-kappa B activity by amitriptyline, desipramine and fluoxetine is not an integral part of the apoptotic signaling cascade triggered by these compounds. Desipramine 292-303 NFKB inhibitor alpha Rattus norvegicus 193-208 11943827-6 2002 Furthermore, the ability of DMI to suppress an acute corticosterone response after swim stress is maintained in CREB-deficient mice. Desipramine 28-31 cAMP responsive element binding protein 1 Mus musculus 112-116 11943827-7 2002 However, upregulation of a molecular target of CREB, BDNF, is abolished in the CREB-deficient mice after chronic administration of DMI. Desipramine 131-134 cAMP responsive element binding protein 1 Mus musculus 47-51 11943827-7 2002 However, upregulation of a molecular target of CREB, BDNF, is abolished in the CREB-deficient mice after chronic administration of DMI. Desipramine 131-134 brain derived neurotrophic factor Mus musculus 53-57 11943827-7 2002 However, upregulation of a molecular target of CREB, BDNF, is abolished in the CREB-deficient mice after chronic administration of DMI. Desipramine 131-134 cAMP responsive element binding protein 1 Mus musculus 79-83 12020742-5 2002 The objective of the present experiment was to determine whether two commonly used antidepressants (desipramine and fluoxetine) were effective in ameliorating IFN-alpha-induced anhedonia in rats. Desipramine 100-111 interferon alpha 1 Homo sapiens 159-168 11704655-2 2001 Previous data demonstrate that the tricyclic antidepressant, desipramine, induces glucocorticoid receptor (GR) translocation from the cytoplasm to the nucleus in L929 cells and increases dexamethasone-induced GR-mediated gene transcription in L929 cells stably transfected with the mouse mammary tumour virus-chloramphenicol acetyltransferase (MMTV-CAT) reporter gene (LMCAT cells) (Pariante et al., 1997). Desipramine 61-72 nuclear receptor subfamily 3, group C, member 1 Mus musculus 82-105 11916794-0 2002 Tramadol inhibits norepinephrine transporter function at desipramine-binding sites in cultured bovine adrenal medullary cells. Desipramine 57-68 solute carrier family 6 member 2 Bos taurus 18-44 11916794-9 2002 These findings indicate that tramadol competitively inhibits NET function at desipramine-binding sites. Desipramine 77-88 solute carrier family 6 member 2 Bos taurus 61-64 11916794-10 2002 IMPLICATIONS: Tramadol competitively inhibits norepinephrine transporter function at desipramine-binding sites in the adrenal medullary cells and probably the noradrenergic neurons of the descending inhibitory system. Desipramine 85-96 solute carrier family 6 member 2 Bos taurus 46-72 11738543-4 2001 Patients on desipramine showed more diffuse rCBF reductions in frontal and temporal regions, more so in the left side. Desipramine 12-23 CCAAT/enhancer binding protein zeta Rattus norvegicus 44-48 11704655-2 2001 Previous data demonstrate that the tricyclic antidepressant, desipramine, induces glucocorticoid receptor (GR) translocation from the cytoplasm to the nucleus in L929 cells and increases dexamethasone-induced GR-mediated gene transcription in L929 cells stably transfected with the mouse mammary tumour virus-chloramphenicol acetyltransferase (MMTV-CAT) reporter gene (LMCAT cells) (Pariante et al., 1997). Desipramine 61-72 nuclear receptor subfamily 3, group C, member 1 Mus musculus 107-109 11704655-2 2001 Previous data demonstrate that the tricyclic antidepressant, desipramine, induces glucocorticoid receptor (GR) translocation from the cytoplasm to the nucleus in L929 cells and increases dexamethasone-induced GR-mediated gene transcription in L929 cells stably transfected with the mouse mammary tumour virus-chloramphenicol acetyltransferase (MMTV-CAT) reporter gene (LMCAT cells) (Pariante et al., 1997). Desipramine 61-72 nuclear receptor subfamily 3, group C, member 1 Mus musculus 209-211 11704655-7 2001 Twenty-four hours coincubation of cells with desipramine, clomipramine or paroxetine, also enhanced GR function in the presence of cortisol, but not of corticosterone. Desipramine 45-56 nuclear receptor subfamily 3, group C, member 1 Mus musculus 100-102 11556901-0 2001 Chronic desipramine treatment selectively potentiates somatostatin-induced dopamine release in the nucleus accumbens. Desipramine 8-19 somatostatin Rattus norvegicus 54-66 11504807-12 2001 The immobility of 5-HT(1B) receptor mutant mice was decreased similarly by desipramine (5.0--20.0 mg/kg i.p.). Desipramine 75-86 5-hydroxytryptamine (serotonin) receptor 1B Mus musculus 18-35 11838318-6 2001 Desipramine (10 mg/kg), a tricycle antidepressant, or fluoxetine (10 mg/kg), a serotonin reuptake inhibitor, significantly reversed the LPS-induced increase in immobility time. Desipramine 0-11 toll-like receptor 4 Mus musculus 136-139 11556901-4 2001 Chronic desipramine treatment resulted in an exaggerated somatostatin-induced increase of dopamine levels, specifically in the nucleus accumbens (3542% compared with 564% of basal in the striatum), whereas acute desipramine treatment had no effect (582% of basal) compared with saline treated rats. Desipramine 8-19 somatostatin Rattus norvegicus 57-69 11415943-9 2001 In addition, TNF-alpha-induced ROS production was inhibited by the acidic sphingomyelinase inhibitor desipramine (5 microM; -80%, n = 4, P < 0.01) and totally blocked by the ceramide-activated protein kinase (CAPK) inhibitor dimethylaminopurine (1 mM; n = 6, P < 0.05). Desipramine 101-112 tumor necrosis factor Homo sapiens 13-22 11454927-7 2001 In addition, desipramine significantly reduced immobility in the Dbh(-/-) mice following pretreatment with the synthetic NE precursor L-threo-3,4-dihydroxyphenylserine, but not saline. Desipramine 13-24 dopamine beta hydroxylase Mus musculus 65-68 11417443-0 2001 Duplication of CYP2D6 predicts high clearance of desipramine but high clearance does not predict duplication of CYP2D6. Desipramine 49-60 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 15-21 11353802-1 2001 Previous studies have demonstrated that chronic treatment of C6 glioma cells with the antidepressants desipramine and fluoxetine increases the Triton X-100 solubility of the G protein Gsalpha (Toki et al., 1999). Desipramine 102-113 GNAS complex locus Homo sapiens 184-191 11353802-6 2001 Desipramine- or fluoxetine-treated cells show a more centralized clustering of Gsalpha in the Golgi region of the cell and a drastic reduction of Gsalpha in the cellular processes. Desipramine 0-11 GNAS complex locus Homo sapiens 79-86 11353802-6 2001 Desipramine- or fluoxetine-treated cells show a more centralized clustering of Gsalpha in the Golgi region of the cell and a drastic reduction of Gsalpha in the cellular processes. Desipramine 0-11 GNAS complex locus Homo sapiens 146-153 11417443-1 2001 OBJECTIVE: Duplication of CYP2D6 causes very rapid metabolism of CYP2D6 substrates such as desipramine. Desipramine 91-102 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 26-32 11417443-1 2001 OBJECTIVE: Duplication of CYP2D6 causes very rapid metabolism of CYP2D6 substrates such as desipramine. Desipramine 91-102 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 65-71 11106872-1 2001 The present study investigated the effect of long-term (15 mg/kg for 15 days) and acute (15 mg/kg, single administration) treatment with desmethylimipramine, a tricyclic antidepressant drug, on calcium/calmodulin-dependent protein kinase II (CaMKII), a kinase implicated in the mechanism of antidepressant drug action. Desipramine 137-156 calcium/calmodulin dependent protein kinase II gamma Homo sapiens 194-240 11303058-3 2001 Immunohistochemical analysis and in situ hybridization reveal that administration of the antidepressant drug desipramine decreases the accumulation of constitutively expressed TNF mRNA in neurons of the rat brain. Desipramine 109-120 tumor necrosis factor Rattus norvegicus 176-179 11246093-0 2001 Prolonged desipramine treatment increases the production of interleukin-10, an anti-inflammatory cytokine, in C57BL/6 mice subjected to the chronic mild stress model of depression. Desipramine 10-21 interleukin 10 Mus musculus 60-74 11246093-5 2001 RESULTS: Prolonged treatment of C57BL/6 mice subjected to CMS with desipramine increases the ability of T cells to produce IL-10 and the ability of B cells to proliferate after stimulation with LPS; and significantly decreases the cytotoxic activity of NK cells and the proliferative responses of lymphocytes after stimulation with Con-A, PHA and anti-CD3 monoclonal antibodies. Desipramine 67-78 interleukin 10 Mus musculus 123-128 11246093-6 2001 Repeated administration of desipramine to non-stressed mice increases the activity of T lymphocytes, lowers that of B lymphocytes, increases the production of IL-10 by T cells and has no significant effect on the activity of NK cells. Desipramine 27-38 interleukin 10 Mus musculus 159-164 11246093-7 2001 CONCLUSION: Prolonged desipramine treatment of stressed and non-stressed C57BL/6 mice induces an increase in the production of IL-10, an anti-inflammatory cytokine. Desipramine 22-33 interleukin 10 Mus musculus 127-132 11354249-9 2001 Inhibition of ceramide production by desipramine (25-50 microM) reduced UV-induced JNK activation in both 293 and Jurkat cells; and protects 293 cells from UV-induced apoptosis. Desipramine 37-48 mitogen-activated protein kinase 8 Homo sapiens 83-86 11106872-2 2001 Similar to selective and non-selective serotonin reuptake inhibitors, long-term, but not acute, treatment with desmethylimipramine markedly increased the activity of CaMKII in the hippocampal synaptic vesicle fraction (+51.9%). Desipramine 111-130 calcium/calmodulin dependent protein kinase II gamma Homo sapiens 166-172 11106872-1 2001 The present study investigated the effect of long-term (15 mg/kg for 15 days) and acute (15 mg/kg, single administration) treatment with desmethylimipramine, a tricyclic antidepressant drug, on calcium/calmodulin-dependent protein kinase II (CaMKII), a kinase implicated in the mechanism of antidepressant drug action. Desipramine 137-156 calcium/calmodulin dependent protein kinase II gamma Homo sapiens 242-248 10951284-8 2000 Furthermore, both palmitoyldihydrosphingosine and desipramine, a chemically and mechanically unrelated acid-sphingomyelinase inhibitor, significantly delay barrier recovery both 2 and 4 h after acute barrier abrogation. Desipramine 50-61 sphingomyelin phosphodiesterase 1 Homo sapiens 103-124 11082428-3 2000 We previously described down-regulation of the NET in cultured cells after continuous exposure to the tricyclic antidepressant desipramine. Desipramine 127-138 solute carrier family 6 member 2 Homo sapiens 47-50 11080533-9 2000 Desipramine decreased epidermal growth factor (EGF) levels in all age groups, but increased the secretion of peroxidase and lysozyme. Desipramine 0-11 epidermal growth factor like 1 Rattus norvegicus 22-45 11080533-9 2000 Desipramine decreased epidermal growth factor (EGF) levels in all age groups, but increased the secretion of peroxidase and lysozyme. Desipramine 0-11 epidermal growth factor like 1 Rattus norvegicus 47-50 11037800-2 2000 Desipramine is extensively metabolized by cytochrome P450 2D6 (CYP2D6), and kinetics of this compound are altered in poor metabolizers. Desipramine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 42-61 11037800-2 2000 Desipramine is extensively metabolized by cytochrome P450 2D6 (CYP2D6), and kinetics of this compound are altered in poor metabolizers. Desipramine 0-11 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 63-69 10913689-8 2001 Chronic treatment with desipramine or trimipramine, which do not directly inhibit 5-HT uptake, compared to fluoxetine and venlafaxine, lead to increases in 5-HT transporter densities in cingulate, agranular insular and perirhinal cortices. Desipramine 23-34 solute carrier family 6 member 4 Rattus norvegicus 156-172 10996474-3 2000 One and two weeks administration of desipramine significantly reduces the secretion of IL-4, an anti-inflammatory cytokine. Desipramine 36-47 interleukin 4 Mus musculus 87-91 10996474-5 2000 Prolonged desipramine administration (seven and 28 days) significantly increased the bioactivity of IL-1. Desipramine 10-21 interleukin 1 complex Mus musculus 100-104 10996474-6 2000 Four weeks of prolonged administration of amitriptyline and desipramine induces a significant increase in the secretion of IL-10, a cytokine with immunosuppressive and anti-inflammatory activities. Desipramine 60-71 interleukin 10 Mus musculus 123-128 10792576-7 2000 Desmethylimipramine (DMI) treatment for 24 days did not affect basal plasma concentrations of corticosterone or insulin, or arcuate nucleus NPY mRNA expressions, but significantly decreased basal levels of locus coeruleus NPY mRNA compared to saline-treated rats. Desipramine 0-19 neuropeptide Y Rattus norvegicus 222-225 12516468-3 2000 Furthermore, using RT-PCR mediated cross-species partial cDNA cloning, it was found that MW-97 and desipramine(DIM) increased NGF, BDNF mRNA in the frontal cortex after chronic administration for 21 days, meanwhile, BDNF mRNA in hippocampus was also increased. Desipramine 99-110 brain-derived neurotrophic factor Rattus norvegicus 131-135 12516468-3 2000 Furthermore, using RT-PCR mediated cross-species partial cDNA cloning, it was found that MW-97 and desipramine(DIM) increased NGF, BDNF mRNA in the frontal cortex after chronic administration for 21 days, meanwhile, BDNF mRNA in hippocampus was also increased. Desipramine 99-110 brain-derived neurotrophic factor Rattus norvegicus 216-220 10871295-3 2000 In situ hybridization to rat brain sections revealed that ICER mRNA expression was significantly increased by uncompetitive NMDA receptor antagonists (MK-801, phencyclidine, ketamine, memantine) but not by the competitive antagonist CPP [(+/-)-3-(2-carboxypiperazin-4-yl)-propyl-1-phosphonic acid] or other psychotropic agents (clozapine, haloperidol, desipramine). Desipramine 352-363 cAMP responsive element modulator Rattus norvegicus 58-62 10895986-0 2000 Metabolism of desipramine in Japanese psychiatric patients: the impact of CYP2D6 genotype on the hydroxylation of desipramine. Desipramine 114-125 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 74-80 10895986-1 2000 We investigated the impact of the genotype of CYP2D6 on the hydroxylation of desipramine in eighteen patients who were administered desipramine hydrochloride per os. Desipramine 77-88 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-52 10895986-1 2000 We investigated the impact of the genotype of CYP2D6 on the hydroxylation of desipramine in eighteen patients who were administered desipramine hydrochloride per os. Desipramine 132-157 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 46-52 10895986-4 2000 The genotyping of CYP2D6 only grossly predicts the steady state concentration of desipramine, mainly predicts the risk of getting very high plasma levels. Desipramine 81-92 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 18-24 10808050-6 2000 Chronic (14 days) administration of the antidepressant drugs desipramine or zimelidine transforms alpha(2)-adrenergic receptor-dependent decreases in TNF levels to increases in levels of TNF in the locus coeruleus. Desipramine 61-72 tumor necrosis factor Rattus norvegicus 150-153 10808050-6 2000 Chronic (14 days) administration of the antidepressant drugs desipramine or zimelidine transforms alpha(2)-adrenergic receptor-dependent decreases in TNF levels to increases in levels of TNF in the locus coeruleus. Desipramine 61-72 tumor necrosis factor Rattus norvegicus 187-190 10837797-14 2000 Treatment of rats with 6-OHDA resulted in a 52% decrease in the PDE4B3 and 58% decrease in 78 kDa PDE4B variant in hypothalamus; administration of desipramine attenuated the 6-OHDA-induced down-regulation of both PDE4B variants. Desipramine 147-158 phosphodiesterase 4B Rattus norvegicus 64-70 10837797-14 2000 Treatment of rats with 6-OHDA resulted in a 52% decrease in the PDE4B3 and 58% decrease in 78 kDa PDE4B variant in hypothalamus; administration of desipramine attenuated the 6-OHDA-induced down-regulation of both PDE4B variants. Desipramine 147-158 phosphodiesterase 4B Rattus norvegicus 64-69 10837797-14 2000 Treatment of rats with 6-OHDA resulted in a 52% decrease in the PDE4B3 and 58% decrease in 78 kDa PDE4B variant in hypothalamus; administration of desipramine attenuated the 6-OHDA-induced down-regulation of both PDE4B variants. Desipramine 147-158 phosphodiesterase 4B Rattus norvegicus 98-103 10837797-16 2000 In contrast, cerebellar PDE4B3 variant is up-regulated by 6-OHDA treatment and were partially normalized to control values by desipramine pretreatment. Desipramine 126-137 phosphodiesterase 4B Rattus norvegicus 24-30 10792576-7 2000 Desmethylimipramine (DMI) treatment for 24 days did not affect basal plasma concentrations of corticosterone or insulin, or arcuate nucleus NPY mRNA expressions, but significantly decreased basal levels of locus coeruleus NPY mRNA compared to saline-treated rats. Desipramine 21-24 neuropeptide Y Rattus norvegicus 222-225 10792576-8 2000 After repeated immobilization (2 h daily, for 4 days), DMI significantly reduced the stress-induced rise in locus coeruleus NPY mRNA levels, but potentiated the stress-induced rise in arcuate nucleus NPY mRNA expression. Desipramine 55-58 neuropeptide Y Rattus norvegicus 124-127 10792576-8 2000 After repeated immobilization (2 h daily, for 4 days), DMI significantly reduced the stress-induced rise in locus coeruleus NPY mRNA levels, but potentiated the stress-induced rise in arcuate nucleus NPY mRNA expression. Desipramine 55-58 neuropeptide Y Rattus norvegicus 200-203 10792576-9 2000 These results demonstrate that: (1) the increase in arcuate nucleus NPY mRNA expressions in stressed rats closely follows the decrease in plasma concentrations of insulin; (2) increases in NPY mRNA expressions occur in the absence of changes in plasma corticosterone; and (3) desipramine treatment potentiated the effect of stress on arcuate nucleus NPY mRNA expressions, but blocked the repeated stress-induced increase in locus coeruleus NPY mRNA expressions. Desipramine 276-287 neuropeptide Y Rattus norvegicus 189-192 10792576-9 2000 These results demonstrate that: (1) the increase in arcuate nucleus NPY mRNA expressions in stressed rats closely follows the decrease in plasma concentrations of insulin; (2) increases in NPY mRNA expressions occur in the absence of changes in plasma corticosterone; and (3) desipramine treatment potentiated the effect of stress on arcuate nucleus NPY mRNA expressions, but blocked the repeated stress-induced increase in locus coeruleus NPY mRNA expressions. Desipramine 276-287 neuropeptide Y Rattus norvegicus 189-192 10792576-9 2000 These results demonstrate that: (1) the increase in arcuate nucleus NPY mRNA expressions in stressed rats closely follows the decrease in plasma concentrations of insulin; (2) increases in NPY mRNA expressions occur in the absence of changes in plasma corticosterone; and (3) desipramine treatment potentiated the effect of stress on arcuate nucleus NPY mRNA expressions, but blocked the repeated stress-induced increase in locus coeruleus NPY mRNA expressions. Desipramine 276-287 neuropeptide Y Rattus norvegicus 189-192 10792576-10 2000 Thus, NPY mRNA expression in the arcuate nucleus and the locus coeruleus is sensitive to the effects of stress and to the antidepressant drug desipramine, but the arcuate nucleus NPY system is regulated by different mechanisms than the locus coeruleus NPY system. Desipramine 142-153 neuropeptide Y Rattus norvegicus 6-9 10706994-3 2000 The current study examined the effect of long-term administration, up to 6 weeks, of fluvoxamine, desipramine or saline on MAO-A and MAO-B activities in rat striatum, frontal cortex and liver. Desipramine 98-109 monoamine oxidase A Rattus norvegicus 123-128 11343573-4 2000 in desipramine- and citalopram-treated rats was abolished, indicating a desensitization of 5-HT1A receptors. Desipramine 3-14 5-hydroxytryptamine receptor 1A Rattus norvegicus 91-97 10706994-3 2000 The current study examined the effect of long-term administration, up to 6 weeks, of fluvoxamine, desipramine or saline on MAO-A and MAO-B activities in rat striatum, frontal cortex and liver. Desipramine 98-109 monoamine oxidase B Rattus norvegicus 133-138 10693959-1 2000 In a previous study, an up-regulation of rolipram-sensitive, low-Km, cyclic AMP phosphodiesterase (PDE4) subtype PDE4A in rat cerebral cortex following repeated treatment of desipramine was observed. Desipramine 174-185 phosphodiesterase 4A Rattus norvegicus 113-118 10693959-2 2000 To determine whether this effect is shared by antidepressants from different pharmacological classes, PDE4A expression was examined using immunoblot analyses following repeated treatment with the norepinephrine re-uptake inhibitor desipramine, the monoamine oxidase inhibitor phenelzine, the atypical antidepressant trazodone, and the serotonin reuptake inhibitor fluoxetine. Desipramine 231-242 phosphodiesterase 4A Rattus norvegicus 102-107 10693959-3 2000 Desipramine, phenelzine, and fluoxetine all increased the intensities of the PDE4A bands in hippocampal preparations; trazodone did not. Desipramine 0-11 phosphodiesterase 4A Rattus norvegicus 77-82 10693959-4 2000 In preparations of cerebral cortex, the intensities of the PDE4A bands were increased following desipramine treatment, not changed following phenelzine or fluoxetine treatment, and decreased following trazodone treatment. Desipramine 96-107 phosphodiesterase 4A Rattus norvegicus 59-64 10633490-2 2000 Coadministration of rolipram or Ro 20-1724 with an antidepressant (either desipramine or Org 4428) for 21 d resulted in a significant induction of BDNF mRNA in hippocampus relative to administration of vehicle. Desipramine 74-85 brain-derived neurotrophic factor Rattus norvegicus 147-151 10854034-5 2000 The induction of BDNF mRNA by MK-801 was attenuated by pre-treatment (1 h prior to MK-801 administration) with the antipsychotics, clozapine (25 mg/kg) and haloperidol (2 mg/kg), but not with the antidepressant desipramine (15 mg/kg). Desipramine 211-222 brain-derived neurotrophic factor Rattus norvegicus 17-21 10718121-2 2000 The formation of metabolite intermediate (MI)-complexes between cytochrome P450 (CYP) and the alkylamine-substituted drugs diltiazem (DTZ) and desipramine (DES) and their effect on CYP activities was investigated in rat liver. Desipramine 143-154 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 64-79 10718121-2 2000 The formation of metabolite intermediate (MI)-complexes between cytochrome P450 (CYP) and the alkylamine-substituted drugs diltiazem (DTZ) and desipramine (DES) and their effect on CYP activities was investigated in rat liver. Desipramine 143-154 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 81-84 10718121-2 2000 The formation of metabolite intermediate (MI)-complexes between cytochrome P450 (CYP) and the alkylamine-substituted drugs diltiazem (DTZ) and desipramine (DES) and their effect on CYP activities was investigated in rat liver. Desipramine 156-159 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 64-79 10718121-2 2000 The formation of metabolite intermediate (MI)-complexes between cytochrome P450 (CYP) and the alkylamine-substituted drugs diltiazem (DTZ) and desipramine (DES) and their effect on CYP activities was investigated in rat liver. Desipramine 156-159 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 81-84 10718121-2 2000 The formation of metabolite intermediate (MI)-complexes between cytochrome P450 (CYP) and the alkylamine-substituted drugs diltiazem (DTZ) and desipramine (DES) and their effect on CYP activities was investigated in rat liver. Desipramine 156-159 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 181-184 10718121-10 2000 By comparison, inhibition of CYP activity by DES was less pronounced than in control liver. Desipramine 45-48 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 29-32 10601816-9 2000 Vehicle treated OB rats displayed a significant impairment in LPS-induced IL-1beta (54%) and TNF-alpha (70%) secretion compared to their sham operated controls, an effect that was potentiated following chronic desipramine treatment. Desipramine 210-221 interleukin 1 beta Rattus norvegicus 74-82 10601816-9 2000 Vehicle treated OB rats displayed a significant impairment in LPS-induced IL-1beta (54%) and TNF-alpha (70%) secretion compared to their sham operated controls, an effect that was potentiated following chronic desipramine treatment. Desipramine 210-221 tumor necrosis factor Rattus norvegicus 93-102 10601816-10 2000 Furthermore, sham animals that were chronically treated with desipramine displayed decreases in LPS-provoked IL-1beta (51%) and TNF-alpha (49%) secretion compared to vehicle treated counterparts. Desipramine 61-72 interleukin 1 beta Rattus norvegicus 109-117 10601816-10 2000 Furthermore, sham animals that were chronically treated with desipramine displayed decreases in LPS-provoked IL-1beta (51%) and TNF-alpha (49%) secretion compared to vehicle treated counterparts. Desipramine 61-72 tumor necrosis factor Rattus norvegicus 128-137 10601816-13 2000 Whilst chronic treatment with desipramine normalized the behavioural hyperactivity observed in OB rats, such treatment further impaired LPS-induced IL-1beta and TNF-alpha secretion in bulbectomized rats. Desipramine 30-41 tumor necrosis factor Rattus norvegicus 161-170 10575045-5 1999 SERT density in the CA3 region of the hippocampus of the same rats, assessed by quantitative autoradiography with tritiated cyanoimipramine ([(3)H]CN-IMI), was decreased by 80-90% in SSRI-treated rats but not in those treated with phenelzine or DMI. Desipramine 245-248 solute carrier family 6 member 4 Rattus norvegicus 0-4 11015030-3 2000 We investigated the effect of 21 days administration, followed by 7 days withdrawal, of fluvoxamine (10 mg/kg), desipramine (10 mg/kg) and lithium carbonate (25 mg/kg) on PBR and CBR binding characteristics in male Sprague-Dawley rats. Desipramine 112-123 translocator protein Rattus norvegicus 171-174 10520140-4 1999 The time course of the effects of the tricyclic antidepressants desipramine and amitriptyline on GR binding, as assessed by [3H]dexamethasone binding using RU 28362, a specific agonist for GR, showed a biphasic mode of stimulation: desipramine significantly increased the GR binding with 2-day exposure by 36% over that in controls and by 99% and 60% with 10- and 14-day exposures, respectively. Desipramine 64-75 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 97-99 10520140-7 1999 Northern blot analysis demonstrated that the GR mRNA level was significantly increased by 14-day treatment with desipramine (+142% over control), amitriptyline (+108%), mianserin (+124%), paroxetine (+42%) and sulpiride (+92%), but not with haloperidol. Desipramine 112-123 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 45-47 10520140-8 1999 Immunocytochemistry for GR revealed that 2- or 14-day treatment with desipramine significantly increased the number of GR-positive cells with dominant immunoreactivity in the nuclei of granule cell-like neurones or in perikarya of pyramidal cell- and granule cell-like neurones. Desipramine 69-80 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 24-26 10520140-8 1999 Immunocytochemistry for GR revealed that 2- or 14-day treatment with desipramine significantly increased the number of GR-positive cells with dominant immunoreactivity in the nuclei of granule cell-like neurones or in perikarya of pyramidal cell- and granule cell-like neurones. Desipramine 69-80 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 119-121 10424850-0 1999 Desipramine treatment decreases 3H-nisoxetine binding and norepinephrine transporter mRNA in SK-N-SHSY5Y cells. Desipramine 0-11 solute carrier family 6 member 2 Homo sapiens 58-84 11015030-6 2000 After withdrawal, PBR density remained decreased in the liver in all three groups and in the kidneys of the desipramine- and lithium-treated animals. Desipramine 108-119 translocator protein Rattus norvegicus 18-21 11015030-7 2000 In the cerebral cortex, CBR density increased in response to all three agents, whereas PBR density decreased significantly in response to desipramine and lithium carbonate. Desipramine 138-149 translocator protein Rattus norvegicus 87-90 11015030-8 2000 Chronic treatment with fluvoxamine, desipramine and lithium carbonate is apparently associated with a modulation in PBR expression in the testes, adrenals, kidneys, liver and brain, and in CBR expression in brain. Desipramine 36-47 translocator protein Rattus norvegicus 116-119 10424850-1 1999 The antidepressant desipramine has been shown to decrease synaptic membrane concentrations of the norepinephrine re-uptake transporter (NET) in vivo and in vitro, on both an acute and a chronic basis. Desipramine 19-30 solute carrier family 6 member 2 Homo sapiens 98-134 10424850-1 1999 The antidepressant desipramine has been shown to decrease synaptic membrane concentrations of the norepinephrine re-uptake transporter (NET) in vivo and in vitro, on both an acute and a chronic basis. Desipramine 19-30 solute carrier family 6 member 2 Homo sapiens 136-139 10424850-3 1999 In this study, we treated SK-N-SHSY5Y cells with 100 nM desipramine for 24 or 72 h, and measured 3H-nisoxetine binding (as an estimate of NETs) and NET mRNA by quantitative reverse transcription polymerase chain reaction. Desipramine 56-67 solute carrier family 6 member 2 Homo sapiens 138-141 10424850-6 1999 We conclude that decreased NET synthesis may contribute to the chronic, but not acute, effect of desipramine to downregulate the NET. Desipramine 97-108 solute carrier family 6 member 2 Homo sapiens 27-30 10424850-6 1999 We conclude that decreased NET synthesis may contribute to the chronic, but not acute, effect of desipramine to downregulate the NET. Desipramine 97-108 solute carrier family 6 member 2 Homo sapiens 129-132 10201278-0 1999 Different effect of desipramine on protein kinase C in platelets between bipolar and major depressive disorders. Desipramine 20-31 proline rich transmembrane protein 2 Homo sapiens 35-51 10408227-7 1999 SKF 525-A, an inhibitor of cytochrome P450 (CYP), markedly suppressed DMI formation at both pH points. Desipramine 70-73 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 27-42 10408227-7 1999 SKF 525-A, an inhibitor of cytochrome P450 (CYP), markedly suppressed DMI formation at both pH points. Desipramine 70-73 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 44-47 10408227-9 1999 These results suggest that CYP enzyme(s) are mainly responsible for DMI formation at pH 8.5 and 7.5, and FMO enzyme(s) also are involved in IMI N-demethylation at a higher pH range in rat liver microsomes, at least in part. Desipramine 68-71 cytochrome P450, family 2, subfamily g, polypeptide 1 Rattus norvegicus 27-30 10218774-9 1999 This presynaptic modulation requires SNARE proteins because cleavage of SNAP-25 with the botulinum neurotoxin A strongly reduced the desipramine-induced glutamate release. Desipramine 133-144 synaptosome associated protein 25 Rattus norvegicus 72-79 9989935-6 1999 On the other hand, AST-RB2 efficiently catalyzed sulfonation of desipramine and DHEA, but had no activity toward 2-naphthol. Desipramine 64-75 sulfotransferase 2A1 Oryctolagus cuniculus 19-26 10201278-2 1999 The PKC activity of platelets incubated with desipramine was determined in vitro. Desipramine 45-56 proline rich transmembrane protein 2 Homo sapiens 4-7 10201278-3 1999 The PKC activity of the major depressive disorder subjects and healthy volunteers was inhibited by desipramine, whereas that of the bipolar disorder subjects showed both inhibition and activation. Desipramine 99-110 proline rich transmembrane protein 2 Homo sapiens 4-7 9861783-5 1998 Desipramine (DESI), another tricyclic antidepressant, also decreased the TRH content (from 63.6 +/- 2.5, at 10(-9) mol/L, to 12.6 +/- 0.4, at 10(-5) mol/L, fmol/well; P < 0.05). Desipramine 0-11 thyrotropin releasing hormone Rattus norvegicus 73-76 9987205-10 1999 In vitro experiments also showed that desipramine increased glucocorticoid receptor mRNA. Desipramine 38-49 nuclear receptor subfamily 3, group C, member 1 Mus musculus 60-83 10374896-4 1999 After chronic desipramine but not after chronic clorgyline treatments, the density (Bmax) of alpha2-adrenoceptors was increased (46%). Desipramine 14-25 adrenoceptor alpha 2A Rattus norvegicus 93-113 10374896-7 1999 The selective upregulation of the alpha2A-adrenoceptor subtype following chronic desipramine administration is compatible with a differential location and function of the alpha2-adrenoceptor subtypes in the rat kidney. Desipramine 81-92 adrenoceptor alpha 2A Rattus norvegicus 34-54 10435604-7 1999 Depressed patients showed a significant decrease in 5-HT2 receptor binding as measured by setoperone binding in frontal, temporal, parietal, and occipital cortical regions following desipramine treatment. Desipramine 182-193 5-hydroxytryptamine receptor 2A Homo sapiens 52-66 9877326-7 1998 The beta1-antagonist atenolol (1.0 micromol/l) potentiated the facilitatory effects of isoprenaline in the presence of DMI and corticosterone. Desipramine 119-122 UDP-GlcNAc:betaGal beta-1,3-N-acetylglucosaminyltransferase 2 Homo sapiens 4-9 9861783-5 1998 Desipramine (DESI), another tricyclic antidepressant, also decreased the TRH content (from 63.6 +/- 2.5, at 10(-9) mol/L, to 12.6 +/- 0.4, at 10(-5) mol/L, fmol/well; P < 0.05). Desipramine 13-17 thyrotropin releasing hormone Rattus norvegicus 73-76 9930326-6 1998 Transfection of H441 cells in the presence of 0.1-1 microgram/ml DMI-2 caused: (1) 10-fold enhancement of CAT activity when the bacterial plasmid was complexed with either surfactant protein A-poly-lysine or transferrin-poly-lysine; (2) 1.5- to two-fold enhancement of CAT activity in cells exposed to lipofectin-DNA complexes: (3) no effect on transfection via calcium phosphate co-precipitation. Desipramine 65-68 transferrin Homo sapiens 208-219 9758674-0 1998 Inhibition of desipramine hydroxylation (Cytochrome P450-2D6) in vitro by quinidine and by viral protease inhibitors: relation to drug interactions in vivo. Desipramine 14-25 cytochrome P450 2D6 Homo sapiens 41-60 9758674-3 1998 Biotransformation of desipramine (DMI) to hydroxydesipramine (OH-DMI), an index reaction used to profile activity of human cytochrome P450-2D6, was studied in vitro using human liver microsomes. Desipramine 34-37 cytochrome P450 2D6 Homo sapiens 123-142 9724254-6 1998 min-1), Na+-dependent mechanism that was inhibited by chlorimipramine > imipramine > fluoxetine > desipramine > zimelidine. Desipramine 107-118 CD59 molecule (CD59 blood group) Homo sapiens 0-5 9669506-2 1998 The selective 5-HT reuptake inhibitors paroxetine, indalpine and fluvoxamine displayed a high affinity for the 5-HT transporter, whereas the norepinephrine reuptake inhibitor desipramine had a high affinity for the norepinephrine transporter. Desipramine 175-186 solute carrier family 6 member 2 Rattus norvegicus 215-241 9778103-3 1998 injection of 10 mg/kg desipramine to naive mice increased the relative weight of their spleens, the response of their splenocytes to the mitogen concavaline-A and their ability to produce IL-10, as compared to saline controls. Desipramine 22-33 interleukin 10 Mus musculus 188-193 9778103-4 1998 Exposing the desipramine-treated mice to a swimming stress significantly reduced these parameters, as well as the levels of IL-2 and IFN-gamma, as compared to desipramine-treated mice. Desipramine 13-24 interleukin 2 Mus musculus 124-128 9778103-4 1998 Exposing the desipramine-treated mice to a swimming stress significantly reduced these parameters, as well as the levels of IL-2 and IFN-gamma, as compared to desipramine-treated mice. Desipramine 13-24 interferon gamma Mus musculus 133-142 9723120-14 1998 Since chronic desipramine, and not fluoxetine, is able to increase hippocampal glucocorticoid receptor (GR) expression, interactions of GR with CREB and SP1 may determine the lack of effect of desipramine on binding activity of the two latter transcription factors in this brain region. Desipramine 14-25 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 79-102 9723120-14 1998 Since chronic desipramine, and not fluoxetine, is able to increase hippocampal glucocorticoid receptor (GR) expression, interactions of GR with CREB and SP1 may determine the lack of effect of desipramine on binding activity of the two latter transcription factors in this brain region. Desipramine 14-25 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 104-106 9723124-0 1998 Bupropion and desipramine increase dopamine transporter mRNA expression in the ventral tegmental area/substantia nigra of rat brain. Desipramine 14-25 solute carrier family 6 member 3 Rattus norvegicus 35-55 9595891-18 1998 DISCUSSION: Treatment with either desipramine or ECT modified noradrenergic functioning in patients with depression, as assessed by growth hormone response to the clonidine challenge. Desipramine 34-45 growth hormone 1 Homo sapiens 132-146 9523572-10 1998 The present findings imply that down-regulation of the hNET in 293-hNET cells induced by desipramine results from a selective reduction in hNET protein levels, presumably a consequence of either a reduction in the translation of hNET mRNA or from an enhanced degradation of hNET protein. Desipramine 89-100 solute carrier family 6 member 2 Homo sapiens 55-59 9564441-10 1998 The decreases in 5-HT1A mRNA and binding, as well as the MR/GR alterations, were prevented in animals that received imipramine or desipramine antidepressant treatment. Desipramine 130-141 5-hydroxytryptamine receptor 1A Rattus norvegicus 17-23 9523560-9 1998 Scatchard analysis of [3H]desipramine binding revealed that IFN-alpha decreased the maximal binding (Bmax) values without any change in the dissociation constant (K(D)) values. Desipramine 26-37 interferon alpha-A Bos taurus 60-69 9625487-8 1998 These findings indicate that the extent of increases in imipramine and desipramine serum and brain levels are greater during elevated serum AAG states than during normal AAG states when imipramine is co-administered with fluoxetine. Desipramine 71-82 nitrogen permease regulator-like 3 Mus musculus 140-143 9625487-8 1998 These findings indicate that the extent of increases in imipramine and desipramine serum and brain levels are greater during elevated serum AAG states than during normal AAG states when imipramine is co-administered with fluoxetine. Desipramine 71-82 nitrogen permease regulator-like 3 Mus musculus 170-173 9523572-10 1998 The present findings imply that down-regulation of the hNET in 293-hNET cells induced by desipramine results from a selective reduction in hNET protein levels, presumably a consequence of either a reduction in the translation of hNET mRNA or from an enhanced degradation of hNET protein. Desipramine 89-100 solute carrier family 6 member 2 Homo sapiens 67-71 9523572-10 1998 The present findings imply that down-regulation of the hNET in 293-hNET cells induced by desipramine results from a selective reduction in hNET protein levels, presumably a consequence of either a reduction in the translation of hNET mRNA or from an enhanced degradation of hNET protein. Desipramine 89-100 solute carrier family 6 member 2 Homo sapiens 67-71 9523572-10 1998 The present findings imply that down-regulation of the hNET in 293-hNET cells induced by desipramine results from a selective reduction in hNET protein levels, presumably a consequence of either a reduction in the translation of hNET mRNA or from an enhanced degradation of hNET protein. Desipramine 89-100 solute carrier family 6 member 2 Homo sapiens 67-71 9523572-10 1998 The present findings imply that down-regulation of the hNET in 293-hNET cells induced by desipramine results from a selective reduction in hNET protein levels, presumably a consequence of either a reduction in the translation of hNET mRNA or from an enhanced degradation of hNET protein. Desipramine 89-100 solute carrier family 6 member 2 Homo sapiens 67-71 9495547-4 1997 Heterologous expression of rNET in HEK293 cells revealed that uptake of [3H]norepinephrine is sodium- and chloride-dependent and highly sensitive to the selective norepinephrine transporter inhibitors desipramine and nisoxetine. Desipramine 201-212 solute carrier family 6 member 2 Rattus norvegicus 27-31 9523572-0 1998 Down-regulation of the human norepinephrine transporter in intact 293-hNET cells exposed to desipramine. Desipramine 92-103 solute carrier family 6 member 2 Homo sapiens 29-55 9523572-0 1998 Down-regulation of the human norepinephrine transporter in intact 293-hNET cells exposed to desipramine. Desipramine 92-103 solute carrier family 6 member 2 Homo sapiens 70-74 9523572-1 1998 The effects of continuous exposure of cultured cells expressing the human norepinephrine transporter (hNET) to the hNET inhibitor desipramine on hNET expression and function were studied. Desipramine 130-141 solute carrier family 6 member 2 Homo sapiens 115-119 9523572-1 1998 The effects of continuous exposure of cultured cells expressing the human norepinephrine transporter (hNET) to the hNET inhibitor desipramine on hNET expression and function were studied. Desipramine 130-141 solute carrier family 6 member 2 Homo sapiens 115-119 9523572-2 1998 Exposure of HEK-293 cells transfected stably with the hNET cDNA (293-hNET cells) to desipramine for 3 days reduced the specific binding of [3H]nisoxetine in membrane homogenates in a concentration-dependent manner. Desipramine 84-95 solute carrier family 6 member 2 Homo sapiens 54-58 9523572-2 1998 Exposure of HEK-293 cells transfected stably with the hNET cDNA (293-hNET cells) to desipramine for 3 days reduced the specific binding of [3H]nisoxetine in membrane homogenates in a concentration-dependent manner. Desipramine 84-95 solute carrier family 6 member 2 Homo sapiens 69-73 9523572-3 1998 The magnitude of the reductions in [3H]nisoxetine binding to hNET was dependent on the length of time of the exposure to desipramine, reaching 77% after a 21-day exposure. Desipramine 121-132 solute carrier family 6 member 2 Homo sapiens 61-65 9523572-6 1998 Similar to binding, hNET protein levels returned to control levels 72 h after cessation of desipramine exposure. Desipramine 91-102 solute carrier family 6 member 2 Homo sapiens 20-24 9523572-7 1998 Northern blotting indicated that exposure of 293-hNET cells to desipramine did not significantly alter hNET mRNA levels. Desipramine 63-74 solute carrier family 6 member 2 Homo sapiens 49-53 9523572-8 1998 Uptake of [3H]norepinephrine by 293-hNET cells was markedly reduced after a 3-day exposure to desipramine. Desipramine 94-105 solute carrier family 6 member 2 Homo sapiens 36-40 9495547-4 1997 Heterologous expression of rNET in HEK293 cells revealed that uptake of [3H]norepinephrine is sodium- and chloride-dependent and highly sensitive to the selective norepinephrine transporter inhibitors desipramine and nisoxetine. Desipramine 201-212 solute carrier family 6 member 2 Bos taurus 163-189 9205822-1 1997 AIMS: In order to anticipate drug-interactions of potential clinical significance the ability of the novel antidepressant, venlafaxine, to inhibit CYP2D6 dependent imipramine and desipramine 2-hydroxylation was investigated in human liver microsomes. Desipramine 179-190 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 147-153 9462878-7 1997 Only DMI (5 mg/kg) suppressed AS at P11. Desipramine 5-8 S100 calcium binding protein A10 Rattus norvegicus 36-39 9428594-0 1997 Synthesis of 11C-labeled desipramine and its metabolite 2-hydroxydesipramine: potential radiotracers for PET studies of the norepinephrine transporter. Desipramine 25-36 solute carrier family 6 member 2 Homo sapiens 124-150 9428594-5 1997 [11C]DMI and [11C]HDMI have potential utility as PET radiotracers for the norepinephrine transporter. Desipramine 5-8 solute carrier family 6 member 2 Homo sapiens 74-100 9357451-7 1997 After administration of the tricyclic antidepressant desipramine, tissue sections obtained from the rat hippocampus and locus coeruleus are devoid of neuronal-associated TNF alpha immunoreactivity. Desipramine 53-64 tumor necrosis factor Rattus norvegicus 170-179 9380019-0 1997 Steroid-independent translocation of the glucocorticoid receptor by the antidepressant desipramine. Desipramine 87-98 nuclear receptor subfamily 3, group C, member 1 Mus musculus 41-64 9380019-5 1997 To examine this latter possibility, we evaluated translocation of the GR from the cytoplasm to the nucleus after 24-hr in vitro treatment of L929 cells (mouse fibroblasts) with the tricyclic antidepressant desipramine (0.1-10 microM) in the presence or absence of the synthetic steroid dexamethasone. Desipramine 206-217 nuclear receptor subfamily 3, group C, member 1 Mus musculus 70-72 9380019-7 1997 Desipramine was found to (i) induce GR translocation from the cytoplasm to the nucleus in the absence of steroids (with no effect alone on GR-mediated gene transcription) and (ii) potentiate dexamethasone-induced GR translocation and dexamethasone-induced GR-mediated gene transcription. Desipramine 0-11 nuclear receptor subfamily 3, group C, member 1 Mus musculus 36-38 9380019-7 1997 Desipramine was found to (i) induce GR translocation from the cytoplasm to the nucleus in the absence of steroids (with no effect alone on GR-mediated gene transcription) and (ii) potentiate dexamethasone-induced GR translocation and dexamethasone-induced GR-mediated gene transcription. Desipramine 0-11 nuclear receptor subfamily 3, group C, member 1 Mus musculus 139-141 9380019-7 1997 Desipramine was found to (i) induce GR translocation from the cytoplasm to the nucleus in the absence of steroids (with no effect alone on GR-mediated gene transcription) and (ii) potentiate dexamethasone-induced GR translocation and dexamethasone-induced GR-mediated gene transcription. Desipramine 0-11 nuclear receptor subfamily 3, group C, member 1 Mus musculus 139-141 9380019-7 1997 Desipramine was found to (i) induce GR translocation from the cytoplasm to the nucleus in the absence of steroids (with no effect alone on GR-mediated gene transcription) and (ii) potentiate dexamethasone-induced GR translocation and dexamethasone-induced GR-mediated gene transcription. Desipramine 0-11 nuclear receptor subfamily 3, group C, member 1 Mus musculus 139-141 9276071-3 1997 This report demonstrates that a single dose of desipramine (10 or 25 mg/kg), a classical tricyclic antidepressant drug acting on the adrenergic system, induced c-fos and zif268 expression in rat hippocampus without affecting c-jun. Desipramine 47-58 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 160-165 9276071-3 1997 This report demonstrates that a single dose of desipramine (10 or 25 mg/kg), a classical tricyclic antidepressant drug acting on the adrenergic system, induced c-fos and zif268 expression in rat hippocampus without affecting c-jun. Desipramine 47-58 early growth response 1 Rattus norvegicus 170-176 9314045-2 1997 We have investigated the effects of long-term treatment with desipramine on the phosphorylation state of microtubule-associated protein 2 (MAP2) and microtubule assembly in the rat cerebral cortex. Desipramine 61-72 microtubule-associated protein 2 Rattus norvegicus 105-137 9314045-2 1997 We have investigated the effects of long-term treatment with desipramine on the phosphorylation state of microtubule-associated protein 2 (MAP2) and microtubule assembly in the rat cerebral cortex. Desipramine 61-72 microtubule-associated protein 2 Rattus norvegicus 139-143 9314045-4 1997 The degree of phosphorylation of serine residues of MAP2 was significantly increased after chronic administration of desipramine without changes in the total concentration of MAP2. Desipramine 117-128 microtubule-associated protein 2 Rattus norvegicus 52-56 9314045-8 1997 These results raise the possibility that the changes in the degree of phosphorylation of MAP2 and microtubule assembly represent intracellular modifications involved in functional changes elicited by long-term treatment with desipramine. Desipramine 225-236 microtubule-associated protein 2 Rattus norvegicus 89-93 9220124-3 1997 This review article will focus on basic pharmacokinetic considerations for elderly patients when psychotropics metabolized by CYP2D6, such as nortriptyline and desipramine, are prescribed. Desipramine 160-171 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 126-132 9220124-12 1997 Significant correlations have been reported between individual CYP2D6 activity and plasma concentrations of nortriptyline and desipramine. Desipramine 126-137 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 63-69 9651108-0 1998 Influence of the antidepressants desipramine and fluoxetine on tryptophan-2,3-dioxygenase in the presence of exogenous melatonin. Desipramine 33-44 tryptophan 2,3-dioxygenase Rattus norvegicus 63-89 9651108-4 1998 Desipramine reduced basal hepatic tryptophan-2,3-dioxygenase activity in the liver while fluoxetine had no observable effect. Desipramine 0-11 tryptophan 2,3-dioxygenase Rattus norvegicus 34-60 9256169-6 1997 The binding of [3H]GBR-12935 to CYP2D6 was decreased partially by substrates or inhibitors of CYP2D isoforms (quinine, quinidine, propranolol, bufuralol, imipramine, and desipramine). Desipramine 170-181 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 32-38 9256169-6 1997 The binding of [3H]GBR-12935 to CYP2D6 was decreased partially by substrates or inhibitors of CYP2D isoforms (quinine, quinidine, propranolol, bufuralol, imipramine, and desipramine). Desipramine 170-181 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 32-37 9205822-5 1997 The inhibitory effect of imipramine and desipramine on liver microsomal CYP2D6 dependent venlafaxine O-demethylation was determined similarly. Desipramine 40-51 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 72-78 9205822-10 1997 Imipramine and desipramine gave marked inhibition of CYP2D6 dependent venlafaxine O-demethylase activity (Ki values of 3.9 +/- 1.7 and 1.7 +/- 0.9 microM, respectively). Desipramine 15-26 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 53-59 9169301-3 1997 In this study, the influence of the selective beta 1- (CGP 20712A) and beta 2- (ICI 118551) adrenergic blockers on the antinociceptive effect of desipramine and nortriptyline was studied in mice using physical and chemical nociceptive tests that implicate different levels of sensory-motor integration in the central nervous system (CNS). Desipramine 145-156 hemoglobin, beta adult major chain Mus musculus 46-52 9169301-3 1997 In this study, the influence of the selective beta 1- (CGP 20712A) and beta 2- (ICI 118551) adrenergic blockers on the antinociceptive effect of desipramine and nortriptyline was studied in mice using physical and chemical nociceptive tests that implicate different levels of sensory-motor integration in the central nervous system (CNS). Desipramine 145-156 hemoglobin, beta adult minor chain Mus musculus 71-77 9063880-6 1997 It has the same specificity as wild type SERT for the antidepressants paroxetine and desipramine. Desipramine 85-96 solute carrier family 6 member 4 Homo sapiens 41-45 9084457-2 1997 At least three different P450s appear to be responsible for the N-demethylation of imipramine to desipramine in vivo: CYP1A2, CYP2C19, and CYP3A4. Desipramine 97-108 cytochrome P450 family 1 subfamily A member 2 Homo sapiens 118-124 9084457-2 1997 At least three different P450s appear to be responsible for the N-demethylation of imipramine to desipramine in vivo: CYP1A2, CYP2C19, and CYP3A4. Desipramine 97-108 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 126-133 9084457-2 1997 At least three different P450s appear to be responsible for the N-demethylation of imipramine to desipramine in vivo: CYP1A2, CYP2C19, and CYP3A4. Desipramine 97-108 cytochrome P450 family 3 subfamily A member 4 Homo sapiens 139-145 9049581-0 1997 Relationship between plasma desipramine levels, CYP2D6 phenotype and clinical response to desipramine: a prospective study. Desipramine 90-101 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 48-54 9049581-1 1997 OBJECTIVE: The clinical relevance of the CYP2D6 oxidation polymorphism in the treatment of depression with desipramine (DMI) was studied prospectively in depressed outpatients. Desipramine 107-118 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 41-47 9049581-1 1997 OBJECTIVE: The clinical relevance of the CYP2D6 oxidation polymorphism in the treatment of depression with desipramine (DMI) was studied prospectively in depressed outpatients. Desipramine 120-123 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 41-47 9503270-3 1997 DMI, as previously reported, significantly increased hippocampal glucocorticoid receptor (GR) mRNA while the equipotent NE uptake inhibitor (+)-oxaprotiline like the inactive (-)-oxaprotiline did not alter hippocampal levels of GR mRNA. Desipramine 0-3 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 65-88 9503270-3 1997 DMI, as previously reported, significantly increased hippocampal glucocorticoid receptor (GR) mRNA while the equipotent NE uptake inhibitor (+)-oxaprotiline like the inactive (-)-oxaprotiline did not alter hippocampal levels of GR mRNA. Desipramine 0-3 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 90-92 9503270-3 1997 DMI, as previously reported, significantly increased hippocampal glucocorticoid receptor (GR) mRNA while the equipotent NE uptake inhibitor (+)-oxaprotiline like the inactive (-)-oxaprotiline did not alter hippocampal levels of GR mRNA. Desipramine 0-3 nuclear receptor subfamily 3, group C, member 1 Rattus norvegicus 228-230 9395259-0 1997 Interaction mechanisms of imipramine and desipramine with enkephalin-degrading aminopeptidases in vitro. Desipramine 41-52 proenkephalin Rattus norvegicus 58-68 9395259-4 1997 The present work shows the effects in vitro of imipramine and its active metabolite desipramine on the activities of two membrane-bound enkephalin-degrading aminopeptidases present in rat brain. Desipramine 84-95 proenkephalin Rattus norvegicus 136-146 8832768-7 1996 Basal and DMI-induced GH levels, on the other hand, did not differ in PTSD versus normal subjects. Desipramine 10-13 growth hormone 1 Homo sapiens 22-24 8893267-1 1996 PURPOSE: The effect of elevated serum alpha-1-acid glycoprotein (AAG) concentrations on the steady-state serum and brain levels of imipramine and its metabolite desipramine was assessed. Desipramine 161-172 nitrogen permease regulator-like 3 Mus musculus 38-63 8893267-1 1996 PURPOSE: The effect of elevated serum alpha-1-acid glycoprotein (AAG) concentrations on the steady-state serum and brain levels of imipramine and its metabolite desipramine was assessed. Desipramine 161-172 nitrogen permease regulator-like 3 Mus musculus 65-68 8893267-11 1996 CONCLUSIONS: Elevated serum AAG impedes the transport of imipramine and desipramine into the brain. Desipramine 72-83 nitrogen permease regulator-like 3 Mus musculus 28-31 8835703-6 1996 In conclusion, by taking into account that the incidence of the PMs of CYP2C19 is much greater (18-23%) than that of CYP2D6 (< 1%) in Japanese population, the individually predetermined assessment of the CYP2C19-mediated metabolic capacity of imipramine would be more valuable than that of the CYP2D6-mediated capacity for forecasting the steady-state concentrations of imipramine and desipramine in Japanese depressive patients, thereby attaining an individualized optimization of imipramine therapy. Desipramine 388-399 cytochrome P450 family 2 subfamily C member 19 Homo sapiens 71-78 9178363-2 1997 The purpose of this study was to examine the phosphorylation of a major PKC substrate involved in modulation of neurotransmitter release, GAP-43, in a synaptosomal preparation from rat cerebral cortex after repeated administration of fluxetine (FL) and desipramine (DMI). Desipramine 253-264 protein kinase C, gamma Rattus norvegicus 72-75 9178363-2 1997 The purpose of this study was to examine the phosphorylation of a major PKC substrate involved in modulation of neurotransmitter release, GAP-43, in a synaptosomal preparation from rat cerebral cortex after repeated administration of fluxetine (FL) and desipramine (DMI). Desipramine 253-264 growth associated protein 43 Rattus norvegicus 138-144 9178363-2 1997 The purpose of this study was to examine the phosphorylation of a major PKC substrate involved in modulation of neurotransmitter release, GAP-43, in a synaptosomal preparation from rat cerebral cortex after repeated administration of fluxetine (FL) and desipramine (DMI). Desipramine 266-269 protein kinase C, gamma Rattus norvegicus 72-75 9178363-2 1997 The purpose of this study was to examine the phosphorylation of a major PKC substrate involved in modulation of neurotransmitter release, GAP-43, in a synaptosomal preparation from rat cerebral cortex after repeated administration of fluxetine (FL) and desipramine (DMI). Desipramine 266-269 growth associated protein 43 Rattus norvegicus 138-144 9178363-8 1997 The extent of phosphorylation of GAP-43 protein by native PKC in synaptosomes of rats treated with either FL or DMI was not significantly different from that observed in control animals. Desipramine 112-115 growth associated protein 43 Rattus norvegicus 33-39 9178363-8 1997 The extent of phosphorylation of GAP-43 protein by native PKC in synaptosomes of rats treated with either FL or DMI was not significantly different from that observed in control animals. Desipramine 112-115 protein kinase C, gamma Rattus norvegicus 58-61 9178363-9 1997 The previously observed suppression of basal PKC activity in rat cortical synaptosomes by FL and DMI treatment was thus not reflected in altered GAP-43 phosphorylation. Desipramine 97-100 protein kinase C, gamma Rattus norvegicus 45-48 8938825-2 1996 The authors studied the effects of coadministration of desipramine, which is a substrate of CYP2D6, on plasma concentrations of bromperidol and its reduced metabolite (reduced bromperidol). Desipramine 55-66 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 92-98 8710929-6 1996 These events are stimulated by NE and by guanethidine, an hNET substrate, and they are blocked by cocaine and the antidepressant desipramine. Desipramine 129-140 solute carrier family 6 member 2 Homo sapiens 58-62 8667208-6 1996 Desipramine blocked the toxicity of MPP+ toward the noradrenaline transporter, but not the dopamine transporter expressing cells. Desipramine 0-11 solute carrier family 6 member 2 Homo sapiens 52-77 8627519-5 1996 Chronic treatment with imipramine and desipramine alone induced Fos-LI in the central nucleus of the amygdala and the dorsolateral bed nucleus of the stria terminalis. Desipramine 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 64-67 8627519-8 1996 Chronic administration of imipramine, desipramine and nisoxetine antagonized the swim induced expression of Fos-LI in the PVN and in limbic cortical regions, including the medial prefrontal ventrolateral orbital and cingulate cortices. Desipramine 38-49 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 108-111 8627519-10 1996 Thus, only antidepressant drugs that affect norepinephrine uptake (i.e., imipramine, desipramine and nisoxetine) antagonized swim stress-induced Fos-LI. Desipramine 85-96 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 145-148 8737449-0 1996 Effect of desipramine on spontaneous activity of hippocampal CA1 neuron after transient cerebral ischemia in rats. Desipramine 10-21 carbonic anhydrase 1 Rattus norvegicus 61-64 8730743-0 1996 Desipramine administration in the olfactory bulbectomized rat: changes in brain beta-adrenoceptor and 5-HT2A binding sites and their relationship to behaviour. Desipramine 0-11 5-hydroxytryptamine receptor 2A Rattus norvegicus 102-108 8730743-16 1996 DMI administration for up to 21 days produced a progressive reduction in the number of 5-HT2A receptors in frontal cortex, without significant alterations in occipital cortex. Desipramine 0-3 5-hydroxytryptamine receptor 2A Rattus norvegicus 87-93 8730743-20 1996 The present results suggest that the action of DMI in this animal model is unlikely to be directly related to a reduction in beta-adrenoceptors but may be related to a reduction in frontal cortical 5-HT2A receptors. Desipramine 47-50 5-hydroxytryptamine receptor 2A Rattus norvegicus 198-204 8848823-6 1996 These results suggest that PB is an inducer of the 2-hydroxylation of DMI, a reaction primarily catalyzed by CYP2D6, but do not provide further information on the specific P450 isoenzyme(s) being induced. Desipramine 70-73 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 109-115 8724644-8 1996 The reduction of NA levels induced by DSP-4 was prevented by the concomitant administration of the NA uptake inhibitor desipramine. Desipramine 119-130 dual specificity phosphatase 26 Homo sapiens 38-43 8883917-5 1996 When the 6-OHDA treatment was preceded by administration of desmethylimipramine which prevents NE depletion, IL-1 treatment increased Fos in the PVN, suggesting that the effect of 6-OHDA was indeed related to the depletion of NE. Desipramine 60-79 interleukin 1 complex Mus musculus 109-113 8883917-5 1996 When the 6-OHDA treatment was preceded by administration of desmethylimipramine which prevents NE depletion, IL-1 treatment increased Fos in the PVN, suggesting that the effect of 6-OHDA was indeed related to the depletion of NE. Desipramine 60-79 FBJ osteosarcoma oncogene Mus musculus 134-137 8741184-3 1996 beta-Adrenoceptor subtypes, beta 1 and beta 2, which coexist in neurons and astrocytes, are differently distributed in the brain and differently modified by desipramine administration which down-regulates beta 1-adrenoceptor in forebrain neurons and astrocytes and beta 2-adrenoceptor in cerebellum neurons. Desipramine 157-168 adrenoceptor beta 1 Rattus norvegicus 205-224 8741184-3 1996 beta-Adrenoceptor subtypes, beta 1 and beta 2, which coexist in neurons and astrocytes, are differently distributed in the brain and differently modified by desipramine administration which down-regulates beta 1-adrenoceptor in forebrain neurons and astrocytes and beta 2-adrenoceptor in cerebellum neurons. Desipramine 157-168 adrenoceptor beta 2 Rattus norvegicus 265-284 8845222-14 1996 These data suggest that NO production attributable to constitutive NOS (cNOS) activity aggravates the hypotension associated with DMI toxicity in the anesthetized rat, and contributes to the pathophysiology of this overdose. Desipramine 130-133 nitric oxide synthase 3 Rattus norvegicus 54-70 8845222-14 1996 These data suggest that NO production attributable to constitutive NOS (cNOS) activity aggravates the hypotension associated with DMI toxicity in the anesthetized rat, and contributes to the pathophysiology of this overdose. Desipramine 130-133 nitric oxide synthase 3 Rattus norvegicus 72-76 8737449-1 1996 AIM: To study the spontaneous firing of CA1 neurons in rat hippocampus after transient cerebral ischemia and the effect of desipramine (Des) on the post-ischemic electric activity of CA1 neurons. Desipramine 123-134 carbonic anhydrase 1 Rattus norvegicus 183-186 8737449-1 1996 AIM: To study the spontaneous firing of CA1 neurons in rat hippocampus after transient cerebral ischemia and the effect of desipramine (Des) on the post-ischemic electric activity of CA1 neurons. Desipramine 136-139 carbonic anhydrase 1 Rattus norvegicus 183-186 8848002-5 1995 In contrast, all chimeras containing dopamine transporter sequences from this region resemble the dopamine transporter, which demonstrates higher affinity for psychomotor stimulants compared with antidepressants (e.g., Ki = 391 +/- 39 nM cocaine compared with 9365 +/- 1260 nM desipramine). Desipramine 277-288 solute carrier family 6 member 3 Homo sapiens 37-57 8848002-5 1995 In contrast, all chimeras containing dopamine transporter sequences from this region resemble the dopamine transporter, which demonstrates higher affinity for psychomotor stimulants compared with antidepressants (e.g., Ki = 391 +/- 39 nM cocaine compared with 9365 +/- 1260 nM desipramine). Desipramine 277-288 solute carrier family 6 member 3 Homo sapiens 98-118 8848002-6 1995 A region including transmembrane domains 1-3 of the norepinephrine transporter also contributes to the interaction of desipramine and nisoxetine, whereas the analogous region of the dopamine transporter influences the affinity for piperazine derivatives (e.g., GBR12909 and LR1111) that are selective for the dopamine transporter. Desipramine 118-129 solute carrier family 6 member 3 Homo sapiens 309-329 7472505-7 1995 Chronic (21 d), but not acute (1 d), administration of several different antidepressant drugs, including tranylcypromine, sertraline, desipramine, or mianserin, significantly increased BDNF mRNA and all but mianserin increased trkB mRNA in hippocampus. Desipramine 134-145 brain-derived neurotrophic factor Rattus norvegicus 185-189 8617667-10 1995 Increased DMI played a major role in the compensatory gain response in both CL-CA and FA-CA groups. Desipramine 10-13 FA complementation group A Homo sapiens 86-91 8564731-7 1995 The serine but not threonine or tyrosine phosphorylation of MAP2 was significantly increased after chronic treatment with DMI, MPR or CTR. Desipramine 122-125 microtubule-associated protein 2 Rattus norvegicus 60-64 7589222-0 1995 Chronic fluoxetine or desmethylimipramine treatment alters 5-HT2 receptor mediated c-fos gene expression. Desipramine 22-41 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 83-88 7582477-0 1995 Protein kinase C in rat brain cortex and hippocampus: effect of repeated administration of fluoxetine and desipramine. Desipramine 106-117 protein kinase C, gamma Rattus norvegicus 0-16 7582477-3 1995 The aim of this study was to investigate the effect of repeated administration of fluoxetine (FL) and desipramine (DMI) on the distribution and activity of protein kinase C (PKC) in subcellular fractions of rat cortex (Cx) and hippocampus (Hc) under basal conditions and in response to a single in vivo administration of 5-HT2A/2C agonist, 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI). Desipramine 102-113 protein kinase C, gamma Rattus norvegicus 156-172 7582477-3 1995 The aim of this study was to investigate the effect of repeated administration of fluoxetine (FL) and desipramine (DMI) on the distribution and activity of protein kinase C (PKC) in subcellular fractions of rat cortex (Cx) and hippocampus (Hc) under basal conditions and in response to a single in vivo administration of 5-HT2A/2C agonist, 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI). Desipramine 102-113 protein kinase C, gamma Rattus norvegicus 174-177 7582477-3 1995 The aim of this study was to investigate the effect of repeated administration of fluoxetine (FL) and desipramine (DMI) on the distribution and activity of protein kinase C (PKC) in subcellular fractions of rat cortex (Cx) and hippocampus (Hc) under basal conditions and in response to a single in vivo administration of 5-HT2A/2C agonist, 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI). Desipramine 115-118 protein kinase C, gamma Rattus norvegicus 156-172 7582477-3 1995 The aim of this study was to investigate the effect of repeated administration of fluoxetine (FL) and desipramine (DMI) on the distribution and activity of protein kinase C (PKC) in subcellular fractions of rat cortex (Cx) and hippocampus (Hc) under basal conditions and in response to a single in vivo administration of 5-HT2A/2C agonist, 1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI). Desipramine 115-118 protein kinase C, gamma Rattus norvegicus 174-177 7582477-15 1995 The activity of PKC in subcellular fractions of Cx and Hc was significantly (by 40-50%) decreased in rats given repeated doses of FL or DMI. Desipramine 136-139 protein kinase C, gamma Rattus norvegicus 16-19 7582477-20 1995 The results indicate that repeated administration of FL and DMI produced similar changes in basal PKC activity but differentially affected the PKC response to the 5-HT2A/2c receptor agonist, DOI. Desipramine 60-63 protein kinase C, gamma Rattus norvegicus 98-101 7582477-20 1995 The results indicate that repeated administration of FL and DMI produced similar changes in basal PKC activity but differentially affected the PKC response to the 5-HT2A/2c receptor agonist, DOI. Desipramine 60-63 protein kinase C, gamma Rattus norvegicus 143-146 7582477-20 1995 The results indicate that repeated administration of FL and DMI produced similar changes in basal PKC activity but differentially affected the PKC response to the 5-HT2A/2c receptor agonist, DOI. Desipramine 60-63 5-hydroxytryptamine receptor 2A Rattus norvegicus 163-169 7883833-4 1995 In rats, DMI reduces the activity of the locus ceruleus, a major source of NE innervation of the hypothalamic paraventricular nucleus, the site of CRH neurons. Desipramine 9-12 corticotropin releasing hormone Rattus norvegicus 147-150 7667349-0 1995 Chronic desipramine alters stress-induced behaviors and regional expression of the immediate early gene, c-fos. Desipramine 8-19 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 105-110 7667349-1 1995 This experiment examined the effects of acute or chronic administration of the antidepressant drug desipramine on conditioned stress-induced behaviors and regional c-fos expression in the brain. Desipramine 99-110 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 164-169 7667349-7 1995 Similarly, Fos immunohistochemistry revealed that the chronic desipramine group showing positive behavioral effects was the only group in which there were significant reductions in the number of stress-induced Fos-positive neurons in five of 60 structures surveyed. Desipramine 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 11-14 7667349-7 1995 Similarly, Fos immunohistochemistry revealed that the chronic desipramine group showing positive behavioral effects was the only group in which there were significant reductions in the number of stress-induced Fos-positive neurons in five of 60 structures surveyed. Desipramine 62-73 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 210-213 7603449-8 1995 Upon membrane depolarization, desipramine reduced the phosphorylation of CREB at Ser-119 and also blocked the depolarization-induced increase in the intracellular free Ca2+ concentration in HIT cells. Desipramine 30-41 cAMP responsive element binding protein 1 Rattus norvegicus 73-77 7612155-5 1995 Moreover, chronic administration of desipramine or sertraline, selective re-uptake inhibitors of norepinephrine, or serotonin, respectively, also significantly down-regulated the induction of c-fos mRNA in response to restraint stress. Desipramine 36-47 Fos proto-oncogene, AP-1 transcription factor subunit Rattus norvegicus 192-197 7883833-13 1995 In summary, DMI increased basal cortisol levels consistent with facilitation of NE neurotransmission and, hence, hypothalamic CRH release. Desipramine 12-15 corticotropin releasing hormone Homo sapiens 126-129 7891324-2 1995 To delineate the pharmacokinetic mechanisms, we studied the effects of a high-affinity, drug-specific Fab fragment on desipramine (DMI) disposition in rats and on DMI kinetics in the isolated, perfused rat liver. Desipramine 118-129 FA complementation group B Homo sapiens 102-105 7891324-2 1995 To delineate the pharmacokinetic mechanisms, we studied the effects of a high-affinity, drug-specific Fab fragment on desipramine (DMI) disposition in rats and on DMI kinetics in the isolated, perfused rat liver. Desipramine 131-134 FA complementation group B Homo sapiens 102-105 7891324-4 1995 Rats received 20 mg/kg DMI intravenously over 30 min followed in 10 min by DMI-specific ovine polyclonal Fab (DMI-Fab) or nonspecific human Fab (control-Fab) (1.1 g/kg; molar Fab-to-DMI ratio, 0.34) intravenously over 20 min. Desipramine 75-78 FA complementation group B Homo sapiens 105-108 7891324-4 1995 Rats received 20 mg/kg DMI intravenously over 30 min followed in 10 min by DMI-specific ovine polyclonal Fab (DMI-Fab) or nonspecific human Fab (control-Fab) (1.1 g/kg; molar Fab-to-DMI ratio, 0.34) intravenously over 20 min. Desipramine 75-78 FA complementation group B Homo sapiens 105-108 7891324-5 1995 The serum DMI concentration increased by 50-fold 5 min after DMI-Fab administration. Desipramine 10-13 FA complementation group B Homo sapiens 65-68 7891324-8 1995 Renal clearance increased by 72% after DMI-Fab and total renal excretion of DMI increased by 7-fold due to the much higher serum DMI concentration. Desipramine 39-42 FA complementation group B Homo sapiens 43-46 7891324-9 1995 Ninety-four percent of DMI-Fab excretion and 87% of DMI excretion occurred in the first 12 h. The percent of DMI bound in urine was markedly increased by DMI-Fab (87.1 vs. 19.1%), as was the molar ratio of DMI to DMI-Fab in urine (0.75 vs. 0.08). Desipramine 23-26 FA complementation group B Homo sapiens 27-30 7891324-9 1995 Ninety-four percent of DMI-Fab excretion and 87% of DMI excretion occurred in the first 12 h. The percent of DMI bound in urine was markedly increased by DMI-Fab (87.1 vs. 19.1%), as was the molar ratio of DMI to DMI-Fab in urine (0.75 vs. 0.08). Desipramine 23-26 FA complementation group B Homo sapiens 158-161 7891324-9 1995 Ninety-four percent of DMI-Fab excretion and 87% of DMI excretion occurred in the first 12 h. The percent of DMI bound in urine was markedly increased by DMI-Fab (87.1 vs. 19.1%), as was the molar ratio of DMI to DMI-Fab in urine (0.75 vs. 0.08). Desipramine 52-55 FA complementation group B Homo sapiens 158-161 7891324-9 1995 Ninety-four percent of DMI-Fab excretion and 87% of DMI excretion occurred in the first 12 h. The percent of DMI bound in urine was markedly increased by DMI-Fab (87.1 vs. 19.1%), as was the molar ratio of DMI to DMI-Fab in urine (0.75 vs. 0.08). Desipramine 52-55 FA complementation group B Homo sapiens 158-161 7550552-1 1995 The tricyclic antidepressant desipramine impaired shuttle-box avoidance acquisition in mice of the CD-1 strain. Desipramine 29-40 CD1 antigen complex Mus musculus 99-103 7898321-0 1994 Desipramine-induced increase in norepinephrine transporter mRNA is not mediated via alpha 2 receptors. Desipramine 0-11 solute carrier family 6 member 2 Rattus norvegicus 32-58 7898321-1 1994 In situ hybridization for the norepinephrine transporter (NET) was performed in rats receiving short-term (2 days) treatment with either an alpha-2 (alpha 2) receptor agonist (clonidine) or antagonist (yohimbine) followed by saline or desipramine (DMI). Desipramine 235-246 solute carrier family 6 member 2 Rattus norvegicus 58-61 7898321-1 1994 In situ hybridization for the norepinephrine transporter (NET) was performed in rats receiving short-term (2 days) treatment with either an alpha-2 (alpha 2) receptor agonist (clonidine) or antagonist (yohimbine) followed by saline or desipramine (DMI). Desipramine 248-251 solute carrier family 6 member 2 Rattus norvegicus 58-61 7898321-6 1994 In the "DMI" group an attentuation of the DMI-induced increase in NET mRNA was observed in the clonidine/DMI animals compared to the saline/DMI animals. Desipramine 8-11 solute carrier family 6 member 2 Rattus norvegicus 66-69 7898321-6 1994 In the "DMI" group an attentuation of the DMI-induced increase in NET mRNA was observed in the clonidine/DMI animals compared to the saline/DMI animals. Desipramine 42-45 solute carrier family 6 member 2 Rattus norvegicus 66-69 7898321-6 1994 In the "DMI" group an attentuation of the DMI-induced increase in NET mRNA was observed in the clonidine/DMI animals compared to the saline/DMI animals. Desipramine 42-45 solute carrier family 6 member 2 Rattus norvegicus 66-69 7898321-6 1994 In the "DMI" group an attentuation of the DMI-induced increase in NET mRNA was observed in the clonidine/DMI animals compared to the saline/DMI animals. Desipramine 42-45 solute carrier family 6 member 2 Rattus norvegicus 66-69 7898321-8 1994 These findings suggest that the DMI-induced increase in NET mRNA is not mediated via alpha 2 receptors for, although clonidine attenuates DMI"s effect, there is no reciprocal enhancement with the alpha 2 antagonist yohimbine. Desipramine 32-35 solute carrier family 6 member 2 Rattus norvegicus 56-59 7898321-10 1994 Additional studies are needed to clarify the mechanism of the DMI-induced increase in NET mRNA and to correlate changes in NET mRNA with transporter expression at the synaptic membrane. Desipramine 62-65 solute carrier family 6 member 2 Rattus norvegicus 86-89 7816863-0 1994 Beta 2- but not beta 1-adrenoceptors are involved in desipramine enhancement of aggressive behavior in long-term isolated mice. Desipramine 53-64 hemoglobin, beta adult minor chain Mus musculus 0-6 7816863-4 1994 ICI118,551 HCl (1.25-5 mg/kg, IP), a selective beta 2-adrenoceptor antagonist, also blocked the desipramine-induced enhancement of aggressive behavior in a dose-dependent manner, whereas metoprolol tartrate (5-20 mg/kg, IP), a selective beta 1-adrenoceptor antagonist, did not affect it. Desipramine 96-107 adrenergic receptor, beta 2 Mus musculus 47-66 7816863-4 1994 ICI118,551 HCl (1.25-5 mg/kg, IP), a selective beta 2-adrenoceptor antagonist, also blocked the desipramine-induced enhancement of aggressive behavior in a dose-dependent manner, whereas metoprolol tartrate (5-20 mg/kg, IP), a selective beta 1-adrenoceptor antagonist, did not affect it. Desipramine 96-107 adrenergic receptor, beta 1 Mus musculus 237-256 7816863-6 1994 Taken together with our previous finding that the desipramine-induced enhancement of aggressive behavior can be blocked by yohimbine, an alpha 2-adrenoceptor antagonist, the present results indicate that not only alpha 2- but also beta 2-adrenoceptor stimulation plays important roles in modulation of aggressive behavior in long-term isolated mice. Desipramine 50-61 adrenergic receptor, beta 2 Mus musculus 231-250 7604141-4 1995 These findings suggest that CBZ induces the 2-hydroxylation of DMI, a reaction primarily catalyzed by the polymorphic CYP2D6 isozyme. Desipramine 63-66 cytochrome P450 family 2 subfamily D member 6 Homo sapiens 118-124 7895065-2 1994 Antidepressant administration of desipramine to rats twice-daily for 2 weeks increased hippocampal TNF levels and transformed the presynaptic TNF response. Desipramine 33-44 tumor necrosis factor Rattus norvegicus 99-102 7895065-2 1994 Antidepressant administration of desipramine to rats twice-daily for 2 weeks increased hippocampal TNF levels and transformed the presynaptic TNF response. Desipramine 33-44 tumor necrosis factor Rattus norvegicus 142-145 7895065-3 1994 One day of desipramine administration resulted in increased locus coeruleus TNF mRNA accumulation and, simultaneously, hippocampal TNF levels escalated. Desipramine 11-22 tumor necrosis factor Rattus norvegicus 76-79 7895065-3 1994 One day of desipramine administration resulted in increased locus coeruleus TNF mRNA accumulation and, simultaneously, hippocampal TNF levels escalated. Desipramine 11-22 tumor necrosis factor Rattus norvegicus 131-134 7895065-6 1994 In addition, TNF potentiation of [3H]norepinephrine release after chronic desipramine administration was reversed in the presence of idazoxan to a greater inhibition than in control slices exposed to idazoxan. Desipramine 74-85 tumor necrosis factor Rattus norvegicus 13-16 7810621-6 1994 In the absence and presence of desipramine, catechol-O-methyltransferase inhibition had no effect on the clearance of NE, Epi, and DA and decreased Iso clearance by 25%. Desipramine 31-42 catechol O-methyltransferase Oryctolagus cuniculus 44-72 7945993-2 1994 Desipramine induced rapid intracellular degradation of mature acid sphingomyelinase when added to the cells in the micromolar range, concomitantly abolishing the enzyme activity. Desipramine 0-11 sphingomyelin phosphodiesterase 1 Homo sapiens 62-83 7945993-5 1994 From these results we conclude that desipramine and possibly also similarly acting tricyclic antidepressants induce proteolytic degradation of acid sphingomyelinase. Desipramine 36-47 sphingomyelin phosphodiesterase 1 Homo sapiens 143-164 8052414-1 1994 The effect of castration combined with either long-term treatment with the tricyclic antidepressant drug desipramine or the sex steroid 17 beta-estradiol on serotonin responses in area CA1 of the hippocampus of male and female rats was examined. Desipramine 105-116 carbonic anhydrase 1 Rattus norvegicus 185-188 8032878-9 1994 Addition of DMI to the CSF significantly increased perfusate NE levels, but produced no change in LH release. Desipramine 12-15 colony stimulating factor 2 Rattus norvegicus 23-26