PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 32786316-2 2020 Pramipexole is classified as a non-ergoline aminobenzothiazole compound that selectively agonizes the dopamine D2-like receptor subfamily, which includes the D2, D3, and D4 receptor subtypes. Pramipexole 0-11 dopamine receptor D4 Homo sapiens 170-181 33111449-11 2021 Thus, the treatment with pramipexole in the evaluated dose and time tended to alter the expression of the PTPRD protein in the spinal cord, in addition to significantly improving the sleep pattern. Pramipexole 25-36 protein tyrosine phosphatase, receptor type, D Rattus norvegicus 106-111 32750392-0 2020 Lack of correlation between dyskinesia and pallidal serotonin transporter expression-induced by L-Dopa and Pramipexole in hemiparkinsonian rats. Pramipexole 107-118 solute carrier family 6 member 4 Rattus norvegicus 52-73 32750392-8 2020 The number of GP SERT-positive axon varicosities was increased in L-Dopa (p < 0.05) and Pramipexole (p < 0.01) treated rats. Pramipexole 88-99 solute carrier family 6 member 4 Rattus norvegicus 17-21 32819568-0 2020 Dopamine receptor D3 agonist (Pramipexole) reduces morphine-induced cardiac fibrosis. Pramipexole 30-41 dopamine receptor D3 Mus musculus 0-20 32540990-0 2020 Nrf2/HO-1 mediates neuroprotective effects of pramipexole by attenuating oxidative damage and mitochondrial perturbation after traumatic brain injury. Pramipexole 46-57 NFE2 like bZIP transcription factor 2 Rattus norvegicus 0-4 32540990-0 2020 Nrf2/HO-1 mediates neuroprotective effects of pramipexole by attenuating oxidative damage and mitochondrial perturbation after traumatic brain injury. Pramipexole 46-57 heme oxygenase 1 Rattus norvegicus 5-9 32540990-13 2020 Further, PPX treatment reduced the Bax/Bcl2 ratio and translocation of Bax to mitochondria and cytochrome-c to cytosol. Pramipexole 9-12 BCL2 associated X, apoptosis regulator Rattus norvegicus 35-38 32540990-13 2020 Further, PPX treatment reduced the Bax/Bcl2 ratio and translocation of Bax to mitochondria and cytochrome-c to cytosol. Pramipexole 9-12 BCL2, apoptosis regulator Rattus norvegicus 39-43 32540990-13 2020 Further, PPX treatment reduced the Bax/Bcl2 ratio and translocation of Bax to mitochondria and cytochrome-c to cytosol. Pramipexole 9-12 BCL2 associated X, apoptosis regulator Rattus norvegicus 71-74 32540990-14 2020 Finally, PPX treatment greatly accelerated the translocation of Nrf2 to the nucleus and upregulated the HO-1 protein expression. Pramipexole 9-12 NFE2 like bZIP transcription factor 2 Rattus norvegicus 64-68 32540990-14 2020 Finally, PPX treatment greatly accelerated the translocation of Nrf2 to the nucleus and upregulated the HO-1 protein expression. Pramipexole 9-12 heme oxygenase 1 Rattus norvegicus 104-108 31538542-4 2020 In order to shed light on this issue, DRD2- and DRD3-overexpressing cells and drd2 KO, drd3 KO and wild-type mice were treated with the DRD2-DRD3 agonist pramipexole. Pramipexole 154-165 dopamine receptor D2 Mus musculus 78-82 31538542-4 2020 In order to shed light on this issue, DRD2- and DRD3-overexpressing cells and drd2 KO, drd3 KO and wild-type mice were treated with the DRD2-DRD3 agonist pramipexole. Pramipexole 154-165 dopamine receptor D2 Mus musculus 136-140 31538542-4 2020 In order to shed light on this issue, DRD2- and DRD3-overexpressing cells and drd2 KO, drd3 KO and wild-type mice were treated with the DRD2-DRD3 agonist pramipexole. Pramipexole 154-165 dopamine receptor D3 Mus musculus 141-145 31538542-5 2020 The results revealed that pramipexole induces autophagy through MTOR inhibition and a DRD3-dependent but DRD2-independent mechanism. Pramipexole 26-37 mechanistic target of rapamycin kinase Mus musculus 64-68 31538542-5 2020 The results revealed that pramipexole induces autophagy through MTOR inhibition and a DRD3-dependent but DRD2-independent mechanism. Pramipexole 26-37 dopamine receptor D3 Mus musculus 86-90 31538542-5 2020 The results revealed that pramipexole induces autophagy through MTOR inhibition and a DRD3-dependent but DRD2-independent mechanism. Pramipexole 26-37 dopamine receptor D2 Mus musculus 105-109 31811458-12 2020 Moreover, addition of miR-494-3p and silence of BDNF mitigated the effect of PPX on MPP+-induced neurotoxicity. Pramipexole 77-80 brain derived neurotrophic factor Homo sapiens 48-52 32172399-0 2020 Pramipexole Reduces zif-268 mRNA Expression in Brain Structures involved in the Generation of Harmaline-Induced Tremor. Pramipexole 0-11 early growth response 1 Rattus norvegicus 20-27 32172399-3 2020 The aim of the present study was to investigate brain targets for the tremorolytic effect of pramipexole by determination of the early activity-dependent gene zif-268 mRNA expression. Pramipexole 93-104 early growth response 1 Rattus norvegicus 159-166 32172399-9 2020 Pramipexole reversed both the harmaline-induced tremor and the increase in zif-268 mRNA expression in the inferior olive, cerebellar cortex and motor cortex. Pramipexole 0-11 early growth response 1 Rattus norvegicus 75-82 32735501-0 2020 Efficacy of pramipexole combined with levodopa for Parkinson"s disease treatment and their effects on QOL and serum TNF-alpha levels. Pramipexole 12-23 tumor necrosis factor Homo sapiens 116-125 32735501-8 2020 CONCLUSION: Pramipexole combined with levodopa relieved PD symptoms and improved the quality of life of PD patients, potentially by suppressing serum TNF-alpha levels. Pramipexole 12-23 tumor necrosis factor Homo sapiens 150-159 32546033-13 2021 miR-19b was found higher in ropinirole drug used group than that of pramipexole group.Conclusion: This study suggests that serum miR-29c expression level might be potential biomarker in the diagnosis of Turkish Parkinson patients. Pramipexole 68-79 microRNA 29c Homo sapiens 129-136 31811458-13 2020 PPX inhibited MPP+-induced neurotoxicity in SK-N-SH and CHP 212 cells by decreasing miR-494-3p and increasing BDNF, indicating the potential therapeutic effect of PPX on Parkinson"s disease. Pramipexole 0-3 brain derived neurotrophic factor Homo sapiens 110-114 31811458-0 2020 Pramipexole Inhibits MPP+-Induced Neurotoxicity by miR-494-3p/BDNF. Pramipexole 0-11 brain derived neurotrophic factor Homo sapiens 62-66 31811458-13 2020 PPX inhibited MPP+-induced neurotoxicity in SK-N-SH and CHP 212 cells by decreasing miR-494-3p and increasing BDNF, indicating the potential therapeutic effect of PPX on Parkinson"s disease. Pramipexole 163-166 brain derived neurotrophic factor Homo sapiens 110-114 31235613-1 2019 A dopamine D2 receptor agonist, pramipexole, has been found to elicit neuroprotection in patients with Parkinson"s disease and restless leg syndrome. Pramipexole 32-43 dopamine receptor D2 Homo sapiens 2-22 31085186-8 2019 Moreover, both rotigotine and pramipexole shortened the duration of freezing in the fear conditioning test, but only in the mSPS-exposed mice. Pramipexole 30-41 spontaneous seizure Mus musculus 124-128 30291939-5 2018 We report that DEX improved LTP maintenance in CA1 neurons of acute hippocampal slices from aged but not young rats. Pramipexole 15-18 carbonic anhydrase 1 Rattus norvegicus 47-50 31324936-2 2019 OBJECTIVES: This study used computational modelling to characterise the reinforcement learning processes underlying patterns of PRL behaviour observed in SUD and OCD and to show how the dopamine D2/3 receptor agonist pramipexole and the D2/3 antagonist amisulpride affected these responses. Pramipexole 217-228 dopamine receptor D2 Homo sapiens 186-208 30423289-5 2019 The interaction was also increased by pramipexole, a D2R agonist commonly used in the treatment of PD, indicating a possible clinically relevance. Pramipexole 38-49 dopamine receptor D2 Mus musculus 53-56 31008016-2 2019 Pramipexole is a small molecule used in the treatment of idiopathic and uremic RLS. Pramipexole 0-11 RLS1 Homo sapiens 79-82 31008016-6 2019 MATERIALS AND METHODS: Our patient was a 63-year-old man on chronic HD for 5 years who had been treated uneventfully with oral pramipexole for uremic RLS since then. Pramipexole 127-138 RLS1 Homo sapiens 150-153 30033298-1 2018 In this present paper, a simple voltammetric method has been reported for the simultaneous determination of acetaminophen (AC), pramipexole (PRX) and carbamazepine (CBZ) using ZSM-5 nanozeolite-TiO2 nanoparticles composite modified carbon paste electrode (ZSM-5/TiO2/CPE). Pramipexole 141-144 carboxypeptidase E Homo sapiens 267-270 30033298-4 2018 The oxidation peak currents of AC, PRX and CBZ at the surface of ZSM-5/TiO2/CPE were increased dramatically against CPE and their overpotentials decreased. Pramipexole 35-38 carboxypeptidase E Homo sapiens 76-79 29953413-0 2018 CircSNCA downregulation by pramipexole treatment mediates cell apoptosis and autophagy in Parkinson"s disease by targeting miR-7. Pramipexole 27-38 leukocyte immunoglobulin like receptor B1 Homo sapiens 123-128 30033298-4 2018 The oxidation peak currents of AC, PRX and CBZ at the surface of ZSM-5/TiO2/CPE were increased dramatically against CPE and their overpotentials decreased. Pramipexole 35-38 carboxypeptidase E Homo sapiens 116-119 28695622-0 2018 Sleep disturbance by pramipexole is modified by Meis1 in mice. Pramipexole 21-32 Meis homeobox 1 Mus musculus 48-53 28695622-4 2018 We report a detailed sleep analysis of heterozygous Meis1 knockout mice and challenge it with pramipexole, a dopamine agonist used in the treatment of RLS. Pramipexole 94-105 Meis homeobox 1 Mus musculus 52-57 29481916-0 2018 Pramipexole and Fingolimod exert neuroprotection in a mouse model of Parkinson"s disease by activation of sphingosine kinase 1 and Akt kinase. Pramipexole 0-11 sphingosine kinase 1 Mus musculus 106-126 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 18-29 dopamine receptor D3 Mus musculus 52-55 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 18-29 interleukin 1 beta Mus musculus 234-251 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 18-29 interleukin 6 Mus musculus 257-270 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 18-29 brain derived neurotrophic factor Mus musculus 273-277 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 18-29 mitogen-activated protein kinase 3 Mus musculus 283-289 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 18-29 cAMP responsive element binding protein 1 Mus musculus 290-294 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 31-34 dopamine receptor D3 Mus musculus 52-55 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 31-34 interleukin 1 beta Mus musculus 234-251 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 31-34 interleukin 6 Mus musculus 257-270 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 31-34 brain derived neurotrophic factor Mus musculus 273-277 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 31-34 mitogen-activated protein kinase 3 Mus musculus 283-289 29390063-6 2018 Pretreatment with pramipexole (PPX), a preferential D3R agonist, showed antidepressant effects on LPS-induced depression-like behavior through preventing changes in LPS-induced proinflammatory cytokines (tumour necrosis factor-alpha, interleukin-1beta, and interleukin-6), BDNF, and ERK1/2-CREB signaling pathway in the VTA and NAc. Pramipexole 31-34 cAMP responsive element binding protein 1 Mus musculus 290-294 29056363-0 2018 Pramipexole reduces soluble mutant huntingtin and protects striatal neurons through dopamine D3 receptors in a genetic model of Huntington"s disease. Pramipexole 0-11 huntingtin Mus musculus 35-45 29361389-8 2018 The post-hoc analyses including the time of evolution and Pramipexol or Ropinirole treatments, confirmed the independent effect of the DRD3 upon ICDs (p = .028) and ICDARs (p = .041) as well as the interaction between DRD3 and Pramipexol treatment upon ICDARs (OR = 4.60, 95% CI 1.20-17.632, p = .026). Pramipexole 58-68 dopamine receptor D3 Homo sapiens 135-139 29361389-8 2018 The post-hoc analyses including the time of evolution and Pramipexol or Ropinirole treatments, confirmed the independent effect of the DRD3 upon ICDs (p = .028) and ICDARs (p = .041) as well as the interaction between DRD3 and Pramipexol treatment upon ICDARs (OR = 4.60, 95% CI 1.20-17.632, p = .026). Pramipexole 227-237 dopamine receptor D3 Homo sapiens 135-139 29531524-0 2018 Ropinirole and Pramipexole Promote Structural Plasticity in Human iPSC-Derived Dopaminergic Neurons via BDNF and mTOR Signaling. Pramipexole 15-26 brain derived neurotrophic factor Homo sapiens 104-108 29531524-0 2018 Ropinirole and Pramipexole Promote Structural Plasticity in Human iPSC-Derived Dopaminergic Neurons via BDNF and mTOR Signaling. Pramipexole 15-26 mechanistic target of rapamycin kinase Homo sapiens 113-117 29531524-5 2018 Ropinirole and pramipexole produced dose-dependent increases of dendritic arborization and soma size after 3 days of culture, effects antagonized by the selective D3R antagonists SB277011-A and S33084 and by the mTOR pathway kinase inhibitors LY294002 and rapamycin. Pramipexole 15-26 mechanistic target of rapamycin kinase Homo sapiens 212-216 29056363-9 2018 Pramipexole reduced striatal levels of soluble mHTT and increased the size of intranuclear inclusions in R6/1 mice. Pramipexole 0-11 huntingtin Mus musculus 47-51 28631520-0 2017 Striatal changes underlie MPEP-mediated suppression of the acquisition and expression of pramipexole-induced place preference in an alpha-synuclein rat model of Parkinson"s disease. Pramipexole 89-100 synuclein alpha Rattus norvegicus 132-147 28631520-3 2017 We previously demonstrated that the rewarding effect of the D2/D3 agonist pramipexole was enhanced after repeated exposure to L-dopa and alpha-synuclein mediated dopaminergic nigral loss with specific transcriptional signatures suggesting a key involvement of the glutamatergic pathway. Pramipexole 74-85 synuclein alpha Rattus norvegicus 137-152 27817855-8 2017 Our results suggest that genotype in DRD3 Ser9Gly was the main factor determining different doses of DAs and PD patients carrying Gly/Gly genotype require higher doses of pramipexole for effective treatment. Pramipexole 171-182 dopamine receptor D3 Homo sapiens 37-41 26801190-0 2017 Pramipexole, a Dopamine D2/D3 Receptor-Preferring Agonist, Prevents Experimental Autoimmune Encephalomyelitis Development in Mice. Pramipexole 0-11 dopamine receptor D2 Mus musculus 24-38 26801190-3 2017 Herein, we hypothesized that pramipexole (PPX), a dopamine D2/D3 receptor-preferring agonist commonly used to treat Parkinson"s disease (PD), would be a suitable therapeutic drug for EAE. Pramipexole 29-40 dopamine receptor D2 Mus musculus 59-73 26801190-3 2017 Herein, we hypothesized that pramipexole (PPX), a dopamine D2/D3 receptor-preferring agonist commonly used to treat Parkinson"s disease (PD), would be a suitable therapeutic drug for EAE. Pramipexole 42-45 dopamine receptor D2 Mus musculus 59-73 26801190-8 2017 Moreover, PPX inhibited the production of inflammatory cytokines, such as IL-17, IL-1beta, and TNF-alpha in peripheral lymphoid tissue. Pramipexole 10-13 interleukin 1 beta Mus musculus 81-89 26801190-8 2017 Moreover, PPX inhibited the production of inflammatory cytokines, such as IL-17, IL-1beta, and TNF-alpha in peripheral lymphoid tissue. Pramipexole 10-13 tumor necrosis factor Mus musculus 95-104 28845959-11 2017 The main pharmacologic options for RLS management are dopaminergic agonists (eg, pramipexole and ropinirole); gabapentinoids also are good options. Pramipexole 81-92 RLS1 Homo sapiens 35-38 28818004-4 2017 Augmentation, which refers to a paradoxical treatment-related increase in RLS symptoms, has been associated with all three dopamine agonists approved for the treatment of RLS - rotigotine, pramipexole, and ropinirole. Pramipexole 189-200 RLS1 Homo sapiens 74-77 28415529-5 2017 It was found that the TMDS-MWCNT/GCE exhibits good catalytic activity toward oxidation of pramipexole (PPX) drug, leading to a concentration range of 0.8 to 600muM with a detection limit of 0.2muM at 3sigma using the differential pulse voltammetry technique, a sensitivity of 0.084muAmuM-1, and a correlation coefficient of 0.991. Pramipexole 90-101 aminomethyltransferase Homo sapiens 22-36 28415529-5 2017 It was found that the TMDS-MWCNT/GCE exhibits good catalytic activity toward oxidation of pramipexole (PPX) drug, leading to a concentration range of 0.8 to 600muM with a detection limit of 0.2muM at 3sigma using the differential pulse voltammetry technique, a sensitivity of 0.084muAmuM-1, and a correlation coefficient of 0.991. Pramipexole 103-106 aminomethyltransferase Homo sapiens 22-36 27958259-1 2017 Acute hyperkinesia after discontinuation of tramadol in a patient with chronic pain using citalopram and pramipexole for restless legs syndrome (RLS) has not been reported. Pramipexole 105-116 RLS1 Homo sapiens 145-148 27958259-8 2017 Sudden discontinuation of tramadol in a patient under pramipexole for RLS may cause severe, choreatic hyperkinesias for hours, which immediately resolve upon intravenous fentanyl. Pramipexole 54-65 RLS1 Homo sapiens 70-73 27958259-9 2017 In patients under pramipexole for RLS and tramadol and fentanyl for chronic pain, sudden discontinuation of tramadol should be avoided to prevent induction of restless body syndrome. Pramipexole 18-29 RLS1 Homo sapiens 34-37 26239766-1 2015 Pramipexole is a D3 dopamine receptor-preferring agonist indicated for the treatment of Parkinson disease. Pramipexole 0-11 dopamine receptor D3 Homo sapiens 17-37 27562098-4 2016 To improve this picture, since 2009 we introduced the non-ergot dopamine agonist pramipexole as an adjunct to l-dopa therapy in the treatment of the most common causes of BH4 deficiency, 6-pyruvoyl tetrahydropterin synthase (PTPS) deficiency and dihydropteridine reductase (DHPR) deficiency. Pramipexole 81-92 6-pyruvoyltetrahydropterin synthase Homo sapiens 187-223 27562098-4 2016 To improve this picture, since 2009 we introduced the non-ergot dopamine agonist pramipexole as an adjunct to l-dopa therapy in the treatment of the most common causes of BH4 deficiency, 6-pyruvoyl tetrahydropterin synthase (PTPS) deficiency and dihydropteridine reductase (DHPR) deficiency. Pramipexole 81-92 6-pyruvoyltetrahydropterin synthase Homo sapiens 225-229 27026509-0 2016 Pramipexole-Induced Hypothermia Reduces Early Brain Injury via PI3K/AKT/GSK3beta pathway in Subarachnoid Hemorrhage rats. Pramipexole 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 68-71 27026509-0 2016 Pramipexole-Induced Hypothermia Reduces Early Brain Injury via PI3K/AKT/GSK3beta pathway in Subarachnoid Hemorrhage rats. Pramipexole 0-11 glycogen synthase kinase 3 beta Rattus norvegicus 72-80 27026509-8 2016 Pramipexole-induced hypothermia ameliorated SAH-induced brain cell death, blood-brain barrier damage and neurobehavioral deficits in a PI3K/AKT/GSK3beta signaling-dependent manner. Pramipexole 0-11 AKT serine/threonine kinase 1 Rattus norvegicus 140-143 27026509-8 2016 Pramipexole-induced hypothermia ameliorated SAH-induced brain cell death, blood-brain barrier damage and neurobehavioral deficits in a PI3K/AKT/GSK3beta signaling-dependent manner. Pramipexole 0-11 glycogen synthase kinase 3 beta Rattus norvegicus 144-152 27026509-9 2016 Therefore, we may conclude that pramipexole-induced hypothermia could effectively inhibit EBI after SAH in rats via PI3K/AKT/GSK3beta signaling pathway. Pramipexole 32-43 AKT serine/threonine kinase 1 Rattus norvegicus 121-124 27026509-9 2016 Therefore, we may conclude that pramipexole-induced hypothermia could effectively inhibit EBI after SAH in rats via PI3K/AKT/GSK3beta signaling pathway. Pramipexole 32-43 glycogen synthase kinase 3 beta Rattus norvegicus 125-133 26628402-0 2016 Adaptive down-regulation of the serotonin transporter in the 6-hydroxydopamine-induced rat model of preclinical stages of Parkinson"s disease and after chronic pramipexole treatment. Pramipexole 160-171 solute carrier family 6 member 4 Rattus norvegicus 32-53 27488420-4 2016 METHODS: Pergolide (0.001-1 muM) and pramipexole (0.01-200 muM) were administered to 8 day primary murine mesencephalic cultures for 24 h. in the presence or absence of desferal, sulpiride or cycloheximide. Pramipexole 37-48 latexin Homo sapiens 59-62 27216859-7 2016 However, the pramipexole-induced increase in waiting impulsivity observed in both sham and lesioned rats, was more pronounced in HI lesioned rats, which displayed a restricted alpha-synuclein-induced dopaminergic neurodegeneration. Pramipexole 13-24 synuclein alpha Rattus norvegicus 176-191 27588058-0 2016 Effects of pramipexole treatment on the alpha-synuclein content in serum exosomes of Parkinson"s disease patients. Pramipexole 11-22 synuclein alpha Homo sapiens 40-55 27588058-14 2016 The therapeutic effect of pramipexole may be associated with its reducing effect on the relative expression of alpha-synuclein in serum exosomes. Pramipexole 26-37 synuclein alpha Homo sapiens 111-126 27261607-1 2016 Pramipexole is a dopamine D2 receptor agonist indicated for treating Parkinson disorder. Pramipexole 0-11 dopamine receptor D2 Mus musculus 17-37 27261607-10 2016 Altogether, our data suggest that the antidepressant-like effect of pramipexole is dependent on the activation of D2 receptor and inhibition of either NMDA receptors and/or NO-cGMP synthesis. Pramipexole 68-79 dopamine receptor D2 Mus musculus 114-125 25739024-9 2015 Chronic pramipexole increased BDNF mRNA, but decreased trkB mRNA in the VTA in lesioned rats. Pramipexole 8-19 brain-derived neurotrophic factor Rattus norvegicus 30-34 26649942-3 2015 In this study, we report that dopamine D2 and D3 receptor (DRD2 and DRD3) activation by pramipexole and quinpirole could enhance BECN1 transcription and promote autophagy activation in several cell lines, including PC12, MES23.5 and differentiated SH-SY5Y cells, and also in tyrosine hydroxylase positive primary midbrain neurons. Pramipexole 88-99 dopamine receptor D2 Rattus norvegicus 59-63 26649942-3 2015 In this study, we report that dopamine D2 and D3 receptor (DRD2 and DRD3) activation by pramipexole and quinpirole could enhance BECN1 transcription and promote autophagy activation in several cell lines, including PC12, MES23.5 and differentiated SH-SY5Y cells, and also in tyrosine hydroxylase positive primary midbrain neurons. Pramipexole 88-99 dopamine receptor D3 Rattus norvegicus 68-72 26649942-3 2015 In this study, we report that dopamine D2 and D3 receptor (DRD2 and DRD3) activation by pramipexole and quinpirole could enhance BECN1 transcription and promote autophagy activation in several cell lines, including PC12, MES23.5 and differentiated SH-SY5Y cells, and also in tyrosine hydroxylase positive primary midbrain neurons. Pramipexole 88-99 beclin 1 Rattus norvegicus 129-134 26649942-6 2015 More importantly, pramipexole treatment ameliorated the SNCA/alpha-synuclein accumulation in rotenone-treated PC12 cells that overexpress wild-type or A53T mutant SNCA by promoting autophagy flux. Pramipexole 18-29 synuclein alpha Rattus norvegicus 56-60 26649942-6 2015 More importantly, pramipexole treatment ameliorated the SNCA/alpha-synuclein accumulation in rotenone-treated PC12 cells that overexpress wild-type or A53T mutant SNCA by promoting autophagy flux. Pramipexole 18-29 synuclein alpha Rattus norvegicus 61-76 26649942-6 2015 More importantly, pramipexole treatment ameliorated the SNCA/alpha-synuclein accumulation in rotenone-treated PC12 cells that overexpress wild-type or A53T mutant SNCA by promoting autophagy flux. Pramipexole 18-29 synuclein alpha Rattus norvegicus 163-167 26649942-8 2015 The inhibition of SNCA accumulation by pramipexole was attenuated by cotreatment with the DRD2 and DRD3 antagonists and Becn1 siRNAs. Pramipexole 39-50 synuclein alpha Rattus norvegicus 18-22 26649942-8 2015 The inhibition of SNCA accumulation by pramipexole was attenuated by cotreatment with the DRD2 and DRD3 antagonists and Becn1 siRNAs. Pramipexole 39-50 dopamine receptor D2 Rattus norvegicus 90-94 26649942-8 2015 The inhibition of SNCA accumulation by pramipexole was attenuated by cotreatment with the DRD2 and DRD3 antagonists and Becn1 siRNAs. Pramipexole 39-50 dopamine receptor D3 Rattus norvegicus 99-103 26649942-8 2015 The inhibition of SNCA accumulation by pramipexole was attenuated by cotreatment with the DRD2 and DRD3 antagonists and Becn1 siRNAs. Pramipexole 39-50 beclin 1 Rattus norvegicus 120-125 26112716-11 2015 In vitro, in the cultured PBMC treated with 10 mumol/L pramipexole, the Nurr1 mRNA levels were significantly increased by 99.61%, 71.75%, 73.16% in 2, 4, and 8 h, respectively (P < 0.001). Pramipexole 55-66 nuclear receptor subfamily 4 group A member 2 Homo sapiens 72-77 26360835-2 2015 Previously the organic cation transporter 2 (OCT2) was shown to be involved in the uptake of pramipexole by renal tubular cells, while the mechanism underlying efflux into tubular lumen remains unclear. Pramipexole 93-104 solute carrier family 22 member 2 Canis lupus familiaris 15-43 26360835-2 2015 Previously the organic cation transporter 2 (OCT2) was shown to be involved in the uptake of pramipexole by renal tubular cells, while the mechanism underlying efflux into tubular lumen remains unclear. Pramipexole 93-104 solute carrier family 22 member 2 Canis lupus familiaris 45-49 26360835-3 2015 Cimetidine, a potent inhibitor of multidrug and toxin extrusion proteins 1 (MATE1) and 2-K (MATE2-K), decreases renal pramipexole clearance in humans. Pramipexole 118-129 solute carrier family 47 member 1 Homo sapiens 76-81 26360835-3 2015 Cimetidine, a potent inhibitor of multidrug and toxin extrusion proteins 1 (MATE1) and 2-K (MATE2-K), decreases renal pramipexole clearance in humans. Pramipexole 118-129 solute carrier family 47 member 2 Homo sapiens 92-99 26360835-5 2015 Pramipexole uptake was investigated using MDCK or HEK cells overexpressing OCT2, MATE1 or MATE2-K and the respective vector controls (Co). Pramipexole 0-11 solute carrier family 22 member 2 Canis lupus familiaris 75-79 26360835-5 2015 Pramipexole uptake was investigated using MDCK or HEK cells overexpressing OCT2, MATE1 or MATE2-K and the respective vector controls (Co). Pramipexole 0-11 solute carrier family 47 member 1 Homo sapiens 81-86 26360835-5 2015 Pramipexole uptake was investigated using MDCK or HEK cells overexpressing OCT2, MATE1 or MATE2-K and the respective vector controls (Co). Pramipexole 0-11 solute carrier family 47 member 2 Homo sapiens 90-97 26360835-6 2015 Transcellular pramipexole transport was investigated in MDCK cells single- or double-transfected with OCT2 and/or MATE1 and in Co cells, separating a basal from an apical compartment in a model for renal tubular secretion. Pramipexole 14-25 solute carrier family 22 member 2 Canis lupus familiaris 102-106 26360835-6 2015 Transcellular pramipexole transport was investigated in MDCK cells single- or double-transfected with OCT2 and/or MATE1 and in Co cells, separating a basal from an apical compartment in a model for renal tubular secretion. Pramipexole 14-25 solute carrier family 47 member 1 Homo sapiens 114-119 26360835-7 2015 Pramipexole uptake was 1.6-, 1.1-, or 1.6-folds in cells overexpressing OCT2, MATE1 or MATE2-K, respectively as compared to Co cells (p<0.05). Pramipexole 0-11 solute carrier family 22 member 2 Canis lupus familiaris 72-76 26360835-7 2015 Pramipexole uptake was 1.6-, 1.1-, or 1.6-folds in cells overexpressing OCT2, MATE1 or MATE2-K, respectively as compared to Co cells (p<0.05). Pramipexole 0-11 solute carrier family 47 member 1 Homo sapiens 78-83 26360835-7 2015 Pramipexole uptake was 1.6-, 1.1-, or 1.6-folds in cells overexpressing OCT2, MATE1 or MATE2-K, respectively as compared to Co cells (p<0.05). Pramipexole 0-11 solute carrier family 47 member 2 Homo sapiens 87-94 26360835-8 2015 In transcellular transport experiments, intracellular pramipexole accumulation was 1.7-folds in MDCK-OCT2 (p<0.001), and transcellular pramipexole transport was 2.2- and 4.0-folds in MDCK-MATE1 and MDCK-OCT2-MATE1 cells as compared to Co cells (p<0.001). Pramipexole 54-65 solute carrier family 22 member 2 Canis lupus familiaris 96-105 26360835-8 2015 In transcellular transport experiments, intracellular pramipexole accumulation was 1.7-folds in MDCK-OCT2 (p<0.001), and transcellular pramipexole transport was 2.2- and 4.0-folds in MDCK-MATE1 and MDCK-OCT2-MATE1 cells as compared to Co cells (p<0.001). Pramipexole 54-65 solute carrier family 22 member 2 Canis lupus familiaris 101-105 26360835-9 2015 Transcellular pramipexole transport was pH dependent and inhibited by cimetidine with IC50 values of 12muM and 5.5muM in MATE1 and OCT2-MATE1 cells, respectively. Pramipexole 14-25 solute carrier family 47 member 1 Homo sapiens 121-126 26360835-9 2015 Transcellular pramipexole transport was pH dependent and inhibited by cimetidine with IC50 values of 12muM and 5.5muM in MATE1 and OCT2-MATE1 cells, respectively. Pramipexole 14-25 solute carrier family 22 member 2 Canis lupus familiaris 131-135 26360835-9 2015 Transcellular pramipexole transport was pH dependent and inhibited by cimetidine with IC50 values of 12muM and 5.5muM in MATE1 and OCT2-MATE1 cells, respectively. Pramipexole 14-25 solute carrier family 47 member 1 Homo sapiens 136-141 26360835-10 2015 Taken together, coordinate activity of OCT2-mediated uptake and MATE-mediated efflux determines pramipexole renal secretion. Pramipexole 96-107 solute carrier family 22 member 2 Canis lupus familiaris 39-43 26360835-11 2015 Reduced OCT2 or MATE transport activity due to genetic variation or drug-drug interactions may affect pramipexole renal secretion. Pramipexole 102-113 solute carrier family 22 member 2 Canis lupus familiaris 8-12 25739024-9 2015 Chronic pramipexole increased BDNF mRNA, but decreased trkB mRNA in the VTA in lesioned rats. Pramipexole 8-19 neurotrophic receptor tyrosine kinase 2 Rattus norvegicus 55-59 25739024-11 2015 The present study indicates that both the 6-OHDA-induced dopaminergic lesion and chronic pramipexole influence BDNF signalling in limbic structures, which may be related to their pro-depressive and antidepressant activity in rats, respectively. Pramipexole 89-100 brain-derived neurotrophic factor Rattus norvegicus 111-115 25076559-8 2014 Moreover, type 2 iodothyronine deiodinase (Dio2) expression in porcine VICs treated with pergolide was shown, by a global analysis of mRNA, to be markedly increased compared to that induced by pramipexole. Pramipexole 193-204 iodothyronine deiodinase 2 Homo sapiens 43-47 25514384-0 2015 Pramipexole protects dopaminergic neurons through paraplegin against 6-hydroxydopamine. Pramipexole 0-11 SPG7 matrix AAA peptidase subunit, paraplegin Homo sapiens 50-60 25514384-8 2015 However, the D3 receptor agonist, pramipexole, protected TH reactivity and restored paraplegin expression to the control level in the presence of 6-OHDA. Pramipexole 34-45 SPG7 matrix AAA peptidase subunit, paraplegin Homo sapiens 84-94 24965504-4 2014 There was a strong association between renal function (eGFR) and dexpramipexole CLr. Pramipexole 65-79 epidermal growth factor receptor Homo sapiens 55-59 25511804-4 2015 We show that prolonged administration of pramipexole (0.1mg/kg/day, 6 to 21 days), a preferential D3R agonist, leads to a decrease in DA uptake in mouse striatum that reflects a reduction in DAT affinity for DA in the absence of any change in DAT density or subcellular distribution. Pramipexole 41-52 dopamine receptor D3 Mus musculus 98-101 25511804-4 2015 We show that prolonged administration of pramipexole (0.1mg/kg/day, 6 to 21 days), a preferential D3R agonist, leads to a decrease in DA uptake in mouse striatum that reflects a reduction in DAT affinity for DA in the absence of any change in DAT density or subcellular distribution. Pramipexole 41-52 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 191-194 25511804-4 2015 We show that prolonged administration of pramipexole (0.1mg/kg/day, 6 to 21 days), a preferential D3R agonist, leads to a decrease in DA uptake in mouse striatum that reflects a reduction in DAT affinity for DA in the absence of any change in DAT density or subcellular distribution. Pramipexole 41-52 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 243-246 25511804-5 2015 The effect of pramipexole was absent in mice with genetically-deleted D3R (D3R(-/-)), yet unaffected in mice genetically deprived of D2R (D2R(-/-)). Pramipexole 14-25 dopamine receptor D3 Mus musculus 70-73 25511804-5 2015 The effect of pramipexole was absent in mice with genetically-deleted D3R (D3R(-/-)), yet unaffected in mice genetically deprived of D2R (D2R(-/-)). Pramipexole 14-25 dopamine receptor D3 Mus musculus 75-78 25511804-6 2015 Pramipexole treatment induced a physical interaction between D3R and DAT, as assessed by co-immunoprecipitation and in situ proximity ligation assay. Pramipexole 0-11 dopamine receptor D3 Mus musculus 61-72 25076559-10 2014 CONCLUSION: There were substantial differences (increased [3H]thymidine incorporation, and Dio2 expression) between pergolide and pramipexole, which might correlate with the mechanism of heart valve disease onset. Pramipexole 130-141 iodothyronine deiodinase 2 Homo sapiens 91-95 23631878-4 2013 Dopamine and dopamine D2-like receptor agonists such as pramipexole and quinpirole reduced the formation of TRAP-positive multi-nucleated cells, cathepsin K mRNA expression, and pit formation area in vitro. Pramipexole 56-67 cathepsin K Homo sapiens 145-156 24289818-7 2014 Among the studied drugs, aripiprazole and pramipexole were found to be the most active one for hCA I (IC50: 3.64 and 5.37 muM) and hCA II (IC50: 4.16 and 4.81 muM) activity, respectively. Pramipexole 42-53 carbonic anhydrase 1 Homo sapiens 95-100 24289818-7 2014 Among the studied drugs, aripiprazole and pramipexole were found to be the most active one for hCA I (IC50: 3.64 and 5.37 muM) and hCA II (IC50: 4.16 and 4.81 muM) activity, respectively. Pramipexole 42-53 latexin Homo sapiens 122-125 24289818-7 2014 Among the studied drugs, aripiprazole and pramipexole were found to be the most active one for hCA I (IC50: 3.64 and 5.37 muM) and hCA II (IC50: 4.16 and 4.81 muM) activity, respectively. Pramipexole 42-53 carbonic anhydrase 2 Homo sapiens 131-137 23681749-0 2013 Pramipexole reduces phosphorylation of alpha-synuclein at serine-129. Pramipexole 0-11 synuclein alpha Homo sapiens 39-54 23681749-7 2013 The present study demonstrates that PPX inhibits the phosphorylation of alpha-synuclein and that this is independent of dopamine receptor activation. Pramipexole 36-39 synuclein alpha Homo sapiens 72-87 23681749-10 2013 Thus, PPX decreases the phosphorylation of alpha-synuclein, and this mechanism may contribute to its protective properties in PD models. Pramipexole 6-9 synuclein alpha Homo sapiens 43-58 23631981-1 2013 A previous clinical trial studied the effect of long-term treatment with levodopa (LD) or the dopamine agonist pramipexole (PPX) on disease progression in Parkinson disease using SPECT with the dopamine transporter (DAT)-radioligand [(123)I]beta-CIT as surrogate marker. Pramipexole 111-122 solute carrier family 6 member 3 Homo sapiens 194-214 23631981-1 2013 A previous clinical trial studied the effect of long-term treatment with levodopa (LD) or the dopamine agonist pramipexole (PPX) on disease progression in Parkinson disease using SPECT with the dopamine transporter (DAT)-radioligand [(123)I]beta-CIT as surrogate marker. Pramipexole 111-122 solute carrier family 6 member 3 Homo sapiens 216-219 24116342-4 2013 METHODS: We present an illustrative patient with concomitant RLS and migraine who showed improvement in her headache frequency and RLS symptoms following immediate-release pramipexole (P-IR) treatment and provide review results from the medical records of patients who experienced both migraines and RLS in our previous cross-sectional study. Pramipexole 172-183 pirin Homo sapiens 184-189 24289818-7 2014 Among the studied drugs, aripiprazole and pramipexole were found to be the most active one for hCA I (IC50: 3.64 and 5.37 muM) and hCA II (IC50: 4.16 and 4.81 muM) activity, respectively. Pramipexole 42-53 latexin Homo sapiens 159-162 23643745-6 2013 Amisulpride also attenuated the hypolocomotion induced by the D2/D3 receptor agonist pramipexole and dopamine precursor l-3,4-dihydroxyphenylalanine, whereas raclopride (and S33084) worsened it. Pramipexole 85-96 dopamine receptor D2 Mus musculus 62-76 23241649-5 2013 Furthermore, we tested the sensitivity of PPX-induced CFL to CIM and to the 5HT2c antagonist SB242084. Pramipexole 42-45 5-hydroxytryptamine receptor 2C Rattus norvegicus 76-81 23483198-1 2013 RATIONALE: Pramipexole, a D2/D3 dopamine receptor agonist, has been implicated in the development of impulse control disorders in patients with Parkinson"s disease. Pramipexole 11-22 dopamine receptor D3 Homo sapiens 29-49 23950645-3 2013 In this meta-analysis, we summarized the efficacy and tolerability of pramipexole in treatment for primary RLS. Pramipexole 70-81 RLS1 Homo sapiens 107-110 23357086-2 2013 Recent studies have reported that the dopamine D2/D3 receptor agonist pramipexole exerts an antidepressant-like effect in the chronic mild stress model and in the behavioral despair model, suggesting dopamine D3 receptor may be an important target for antidepressant actions. Pramipexole 70-81 dopamine receptor D3 Mus musculus 200-220 23950645-2 2013 Pramipexole, a potent dopamine D2/3 agonist, is recommended as "effective" in the short-term and "possibly effective" in the long-term treatment of primary RLS in the European guidelines on management of RLS. Pramipexole 0-11 RLS1 Homo sapiens 156-159 23311775-0 2013 Age-dependent dystonia in striatal Ggamma7 deficient mice is reversed by the dopamine D2 receptor agonist pramipexole. Pramipexole 106-117 dopamine receptor D2 Mus musculus 77-97 23311775-6 2013 Taking advantage of the finding of D2R down-regulation in Ggamma7 knockout mice and the dopamine-mediated synergistic relationship in the control of locomotion between D2R-striatopallidal and D1R-stritonigral neurons, we hypothesized that D2-agonist pramipexole would reverse behavioral dyskinesia caused by defective D1R/Galphaolf signaling. Pramipexole 250-261 dopamine receptor D1 Mus musculus 192-195 23950645-2 2013 Pramipexole, a potent dopamine D2/3 agonist, is recommended as "effective" in the short-term and "possibly effective" in the long-term treatment of primary RLS in the European guidelines on management of RLS. Pramipexole 0-11 RLS1 Homo sapiens 204-207 23950645-4 2013 Results of this meta-analysis showed a favorable effect of pramipexole versus placebo on RLS symptoms (mean change on International RLS Study Group Rating Scale [IRLS] score: mean difference [MD] = -5.96; 95% confidence interval [CI]: -7.79 to -4.41, P < 0.00001) and sleep quality (pooled standard mean difference [SMD] = -0.48, 95% CI: -0.61 to -0.35, P < 0.00001). Pramipexole 59-70 RLS1 Homo sapiens 89-92 23950645-4 2013 Results of this meta-analysis showed a favorable effect of pramipexole versus placebo on RLS symptoms (mean change on International RLS Study Group Rating Scale [IRLS] score: mean difference [MD] = -5.96; 95% confidence interval [CI]: -7.79 to -4.41, P < 0.00001) and sleep quality (pooled standard mean difference [SMD] = -0.48, 95% CI: -0.61 to -0.35, P < 0.00001). Pramipexole 59-70 RLS1 Homo sapiens 132-135 22325981-4 2012 Recently, we demonstrated that the dopamine agonist pramipexole improves the therapeutic effect of L-dopa in 6-pyruvoyl tetrahydropterin synthase (PTPS) deficiency, the most common disorder of BH4 metabolism. Pramipexole 52-63 6-pyruvoyltetrahydropterin synthase Homo sapiens 147-151 22153972-2 2012 Pramipexole was generally well-tolerated (82% trial-completion), and yielded greater decreases in PANSS-total scores (drug/placebo=2.1; p=0.04), with similar decreases in PANSS positive and negative scores and 6.7-fold greater reduction of serum prolactin concentrations compared to placebo. Pramipexole 0-11 prolactin Homo sapiens 246-255 22808933-8 2012 Moreover, further studies should investigate the extended-release formulation of ropinirole and pramipexole in RLS patients affected by all day long distressing symptoms. Pramipexole 96-107 RLS1 Homo sapiens 111-114 22333559-7 2012 Fourteen patients with RBD (80.0%) achieved symptomatic improvement of RBD with pramipexole treatment, which reduced REM density and PLM index during non-REM sleep despite the unchanged amount of RWA. Pramipexole 80-91 FXYD domain containing ion transport regulator 1 Homo sapiens 133-136 22227455-1 2012 BACKGROUND: In rats, prepulse inhibition (PPI) of acoustic startle is disrupted by systemic administration of dopaminergic agonists, such as the dopamine D3 receptor (D3R)-preferential agonist pramipexole (PPX). Pramipexole 193-204 dopamine receptor D3 Rattus norvegicus 145-165 22227455-1 2012 BACKGROUND: In rats, prepulse inhibition (PPI) of acoustic startle is disrupted by systemic administration of dopaminergic agonists, such as the dopamine D3 receptor (D3R)-preferential agonist pramipexole (PPX). Pramipexole 206-209 dopamine receptor D3 Rattus norvegicus 145-165 22101745-7 2011 RESULTS: Both pramipexole and dual-release L/B were effective in reducing PLM and RLS symptoms. Pramipexole 14-25 FXYD domain containing ion transport regulator 1 Homo sapiens 74-77 22023785-10 2012 CONCLUSION: Chronic PPX administration led to a net enhancement in DA and 5-HT neurotransmission, as indicated by the increased tonic activation of postsynaptic D2 and 5-HT1A receptors in forebrain structures. Pramipexole 20-23 5-hydroxytryptamine receptor 1A Rattus norvegicus 168-174 22986363-0 2012 Pramipexole upregulates dopamine receptor D2 and D3 expression in rat striatum. Pramipexole 0-11 solute carrier family 3 member 1 Rattus norvegicus 42-51 23391874-2 2012 Our previous study demonstrated that pergolide and pramipexole stimulated elevation of plasma arginine vasopressin (AVP) levels in some patients with Parkinson"s disease (PD), but that levodopa/carbidopa (300/30 mg/day) did not affect plasma AVP levels in treatment-naive PD patients. Pramipexole 51-62 arginine vasopressin Homo sapiens 103-114 22467109-2 2012 Our previous study demonstrated that in some patients with Parkinson"s disease (PD), pergolide and pramipexole stimulate elevations of plasma arginine vasopressin (AVP) levels even at doses that are lower than the ordinary maintenance dose. Pramipexole 99-110 arginine vasopressin Homo sapiens 151-162 21272591-8 2011 A 24 h treatment with PPX does not modify the morphology of neurosphere-derived cells, but causes an increase of glial fibrillary acidic protein (GFAP)-positive cells, an effect sensitive to both D2 and D3 antagonism. Pramipexole 22-25 glial fibrillary acidic protein Mus musculus 113-144 21683731-2 2011 In mice, the dopamine D3 receptor-preferential agonist pramipexole (PPX) alters locomotor activity in a biphasic manner at doses that have no effect on PPI. Pramipexole 55-66 dopamine receptor D3 Mus musculus 13-33 21683731-2 2011 In mice, the dopamine D3 receptor-preferential agonist pramipexole (PPX) alters locomotor activity in a biphasic manner at doses that have no effect on PPI. Pramipexole 68-71 dopamine receptor D3 Mus musculus 13-33 21892895-8 2011 Pramipexole has a very low affinity for serotoninergic 5-HT2A and 5-HT2B receptors, as well as D1-type receptors. Pramipexole 0-11 5-hydroxytryptamine receptor 2A Homo sapiens 55-61 21272591-8 2011 A 24 h treatment with PPX does not modify the morphology of neurosphere-derived cells, but causes an increase of glial fibrillary acidic protein (GFAP)-positive cells, an effect sensitive to both D2 and D3 antagonism. Pramipexole 22-25 glial fibrillary acidic protein Mus musculus 146-150 21408026-1 2011 The purpose of this study was to determine the binding sites of pramipexole in extrastriatal dopaminergic regions because its antidepressive effects have been speculated to occur by activating the dopamine D(2) receptor subfamily in extrastriatal areas. Pramipexole 64-75 dopamine receptor D2 Homo sapiens 197-219 20925067-1 2011 To compare the safety and efficacy of low dosages of pramipexole given twice daily (bid) in early Parkinson"s disease (PD) with those of a standard 3 times daily (tid) regimen in a randomized, double-blind, placebo controlled trial involving 311 early PD patients not receiving dopaminergic treatment. Pramipexole 53-64 BH3 interacting domain death agonist Homo sapiens 84-87 20680652-1 2011 Substrates for the Organic Cation Transporter 1, encoded by the SLC22A1 gene, are metformin, amantadine, pramipexole, and, possibly, levodopa. Pramipexole 105-116 solute carrier family 22 member 1 Homo sapiens 19-47 20680652-1 2011 Substrates for the Organic Cation Transporter 1, encoded by the SLC22A1 gene, are metformin, amantadine, pramipexole, and, possibly, levodopa. Pramipexole 105-116 solute carrier family 22 member 1 Homo sapiens 64-71 20697890-3 2010 Here we used multi-tracer positron emission tomography imaging and a unilateral 6-hydroxydopamine (6-OHDA) rat model of PD to evaluate in vivo the effects of chronic levodopa and pramipexole treatments on measurements of vesicular monoamine transporter type 2 (VMAT2), dopamine transporter (DAT) levels, and on levodopa-induced changes in synaptic DA levels [Delta(DA)]. Pramipexole 179-190 solute carrier family 18 member A2 Rattus norvegicus 221-259 20965780-0 2011 Randomized trial of pramipexole for patients with restless legs syndrome (RLS) and RLS-related impairment of mood. Pramipexole 20-31 RLS1 Homo sapiens 74-77 20965780-0 2011 Randomized trial of pramipexole for patients with restless legs syndrome (RLS) and RLS-related impairment of mood. Pramipexole 20-31 RLS1 Homo sapiens 83-86 20965780-2 2011 We investigated the change of RLS-related mood impairment during treatment of RLS with pramipexole, a dopamine D(3)/D(2) agonist. Pramipexole 87-98 RLS1 Homo sapiens 30-33 20965780-2 2011 We investigated the change of RLS-related mood impairment during treatment of RLS with pramipexole, a dopamine D(3)/D(2) agonist. Pramipexole 87-98 RLS1 Homo sapiens 78-81 20965780-10 2011 CONCLUSIONS: In patients with RLS-related mood disturbance, pramipexole improved RLS while also improving RLS-related mood impairment. Pramipexole 60-71 RLS1 Homo sapiens 30-33 20965780-10 2011 CONCLUSIONS: In patients with RLS-related mood disturbance, pramipexole improved RLS while also improving RLS-related mood impairment. Pramipexole 60-71 RLS1 Homo sapiens 81-84 20965780-10 2011 CONCLUSIONS: In patients with RLS-related mood disturbance, pramipexole improved RLS while also improving RLS-related mood impairment. Pramipexole 60-71 RLS1 Homo sapiens 81-84 21044864-12 2011 CONCLUSIONS: Pramipexole produced long-term improvement of the RLS-like symptoms in our proposed animal model, caused a partial recovery of spinal iron deficiency, and modestly increased D1 receptor affinity. Pramipexole 13-24 dopamine receptor D1 Mus musculus 187-198 20697890-3 2010 Here we used multi-tracer positron emission tomography imaging and a unilateral 6-hydroxydopamine (6-OHDA) rat model of PD to evaluate in vivo the effects of chronic levodopa and pramipexole treatments on measurements of vesicular monoamine transporter type 2 (VMAT2), dopamine transporter (DAT) levels, and on levodopa-induced changes in synaptic DA levels [Delta(DA)]. Pramipexole 179-190 solute carrier family 18 member A2 Rattus norvegicus 261-266 20697890-3 2010 Here we used multi-tracer positron emission tomography imaging and a unilateral 6-hydroxydopamine (6-OHDA) rat model of PD to evaluate in vivo the effects of chronic levodopa and pramipexole treatments on measurements of vesicular monoamine transporter type 2 (VMAT2), dopamine transporter (DAT) levels, and on levodopa-induced changes in synaptic DA levels [Delta(DA)]. Pramipexole 179-190 solute carrier family 6 member 3 Rattus norvegicus 269-289 20697890-3 2010 Here we used multi-tracer positron emission tomography imaging and a unilateral 6-hydroxydopamine (6-OHDA) rat model of PD to evaluate in vivo the effects of chronic levodopa and pramipexole treatments on measurements of vesicular monoamine transporter type 2 (VMAT2), dopamine transporter (DAT) levels, and on levodopa-induced changes in synaptic DA levels [Delta(DA)]. Pramipexole 179-190 solute carrier family 6 member 3 Rattus norvegicus 291-294 19396436-0 2009 Association of the DRD2 and DRD3 polymorphisms with response to pramipexole in Parkinson"s disease patients. Pramipexole 64-75 dopamine receptor D2 Homo sapiens 19-23 20123564-7 2009 A placebo-controlled study of pramipexole in dPD is ongoing. Pramipexole 30-41 dachs Drosophila melanogaster 45-48 20021407-5 2009 Six antagonists, Bromocriptine, Cabergoline, Etilevodopa, Lysuride, Melevodopa and Pramipexole, were found more potent for binding and the active amino acids residues were identified (http://www.rcsb.org/pdb/) in A2A receptor. Pramipexole 83-94 immunoglobulin kappa variable 2D-29 Homo sapiens 213-216 20635141-1 2010 Pramipexole (PPX), a dopamine (DA) receptor D3 preferring agonist, has been used as monotherapy or adjunct therapy to treat Parkinson"s disease (PD) for many years. Pramipexole 0-11 dopamine receptor D3 Mus musculus 21-46 20635141-1 2010 Pramipexole (PPX), a dopamine (DA) receptor D3 preferring agonist, has been used as monotherapy or adjunct therapy to treat Parkinson"s disease (PD) for many years. Pramipexole 13-16 dopamine receptor D3 Mus musculus 21-46 20496886-2 2010 In rats, rOct1 and rOct2 contribute toward pramipexole excretion into urine. Pramipexole 43-54 solute carrier family 22 member 1 Rattus norvegicus 9-14 20496886-2 2010 In rats, rOct1 and rOct2 contribute toward pramipexole excretion into urine. Pramipexole 43-54 solute carrier family 22 member 2 Rattus norvegicus 19-24 20496886-3 2010 The objective of this study was to assess whether pramipexole is a substrate for human OCT1-3. Pramipexole 50-61 solute carrier family 22 member 1 Homo sapiens 87-93 20496886-5 2010 hOCT2 transported pramipexole in a high affinity manner (K(t) = 15.4 +/- 4.1 microM, J(max) = 0.476 +/- 0.028 pmol/s/cm(2)). Pramipexole 18-29 POU class 2 homeobox 2 Homo sapiens 0-5 20496886-6 2010 hOCT3 transported pramipexole in a low affinity manner (K(t) = 138 +/- 31 microM, J(max) = 1.10 +/- 0.08 pmol/s/cm(2)). Pramipexole 18-29 solute carrier family 22 member 3 Homo sapiens 0-5 20496886-8 2010 The human intestinal absorption of pramipexole may involve transport by OCT3 and possibly OCT2. Pramipexole 35-46 solute carrier family 22 member 3 Homo sapiens 72-76 20496886-8 2010 The human intestinal absorption of pramipexole may involve transport by OCT3 and possibly OCT2. Pramipexole 35-46 POU class 2 homeobox 2 Homo sapiens 90-94 20496886-9 2010 OCT2- and OCT3-mediated transport of pramipexole have implications in the drug"s elimination from the kidney and distribution in the brain, respectively. Pramipexole 37-48 POU class 2 homeobox 2 Homo sapiens 0-4 20496886-9 2010 OCT2- and OCT3-mediated transport of pramipexole have implications in the drug"s elimination from the kidney and distribution in the brain, respectively. Pramipexole 37-48 solute carrier family 22 member 3 Homo sapiens 10-14 20122926-4 2010 Indeed, the dopamine precursor, l-dopa (1-10 mg/kg), and the D(3)/D(2) receptor agonist pramipexole (0.0001-0.001 mg/kg) promoted stepping activity in the drag test (a test for akinesia/bradykinesia). Pramipexole 88-99 dopamine receptor D2 Mus musculus 66-79 20385162-1 2010 The preferential dopamine D3 receptor agonist pramipexole (PRA) disrupts prepulse inhibition (PPI) of acoustic startle, an operational measure of sensorimotor gating, in rats. Pramipexole 46-57 dopamine receptor D3 Rattus norvegicus 17-37 19962941-5 2010 In all patients, variables on SIT did not show any differences between the two groups, whereas a significant improvement was shown in the pramipexole group compared with the placebo group for patients with a SIT-PLM index at baseline >or=15. Pramipexole 138-149 FXYD domain containing ion transport regulator 1 Homo sapiens 212-215 20120624-11 2010 In the RLS treatment group, a statistically significant dose effect was found for pramipexole in those subjects confirmed to have ICDs by both the questionnaire and phone interview. Pramipexole 82-93 RLS1 Homo sapiens 7-10 19647776-7 2009 Pramipexole inhibited rotenone-induced DA neuronal death and motor deficits, and reduced immunoreactivity for alpha-synuclein. Pramipexole 0-11 synuclein, alpha Mus musculus 110-125 19647776-8 2009 In addition, pramipexole inhibited the in vitro oligomerization of human wild-type alpha-synuclein by H(2)O(2)plus cytochrome c. Pramipexole 13-24 synuclein, alpha Mus musculus 83-98 19647776-8 2009 In addition, pramipexole inhibited the in vitro oligomerization of human wild-type alpha-synuclein by H(2)O(2)plus cytochrome c. Pramipexole 13-24 cytochrome c, somatic Homo sapiens 115-127 19647776-12 2009 Furthermore, pramipexole increased Bcl-2 immunoreactivity in DA neurons in the SNpc. Pramipexole 13-24 B cell leukemia/lymphoma 2 Mus musculus 35-40 19647776-13 2009 These results suggest that pramipexole may protect DA neurons against exposure to rotenone by chronic oral administration, and this effect is mediated by multiple functions including scavenging of *OH and induction of Bcl-2 protein. Pramipexole 27-38 B cell leukemia/lymphoma 2 Mus musculus 218-223 20161553-2 2009 The standard treatment recommendations for these disorders are pharmacologic; most recently both conditions are most typically managed with pramipexole or ropinerole, which are FDA approved for the treatment of RLS. Pramipexole 140-151 RLS1 Homo sapiens 211-214 19765187-7 2009 Treatment with either l-dopa or PPX in MPTP-treated mice led to significantly decreased expressions of JNK phosphorylation, Bax, and cytochrome c and to an increased level of Bcl-2 expression with a similar degree, compared with the levels in MPTP-only treated mice. Pramipexole 32-35 mitogen-activated protein kinase 8 Mus musculus 103-106 19765187-7 2009 Treatment with either l-dopa or PPX in MPTP-treated mice led to significantly decreased expressions of JNK phosphorylation, Bax, and cytochrome c and to an increased level of Bcl-2 expression with a similar degree, compared with the levels in MPTP-only treated mice. Pramipexole 32-35 BCL2-associated X protein Mus musculus 124-127 19765187-7 2009 Treatment with either l-dopa or PPX in MPTP-treated mice led to significantly decreased expressions of JNK phosphorylation, Bax, and cytochrome c and to an increased level of Bcl-2 expression with a similar degree, compared with the levels in MPTP-only treated mice. Pramipexole 32-35 B cell leukemia/lymphoma 2 Mus musculus 175-180 19704083-1 2009 OBJECTIVE: To report the efficacy, tolerability, and safety of the dopamine agonist pramipexole in a series of 5 patients affected by inherited 6-pyruvoyl tetrahydropterin synthase (PTPS) deficiency and needing l-3,4 dihydroxyphenylalanine (l-dopa) therapy. Pramipexole 84-95 6-pyruvoyltetrahydropterin synthase Homo sapiens 144-180 19704083-1 2009 OBJECTIVE: To report the efficacy, tolerability, and safety of the dopamine agonist pramipexole in a series of 5 patients affected by inherited 6-pyruvoyl tetrahydropterin synthase (PTPS) deficiency and needing l-3,4 dihydroxyphenylalanine (l-dopa) therapy. Pramipexole 84-95 6-pyruvoyltetrahydropterin synthase Homo sapiens 182-186 19704083-12 2009 CONCLUSIONS: The addition of pramipexole to the treatment of 6-pyruvoyl tetrahydropterin synthase deficiency improves the results of l-3,4 dihydroxyphenylalanine therapy. Pramipexole 29-40 6-pyruvoyltetrahydropterin synthase Homo sapiens 61-97 19396436-0 2009 Association of the DRD2 and DRD3 polymorphisms with response to pramipexole in Parkinson"s disease patients. Pramipexole 64-75 dopamine receptor D3 Homo sapiens 28-32 19396436-1 2009 OBJECTIVE: To evaluate the impact of the DRD2 TaqIA and DRD3 Ser9Gly polymorphisms on the efficacy of pramipexole in treating patients with Parkinson"s disease (PD). Pramipexole 102-113 dopamine receptor D2 Homo sapiens 41-45 19396436-1 2009 OBJECTIVE: To evaluate the impact of the DRD2 TaqIA and DRD3 Ser9Gly polymorphisms on the efficacy of pramipexole in treating patients with Parkinson"s disease (PD). Pramipexole 102-113 dopamine receptor D3 Homo sapiens 56-60 19396436-7 2009 When the subjects were grouped by the DRD3 Ser9Gly polymorphism, the response rates for pramipexole treatment were significantly higher in the Ser/Ser group (60%) than in the group containing the Gly allele (13%). Pramipexole 88-99 dopamine receptor D3 Homo sapiens 38-42 19396436-8 2009 There was a significant association between the DRD3 Ser9Gly polymorphism and response rate to pramipexole in PD patients (P = 0.024). Pramipexole 95-106 dopamine receptor D3 Homo sapiens 48-52 19396436-10 2009 CONCLUSIONS: DRD3 Ser9Gly polymorphisms are significantly associated with the therapeutic efficacy of pramipexole in Chinese patients with PD. Pramipexole 102-113 dopamine receptor D3 Homo sapiens 13-17 18455202-4 2008 Dopamine, as well as the dopamine receptor agonists pramipexole and quinpirole, acted as full agonists in the assay as reflected by their ability to elicit marked concentration dependent increases in the BRET signal signifying beta-arrestin2 recruitment to the D2 receptor. Pramipexole 52-63 arrestin beta 2 Homo sapiens 227-241 21687007-7 2009 Accordingly, pramipexole has a higher selectivity for D4 receptor than other dopamine agonists. Pramipexole 13-24 dopamine receptor D4 Homo sapiens 54-65 18598736-4 2008 Among the antiparkinsonian drugs tested, levodopa, bromocriptine, pergolide and pramipexole were ABCB1 substrates. Pramipexole 80-91 ATP binding cassette subfamily B member 1A Rattus norvegicus 97-102 18555604-4 2008 Pretreatment with 100 microM pramipexole rescued TH-positive neurons and MAP2-positive neurons from the toxicity of lactacystin. Pramipexole 29-40 microtubule associated protein 2 Homo sapiens 73-77 18555604-10 2008 Pramipexole increased the levels of brain derived neurotrophic factor (BDNF) in the conditioned medium of astrocyte cultures. Pramipexole 0-11 brain derived neurotrophic factor Homo sapiens 36-69 18555604-10 2008 Pramipexole increased the levels of brain derived neurotrophic factor (BDNF) in the conditioned medium of astrocyte cultures. Pramipexole 0-11 brain derived neurotrophic factor Homo sapiens 71-75 18555604-12 2008 We demonstrated that pramipexole had the protective effect against lactacystin toxicity, mediated by a neurotrophic effect of astrocyte-produced factors including BDNF. Pramipexole 21-32 brain derived neurotrophic factor Homo sapiens 163-167 18611302-4 2008 Although two dopamine agonists, ropinirole and pramipexole, have been approved by the FDA for the treatment of RLS and are currently the first-line treatment for daily symptoms, there is very little information on the teratogenic risks of these new medications. Pramipexole 47-58 RLS1 Homo sapiens 111-114 19220275-1 2009 AIMS: To assess the sleepiness induced by pramipexole, a D2/D3-dopamine receptor agonist commonly used in Parkinson"s disease and restless legs syndrome, without the problem of the confounding factors related to the disease. Pramipexole 42-53 dopamine receptor D3 Homo sapiens 60-80 19750232-8 2009 The first line treatment for idiopathic RLS is represented by dopamine agonists, in particular nonergot-derived ropinirole and pramipexole, whereas ergot dopamine agonists (cabergoline and pergolide) are no longer in first-line use given the risks of cardiac valvulopathy. Pramipexole 127-138 RLS1 Homo sapiens 40-43 20666116-3 2009 The dopamine agonists ropinirole and pramipexole are currently considered to be the agents of choice for the treatment of RLS. Pramipexole 37-48 RLS1 Homo sapiens 122-125 18952497-0 2008 Effect of pramipexole on RLS symptoms and sleep: a randomized, double-blind, placebo-controlled trial. Pramipexole 10-21 RLS1 Homo sapiens 25-28 18952497-2 2008 Our goal was to test the ability of pramipexole to improve sleep in RLS patients and to reconfirm its efficacy for primary RLS symptoms. Pramipexole 36-47 RLS1 Homo sapiens 68-71 18952497-10 2008 CONCLUSIONS: Pramipexole is effective and well-tolerated for RLS and related sleep disturbance. Pramipexole 13-24 RLS1 Homo sapiens 61-64 18537487-9 2008 Several large, double-blind, placebo-controlled studies have demonstrated that dopamine agonists, such as ropinirole and pramipexole, are an efficacious first-line therapy for the treatment of RLS symptoms. Pramipexole 121-132 RLS1 Homo sapiens 193-196 17578485-10 2007 Pramipexole reduced prolactin and increased GH concentrations. Pramipexole 0-11 prolactin Homo sapiens 20-29 17578485-10 2007 Pramipexole reduced prolactin and increased GH concentrations. Pramipexole 0-11 growth hormone 1 Homo sapiens 44-46 17534955-2 2007 In the CALM-PD trial, subjects were initially randomized to levodopa or pramipexole and could later add levodopa if needed. Pramipexole 72-83 synaptosome associated protein 91 Homo sapiens 7-11 16802163-7 2006 RESULTS: Pramipexole reduced alertness and pupillary light reflex response amplitude, tended to reduce core temperature, reduced prolactin levels and increased GH levels. Pramipexole 9-20 growth hormone 1 Homo sapiens 160-162 17356338-5 2007 A subsequent therapeutic trial with pramipexole, a dopamine agonist with preferential D3 dopamine receptor activity, resulted in the recurrence of PG. Pramipexole 36-47 dopamine receptor D3 Homo sapiens 86-106 17161393-3 2007 Pramipexole, a dopamine D2/D3 receptor agonist used clinically in the treatment of Parkinson disease, did not affect glutamate-induced calcium influx but blocked dopaminergic neuronal death induced by glutamate. Pramipexole 0-11 dopamine receptor D2 Homo sapiens 15-38 16401653-8 2006 Pramipexole reduced alertness, caused pupil dilatation, increased heart rate, reduced prolactin and thyroid stimulating hormone, and increased growth hormone level. Pramipexole 0-11 prolactin Homo sapiens 86-95 16401653-8 2006 Pramipexole reduced alertness, caused pupil dilatation, increased heart rate, reduced prolactin and thyroid stimulating hormone, and increased growth hormone level. Pramipexole 0-11 growth hormone 1 Homo sapiens 143-157 16930379-4 2006 Amongst the non-ergoline dopamine agonists, pramipexole has been reported to reduce RLS symptoms, but robust trial data are limited. Pramipexole 44-55 RLS1 Homo sapiens 84-87 16982285-9 2006 The occurrence of dyskinesia may be linked to stimulation of the dopamine D(1) receptor, for which cabergoline and pergolide have a similar and relatively high affinity; bromocriptine, pramipexole, and ropinirole have been associated with a lower risk of dyskinesias. Pramipexole 185-196 dopamine receptor D1 Homo sapiens 65-87 16675366-14 2006 The dopamine agonists used to treat Parkinson"s disease--bromocriptine, pergolide, pramipexole, ropinerole--usually reverse antipsychotic-induced prolactin increases without compromising psychiatric effectiveness. Pramipexole 83-94 prolactin Homo sapiens 146-155 16963794-5 2006 Moreover, progesterone and BD 1047 (a sigma(1) receptor antagonist) counteracted the antidepressant-like effect induced by co-administration of pramipexole and sertraline (but not pramipexole and fluoxetine). Pramipexole 144-155 sigma non-opioid intracellular receptor 1 Rattus norvegicus 38-55 16362428-6 2006 In PC12 cells, pramipexole inhibited H2O2-induced lactate dehydrogenase (LDH) leakage, as well as H2O2-induced cytochrome c release and caspase-3 activation with the resultant apoptosis. Pramipexole 15-26 caspase 3 Rattus norvegicus 136-145 16362428-8 2006 Pramipexole inhibited H2O2-induced JNK and p38 MAP kinase, but not ERK1/2 phosphorylation. Pramipexole 0-11 mitogen-activated protein kinase 8 Rattus norvegicus 35-38 16362428-8 2006 Pramipexole inhibited H2O2-induced JNK and p38 MAP kinase, but not ERK1/2 phosphorylation. Pramipexole 0-11 mitogen activated protein kinase 14 Rattus norvegicus 43-57 16362428-9 2006 Furthermore, in these cells experiments with a fluorescent probe, 2-[6-(4"-amino)phenoxy-3H-xanthen-3-on-9-yl]benzoic acid, revealed that pramipexole, the JNK inhibitor SP600125 and the p38 MAP kinase inhibitor SB203580 inhibited the generation of H2O2-induced reactive oxygen species. Pramipexole 138-149 mitogen-activated protein kinase 8 Rattus norvegicus 155-158 16362428-9 2006 Furthermore, in these cells experiments with a fluorescent probe, 2-[6-(4"-amino)phenoxy-3H-xanthen-3-on-9-yl]benzoic acid, revealed that pramipexole, the JNK inhibitor SP600125 and the p38 MAP kinase inhibitor SB203580 inhibited the generation of H2O2-induced reactive oxygen species. Pramipexole 138-149 mitogen activated protein kinase 14 Rattus norvegicus 186-200 16362428-11 2006 These results suggest that pramipexole exerts a protective effect against oxidative stress-induced PC12 cell death in part through an inhibition of JNK and p38 MAP kinase. Pramipexole 27-38 mitogen-activated protein kinase 8 Rattus norvegicus 148-151 16362428-11 2006 These results suggest that pramipexole exerts a protective effect against oxidative stress-induced PC12 cell death in part through an inhibition of JNK and p38 MAP kinase. Pramipexole 27-38 mitogen activated protein kinase 14 Rattus norvegicus 156-159 16088951-3 2005 The K(i) values for the human cloned D2(High) and D3(High) receptors, respectively, were 19 and 9 nM for pramipexole, 0.24 and 0.6 nM for +PHNO, 0.7 and 1.3 nM for bromocriptine, 0.5 and 2.6 nM for apomorphine, and 0.09 and 0.25 nM for (-)N-propylnorapomorphine. Pramipexole 105-116 iodothyronine deiodinase 3 Homo sapiens 50-68 17314491-1 2006 BACKGROUND: Pramipexole is a D3 dopaminergic agonist that has shown a major effect on both sensory and motor manifestations of restless legs syndrome (RLS) in long-term trials. Pramipexole 12-23 RLS1 Homo sapiens 151-154 17314491-3 2006 OBJECTIVE: To evaluate the acute effect of a low dosage of pramipexole (0.125 mg) on sensory symptoms and motor signs of RLS and on the macro- and microstructure of sleep. Pramipexole 59-70 RLS1 Homo sapiens 121-124 17314491-6 2006 Pramipexole 0.125 mg was administered before the second night at 9:00 p.m. A visual analog scale was used to assess the sensory symptoms of RLS. Pramipexole 0-11 RLS1 Homo sapiens 140-143 17314491-8 2006 RESULTS: After the acute administration of pramipexole, we observed a significant improvement of the sensory symptoms and motor signs of RLS. Pramipexole 43-54 RLS1 Homo sapiens 137-140 17314491-10 2006 CONCLUSIONS: Our results suggest that low doses of pramipexole are effective in reducing sensory symptoms and motor signs of RLS, even after the first administration. Pramipexole 51-62 RLS1 Homo sapiens 125-128 15640376-0 2005 Transport of the dopamine D2 agonist pramipexole by rat organic cation transporters OCT1 and OCT2 in kidney. Pramipexole 37-48 solute carrier family 22 member 1 Rattus norvegicus 84-88 15640376-7 2005 These results demonstrate that both rOCT1 and rOCT2 are involved in the renal uptake of pramipexole across the basolateral membrane of the proximal tubular epithelial cells. Pramipexole 88-99 solute carrier family 22 member 2 Rattus norvegicus 46-51 15640376-0 2005 Transport of the dopamine D2 agonist pramipexole by rat organic cation transporters OCT1 and OCT2 in kidney. Pramipexole 37-48 solute carrier family 22 member 2 Rattus norvegicus 93-97 15640376-2 2005 The uptake of [14C]pramipexole by Xenopus oocytes injected with complementary RNA of either rat organic cation transporter (rOCT) 1 or rOCT2 was significantly higher than that by water-injected oocytes: the kinetic parameters, K(m) and V(max), of pramipexole uptake were 49.5 muM and 234 pmol/60 min/oocyte for rOCT1, and 16.9 microM and 12.8 pmol/60 min/oocyte for rOCT2. Pramipexole 19-30 solute carrier family 22 member 2 Rattus norvegicus 135-140 15640376-2 2005 The uptake of [14C]pramipexole by Xenopus oocytes injected with complementary RNA of either rat organic cation transporter (rOCT) 1 or rOCT2 was significantly higher than that by water-injected oocytes: the kinetic parameters, K(m) and V(max), of pramipexole uptake were 49.5 muM and 234 pmol/60 min/oocyte for rOCT1, and 16.9 microM and 12.8 pmol/60 min/oocyte for rOCT2. Pramipexole 19-30 solute carrier family 22 member 1 Rattus norvegicus 311-316 15640376-2 2005 The uptake of [14C]pramipexole by Xenopus oocytes injected with complementary RNA of either rat organic cation transporter (rOCT) 1 or rOCT2 was significantly higher than that by water-injected oocytes: the kinetic parameters, K(m) and V(max), of pramipexole uptake were 49.5 muM and 234 pmol/60 min/oocyte for rOCT1, and 16.9 microM and 12.8 pmol/60 min/oocyte for rOCT2. Pramipexole 19-30 solute carrier family 22 member 2 Rattus norvegicus 366-371 15640376-6 2005 The IC50 values of procainamide and corticosterone for the uptake of [14C]pramipexole by rOCT1, rOCT2, and kidney slices were 7.7, 167.0, and 47.0 microM and 163.7, 10.7, and 47.7 microM, respectively. Pramipexole 74-85 solute carrier family 22 member 1 Rattus norvegicus 89-94 15640376-6 2005 The IC50 values of procainamide and corticosterone for the uptake of [14C]pramipexole by rOCT1, rOCT2, and kidney slices were 7.7, 167.0, and 47.0 microM and 163.7, 10.7, and 47.7 microM, respectively. Pramipexole 74-85 solute carrier family 22 member 2 Rattus norvegicus 96-101 15640376-7 2005 These results demonstrate that both rOCT1 and rOCT2 are involved in the renal uptake of pramipexole across the basolateral membrane of the proximal tubular epithelial cells. Pramipexole 88-99 solute carrier family 22 member 1 Rattus norvegicus 36-41 15569251-5 2004 However, pramipexole reduced caspase-3 activation, decreased the release of cytochrome c and prevented the fall in the mitochondrial membrane potential induced by MPP+ and rotenone. Pramipexole 9-20 caspase 3 Homo sapiens 29-38 15569251-5 2004 However, pramipexole reduced caspase-3 activation, decreased the release of cytochrome c and prevented the fall in the mitochondrial membrane potential induced by MPP+ and rotenone. Pramipexole 9-20 cytochrome c, somatic Homo sapiens 76-88 15473989-0 2004 Involvement of dopamine D(2)/D(3) receptors and BDNF in the neuroprotective effects of S32504 and pramipexole against 1-methyl-4-phenylpyridinium in terminally differentiated SH-SY5Y cells. Pramipexole 98-109 brain derived neurotrophic factor Homo sapiens 48-52 14655914-2 2003 DESIGN: Retrospective review of the records of consecutive patients treated with pramipexole for RLS. Pramipexole 81-92 RLS1 Homo sapiens 97-100 14985923-12 2004 CONCLUSIONS: Taken together, these studies suggest that the D2 receptor rather than the D3 receptor is important for the antidepressant-like activity observed for pramipexole in the mouse forced swim test. Pramipexole 163-174 dopamine receptor D2 Mus musculus 60-71 15473989-3 2004 To test if the D(3) receptor preferring agonists S32504 and pramipexole act through D(2) or D(3) receptors and via brain-derived neurotrophic factor (BDNF)-dependent pathways, we utilized a terminally differentiated neuroblastoma SH-SY5Y cell line exhibiting a dopaminergic phenotype. Pramipexole 60-71 brain derived neurotrophic factor Homo sapiens 115-148 15473989-3 2004 To test if the D(3) receptor preferring agonists S32504 and pramipexole act through D(2) or D(3) receptors and via brain-derived neurotrophic factor (BDNF)-dependent pathways, we utilized a terminally differentiated neuroblastoma SH-SY5Y cell line exhibiting a dopaminergic phenotype. Pramipexole 60-71 brain derived neurotrophic factor Homo sapiens 150-154 15473989-8 2004 Antibody directed against BDNF concentration-dependently blocked both the neuroprotective effects of BDNF and those of pramipexole and S32504 against MPP(+). Pramipexole 119-130 brain derived neurotrophic factor Homo sapiens 26-30 15473914-0 2004 Low dose pramipexole is neuroprotective in the MPTP mouse model of Parkinson"s disease, and downregulates the dopamine transporter via the D3 receptor. Pramipexole 9-20 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 110-130 15473914-2 2004 We also determined whether subchronic treatment with pramipexole regulates dopamine transporter function, thereby reducing intracellular transport of the active metabolite of MPTP, 1-methyl-4-phenylpyridinium (MPP+). Pramipexole 53-64 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 75-95 15473914-9 2004 MPTP- induced loss of striatal innervation, as measured by DAT-immunoreactivity, was partially prevented by pramipexole, but not with regard to TH-IR. Pramipexole 108-119 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 59-62 15473914-10 2004 Pramipexole also reduced DAT- immunoreactivity in non-MPTP treated mice. Pramipexole 0-11 solute carrier family 6 (neurotransmitter transporter, dopamine), member 3 Mus musculus 25-28 15170083-4 2004 The neuroprotective effects of pramipexole seemed to be derived from several mechanisms: stimulation of D(2) autoreceptor, stimulation of D(3) receptor, inhibition of oxidative reaction and following radical production, increase of Bcl-2 protein and inhibition of apoptotic cell death, and production of neurotrophic factor. Pramipexole 31-42 BCL2 apoptosis regulator Homo sapiens 232-237 14655914-4 2003 PATIENTS: 60 consecutive patients treated with pramipexole for RLS. Pramipexole 47-58 RLS1 Homo sapiens 63-66 14655914-6 2003 MEASUREMENTS AND RESULTS: Pramipexole was completely effective in controlling RLS in 67%, partially effective in 27%, and ineffective in 7% of patients. Pramipexole 26-37 RLS1 Homo sapiens 78-81 14655914-14 2003 CONCLUSIONS: Pramipexole was effective for RLS with continued response with time. Pramipexole 13-24 RLS1 Homo sapiens 43-46 10924701-0 2000 Increase of bcl-2 protein in neuronal dendritic processes of cerebral cortex and hippocampus by the antiparkinsonian drugs, talipexole and pramipexole. Pramipexole 139-150 BCL2, apoptosis regulator Rattus norvegicus 12-17 14521483-5 2003 Dopaminergic agents including levodopa and dopamine agonists such as pergolide, pramipexole, cabergoline and ropinirole are regarded as the treatment of choice for idiopathic RLS, however, the development of augmentation of symptoms, especially under levodopa therapy, may be a major problem. Pramipexole 80-91 RLS1 Homo sapiens 175-178 12954356-0 2003 Pramipexole inhibits MPTP toxicity in mice by dopamine D3 receptor dependent and independent mechanisms. Pramipexole 0-11 dopamine receptor D3 Mus musculus 46-66 12954356-2 2003 Pramipexole retained the ability to inhibit 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced dopamine depletion in mice in which the dopamine D3 receptor had been deleted. Pramipexole 0-11 dopamine receptor D3 Mus musculus 144-164 12954356-4 2003 Furthermore, the dopamine D3 receptor selective antagonist 2-(3-[4-(2-tert-butyl-6-trifluoromethyl-4-pyrimidinyl)-1-piperazinyl]propylthio)-4-pyrimidinol (A-437203) partially inhibited the neuroprotective effect of pramipexole in dopamine D3 receptor expressing mice but not in receptor-deleted mice. Pramipexole 215-226 dopamine receptor D3 Mus musculus 17-37 12482941-6 2002 At clinically relevant doses, pramipexole produced statistically robust decreases in rCBF in bilateral orbitofrontal cortex, thalamus, operculum, posterior and anterior (subgenual) cingulate cortex, and insula (in decreasing order of significance). Pramipexole 30-41 CCAAT/enhancer binding protein zeta Rattus norvegicus 85-89 12482941-8 2002 The dose-response curve and duration of pramipexole"s effects suggest that these rCBF responses indicate functional effects of a D3-preferring agonist. Pramipexole 40-51 CCAAT/enhancer binding protein zeta Rattus norvegicus 81-85 11419913-5 2001 However, the finding that pramipexole can normalise mitochondrial membrane potential and inhibit activity of caspase-3 in cytoplasmic hybrid cells derived from mitochondrial DNA of patients with nonfamilial Alzheimer"s disease suggests an even broader implication for the neuroprotective role of dopamine agonists. Pramipexole 26-37 caspase 3 Homo sapiens 109-118 11261747-2 2001 In contrast to pergolide, a D1 and D2 dopamine agonist, pramipexole is a nonergoline dopamine agonist with D2 and preferential D3 dopamine receptor activity. Pramipexole 56-67 dopamine receptor D3 Homo sapiens 127-147 12737528-3 2003 Recently, bromocriptine, pramipexole and several other agonists of the dopamine D2-receptor subfamily (including D2, D3 and D4-subtypes) have been shown to have neuroprotective effects in parkinsonian models in vitro and in vivo. Pramipexole 25-36 dopamine receptor D2 Homo sapiens 71-91 12464116-5 2002 Pramipexole in particular has shown efficacy in reducing toxicity to MPTP, MPP, rotenone and 6-hydroxydopamine. Pramipexole 0-11 M-phase phosphoprotein 6 Homo sapiens 75-78 11926889-0 2002 Dopamine transporter brain imaging to assess the effects of pramipexole vs levodopa on Parkinson disease progression. Pramipexole 60-71 solute carrier family 6 member 3 Homo sapiens 0-20 11402115-3 2001 METHODS: A randomized, assessor-blinded, placebo-controlled clinical trial was performed in subjects with early PD to determine whether L-dopa or pramipexole might regulate striatal DAT binding as measured by PET with [(11)C]RTI-32. Pramipexole 146-157 solute carrier family 6 member 3 Homo sapiens 182-185 11402115-5 2001 RESULTS: Mean interval change in DAT binding was significantly reduced by 16% to 22% in all striatal regions (caudate, anterior and posterior putamen) of the L-dopa-treated patients, whereas significant changes in the pramipexole-treated patients were limited to the contralateral caudate (-15%), ipsilateral anterior putamen (-14%), and posterior putamen (-20%). Pramipexole 218-229 solute carrier family 6 member 3 Homo sapiens 33-36 11402115-8 2001 CONCLUSIONS: Short-term therapy with L-dopa and, to a lesser extent, pramipexole can modestly down-regulate striatal DAT in patients with early PD. Pramipexole 69-80 solute carrier family 6 member 3 Homo sapiens 117-120 11346069-1 2001 OBJECTIVE: To review the use of newer dopamine agonists pramipexole and ropinirole, in the treatment of restless legs syndrome (RLS). Pramipexole 56-67 RLS1 Homo sapiens 128-131 11346069-4 2001 DATA SYNTHESIS: A number of small clinical studies of short duration have examined the use of pramipexole and ropinirole in the treatment of RLS. Pramipexole 94-105 RLS1 Homo sapiens 141-144 11346069-7 2001 CONCLUSIONS: Although the published trials show promising results for efficacy of pramipexole and ropinirole in the treatment of RLS, they are subject to many limitations. Pramipexole 82-93 RLS1 Homo sapiens 129-132 11301060-0 2001 Release and aggregation of cytochrome c and alpha-synuclein are inhibited by the antiparkinsonian drugs, talipexole and pramipexole. Pramipexole 120-131 cytochrome c, somatic Homo sapiens 27-39 11301060-0 2001 Release and aggregation of cytochrome c and alpha-synuclein are inhibited by the antiparkinsonian drugs, talipexole and pramipexole. Pramipexole 120-131 synuclein alpha Homo sapiens 44-59 11301060-2 2001 In the present study, we examined the effects of talipexole and pramipexole on the release of cytochrome c and alpha-synuclein, their aggregations, and activation of caspases. Pramipexole 64-75 cytochrome c, somatic Homo sapiens 94-106 11301060-2 2001 In the present study, we examined the effects of talipexole and pramipexole on the release of cytochrome c and alpha-synuclein, their aggregations, and activation of caspases. Pramipexole 64-75 synuclein alpha Homo sapiens 111-126 11301060-4 2001 Talipexole and pramipexole at low concentration (0.1-1 mM) significantly inhibited the accumulation of cytochrome c or alpha-synuclein in the cytosolic fraction. Pramipexole 15-26 cytochrome c, somatic Homo sapiens 103-115 11301060-4 2001 Talipexole and pramipexole at low concentration (0.1-1 mM) significantly inhibited the accumulation of cytochrome c or alpha-synuclein in the cytosolic fraction. Pramipexole 15-26 synuclein alpha Homo sapiens 119-134 11301060-7 2001 These results suggest that talipexole and pramipexole may have protective effects against the neurodegeneration, which is induced by intracellular accumulation of cytochrome c and alpha-synuclein. Pramipexole 42-53 cytochrome c, somatic Homo sapiens 163-175 11301060-7 2001 These results suggest that talipexole and pramipexole may have protective effects against the neurodegeneration, which is induced by intracellular accumulation of cytochrome c and alpha-synuclein. Pramipexole 42-53 synuclein alpha Homo sapiens 180-195 10924701-1 2000 Treatment of rats for 4 days with the antiparkinsonian drugs, talipexole and pramipexole, markedly increased Bcl-2 immunoreactivity in neuronal dendritic processes in both cerebral cortex and hippocampus, but treatment for 1 day with either of these drugs did not. Pramipexole 77-88 BCL2, apoptosis regulator Rattus norvegicus 109-114 7814630-4 1995 Western blot analysis revealed that in renal cortical membranes, fenoldopam and pramipexole increased expression of PLC beta 1 and decreased expression of PLC gamma 1; PLC delta was unchanged. Pramipexole 80-91 phospholipase C beta 1 Rattus norvegicus 116-126 11054154-7 2000 Pramipexole is a full dopamine agonist with high selectivity for the D2 dopamine receptor family. Pramipexole 0-11 dopamine receptor D2 Homo sapiens 69-89 11054154-9 2000 Pramipexole has a 5- to 7-fold greater affinity for the D3 receptor subtype with lower affinities for the D2 and D4 receptor subtypes. Pramipexole 0-11 dopamine receptor D4 Homo sapiens 113-124 10812530-1 2000 Pramipexole, a dopamine D2 receptor agonist, was tested in 174 patients with major depression, with or without melancholia and without psychotic features. Pramipexole 0-11 dopamine receptor D2 Homo sapiens 15-35 10201413-1 1999 OBJECTIVES: Pramipexole, a non-ergot dopamine D2/D3 receptor agonist, was investigated as an add on drug in advanced parkinsonian patients with motor fluctuations to assess efficacy, safety, and tolerance. Pramipexole 12-23 dopamine receptor D2 Homo sapiens 37-60 10234741-0 1999 [3H]Pramipexole: a selective radioligand for the high affinity dopamine D2 receptor in bovine striatal membranes. Pramipexole 4-15 dopamine receptor D2 Bos taurus 63-83 9855633-7 1998 Similarly, pramipexole pretreatment increased Bcl-2 and inhibited MPP+-induced apoptosis. Pramipexole 11-22 BCL2 apoptosis regulator Homo sapiens 46-51 8665547-1 1995 We evaluated the efficacy, safety, tolerability, and pharmacokinetics of pramipexole, a novel dopamine D2 receptor agonist, in early Parkinson"s disease (PD). Pramipexole 73-84 dopamine receptor D2 Homo sapiens 94-114 7814630-4 1995 Western blot analysis revealed that in renal cortical membranes, fenoldopam and pramipexole increased expression of PLC beta 1 and decreased expression of PLC gamma 1; PLC delta was unchanged. Pramipexole 80-91 phospholipase C, gamma 1 Rattus norvegicus 155-166 7814630-5 1995 In the cytosol, pramipexole and fenoldopam increased expression of both PLC beta 1 and PLC gamma 1. Pramipexole 16-27 phospholipase C beta 1 Rattus norvegicus 72-82 7814630-5 1995 In the cytosol, pramipexole and fenoldopam increased expression of both PLC beta 1 and PLC gamma 1. Pramipexole 16-27 phospholipase C, gamma 1 Rattus norvegicus 87-98 8748629-1 1995 We evaluated the efficacy, safety and tolerability of a new dopamine D-2 receptor agonist, pramipexole [(S)-2-amino-4,5,6,7-tetrahydro-6-propylamino-benzathiazol-dihydro chloride], as adjunctive therapy in patients with advanced Parkinson"s disease (PD). Pramipexole 91-102 dopamine receptor D2 Homo sapiens 60-81 34218945-9 2022 CONCLUSIONS: Pramipexole improved behavior of PD model rat, and down regulated the mRNA expression and concentrations of IL-6 and TNF-alpha in their CNS. Pramipexole 13-24 interleukin 6 Rattus norvegicus 121-125 2142088-2 1990 The basal serum prolactin levels were reduced dose dependently by s.c. administration of talipexole or SND 919 at doses of 5-100 micrograms/kg. Pramipexole 103-110 prolactin Rattus norvegicus 16-25 2142088-5 1990 In vitro, the spontaneous prolactin release into perfusates from isolated anterior pituitary was inhibited by talipexole or SND 919 added at concentrations ranging from 10(-9) to 10(-6) M. This inhibitory effect of SND 919 was blocked by concurrent application of a dopamine D-2 receptor antagonist, YM-09151-2. Pramipexole 124-131 prolactin Rattus norvegicus 26-35 2142088-5 1990 In vitro, the spontaneous prolactin release into perfusates from isolated anterior pituitary was inhibited by talipexole or SND 919 added at concentrations ranging from 10(-9) to 10(-6) M. This inhibitory effect of SND 919 was blocked by concurrent application of a dopamine D-2 receptor antagonist, YM-09151-2. Pramipexole 215-222 prolactin Rattus norvegicus 26-35 2142088-9 1990 The results suggest that talipexole and SND 919 have a selective dopamine D-2 receptor agonistic property and are almost completely effective to counteract the enhancement of prolactin release induced by estrogens via stimulation of dopamine D-2 receptors in the anterior pituitary. Pramipexole 40-47 prolactin Rattus norvegicus 175-184 1357983-2 1992 Because dopamine has actions on receptors (e.g., adrenergic, serotonin) other than dopamine, we studied a novel dopamine agonist, pramipexole, which has a selectivity to both DA1 and DA2-receptor subtypes. Pramipexole 130-141 RT1 class II, locus Da Rattus norvegicus 175-178 1357983-5 1992 The renal effects of pramipexole were mainly due to actions at the DA1 receptor, since these effects were completely blocked by the coinfusion of a DA1 antagonist, SKF 83742. Pramipexole 21-32 RT1 class II, locus Da Rattus norvegicus 67-70 1357983-5 1992 The renal effects of pramipexole were mainly due to actions at the DA1 receptor, since these effects were completely blocked by the coinfusion of a DA1 antagonist, SKF 83742. Pramipexole 21-32 RT1 class II, locus Da Rattus norvegicus 148-151 1350237-3 1992 Pramipexole decreased serum prolactin levels in a dose-dependent manner, with a maximum effect after 2 to 4 hours. Pramipexole 0-11 prolactin Homo sapiens 28-37 34218945-9 2022 CONCLUSIONS: Pramipexole improved behavior of PD model rat, and down regulated the mRNA expression and concentrations of IL-6 and TNF-alpha in their CNS. Pramipexole 13-24 tumor necrosis factor Rattus norvegicus 130-139 34810175-3 2022 Previous small randomised controlled trials suggested that pramipexole, a dopamine D2/3 receptor agonist, may be effective for treating patients with unipolar and bipolar depression as it is known to influence motivational drive and reward processing. Pramipexole 59-70 dopamine receptor D2 Homo sapiens 74-96 34699917-0 2021 Pramipexole Regulates Depression-Like Behavior via Dopamine D3 Receptor in a Mouse Model of Parkinson"s Disease. Pramipexole 0-11 dopamine receptor D3 Mus musculus 51-71 34699917-7 2021 Pramipexole significantly improved depression-like behavior in DRD2-/- mice but not in DRD3-/- mice. Pramipexole 0-11 dopamine receptor D2 Mus musculus 63-67 34699917-8 2021 These results demonstrate that the antidepressive effect of pramipexole is mediated by DRD3 but not DRD2. Pramipexole 60-71 dopamine receptor D3 Mus musculus 87-91 34451897-1 2021 Treatment with the dopamine D2/D3 receptor agonist pramipexole has demonstrated promising clinical effects in patients with depression. Pramipexole 51-62 dopamine receptor D2 Homo sapiens 19-42 34432265-10 2021 We observed that PPX treatment downregulated IL-1beta levels and increased BNDF content in the spinal cord after EAE induction. Pramipexole 17-20 interleukin 1 alpha Mus musculus 45-53 34708087-7 2021 In response to scratch injury, WT cardiac fibroblasts treated with the D3R agonist, pramipexole, displayed enhanced migration compared to control WT and D3KO cells. Pramipexole 84-95 dopamine receptor D3 Mus musculus 71-74 35605609-0 2022 Enhanced taste recognition following subacute treatment with the dopamine D2/D3 receptor agonist pramipexole in healthy volunteers. Pramipexole 97-108 dopamine receptor D2 Homo sapiens 65-88 34249258-0 2021 Dopaminergic agonist pramipexole improves memory and increases IL-10 production in LPS-challenged rats. Pramipexole 21-32 interleukin 10 Rattus norvegicus 63-68 34249258-12 2021 Interleukin (IL)-10 serum levels were elevated by 1 mg/kg PMX. Pramipexole 58-61 interleukin 10 Rattus norvegicus 0-19 34249258-14 2021 PMX at all doses significantly decreased BDNF serum concentration. Pramipexole 0-3 brain-derived neurotrophic factor Rattus norvegicus 41-45 35609673-3 2022 RLS can generally be well treated with medications such as alpha-2-delta calcium channel ligands (A2Ds) gabapentin, pregabalin, gabapentin enacarbil or, if these are poorly tolerated or lack efficacy, dopamine agonists (DAs), pramipexole, ropinirole or rotigotine. Pramipexole 226-237 RLS1 Homo sapiens 0-3 33493581-0 2021 Pramipexole attenuates neuronal injury in Parkinson"s disease by targeting miR-96 to activate BNIP3-mediated mitophagy. Pramipexole 0-11 microRNA 96 Mus musculus 75-81 35524855-7 2022 After treatment with Pramipexole, the neurofunctional deficits, behavioral disability, and aggravated pathological changes were dramatically reversed, accompanied by the alleviated BBB permeability, and upregulated occludin, an important tight junction protein. Pramipexole 21-32 occludin Mus musculus 215-223 35524855-10 2022 Increased fluorescence intensity of FITC-dextran, reduced value of TEER, and downregulated occludin and KLF2 were observed in TBI-exposed cells, all of which were greatly reversed by 10 and 20 muM Pramipexole. Pramipexole 197-208 occludin Mus musculus 91-99 35524855-10 2022 Increased fluorescence intensity of FITC-dextran, reduced value of TEER, and downregulated occludin and KLF2 were observed in TBI-exposed cells, all of which were greatly reversed by 10 and 20 muM Pramipexole. Pramipexole 197-208 Kruppel-like factor 2 (lung) Mus musculus 104-108 35524855-11 2022 Furthermore, in KLF2-silenced bEnd.3 cells, the protective ability of Pramipexole against endothelial permeability and the expression level of occludin were dramatically abolished. Pramipexole 70-81 Kruppel-like factor 2 (lung) Mus musculus 16-20 35524855-11 2022 Furthermore, in KLF2-silenced bEnd.3 cells, the protective ability of Pramipexole against endothelial permeability and the expression level of occludin were dramatically abolished. Pramipexole 70-81 occludin Mus musculus 143-151 35524855-12 2022 Collectively, our results suggest that Pramipexole protected against TBI-induced BBB dysfunction by mediating KLF2. Pramipexole 39-50 Kruppel-like factor 2 (lung) Mus musculus 110-114 35463692-3 2022 Compared with that in normal group, the positive expression of BDNF was substantially increased in model group and pramipexole group, and its positive expression in pramipexole group was notably higher than that in model group. Pramipexole 115-126 brain-derived neurotrophic factor Rattus norvegicus 63-67 35463692-3 2022 Compared with that in normal group, the positive expression of BDNF was substantially increased in model group and pramipexole group, and its positive expression in pramipexole group was notably higher than that in model group. Pramipexole 165-176 brain-derived neurotrophic factor Rattus norvegicus 63-67 35463692-5 2022 The relative expression levels of miR-103a and miR-30b in model and pramipexole groups were markedly higher than those in normal group, and pramipexole group had remarkably higher relative expression levels of miR-103a and miR-30b than model group. Pramipexole 68-79 microRNA 30b Rattus norvegicus 47-54 35463692-5 2022 The relative expression levels of miR-103a and miR-30b in model and pramipexole groups were markedly higher than those in normal group, and pramipexole group had remarkably higher relative expression levels of miR-103a and miR-30b than model group. Pramipexole 68-79 microRNA 30b Rattus norvegicus 223-230 35463692-5 2022 The relative expression levels of miR-103a and miR-30b in model and pramipexole groups were markedly higher than those in normal group, and pramipexole group had remarkably higher relative expression levels of miR-103a and miR-30b than model group. Pramipexole 140-151 microRNA 30b Rattus norvegicus 47-54 35463692-5 2022 The relative expression levels of miR-103a and miR-30b in model and pramipexole groups were markedly higher than those in normal group, and pramipexole group had remarkably higher relative expression levels of miR-103a and miR-30b than model group. Pramipexole 140-151 microRNA 30b Rattus norvegicus 223-230 35463692-6 2022 It was found through ELISA that model and pramipexole groups had markedly raised IL-1beta and IL-18 content compared with normal group, and their content in pramipexole group was remarkably lower than that in model group. Pramipexole 42-53 interleukin 1 alpha Rattus norvegicus 81-89 35463692-6 2022 It was found through ELISA that model and pramipexole groups had markedly raised IL-1beta and IL-18 content compared with normal group, and their content in pramipexole group was remarkably lower than that in model group. Pramipexole 42-53 interleukin 18 Rattus norvegicus 94-99 35463692-8 2022 Pramipexole may up-regulate the expressions of BDNF, miR-103a and miR-30b to inhibit the apoptosis and inflammation in Parkinson"s disease model rats. Pramipexole 0-11 brain-derived neurotrophic factor Rattus norvegicus 47-51 35463692-8 2022 Pramipexole may up-regulate the expressions of BDNF, miR-103a and miR-30b to inhibit the apoptosis and inflammation in Parkinson"s disease model rats. Pramipexole 0-11 microRNA 30b Rattus norvegicus 66-73 33493581-11 2021 miR-96 overexpression or BNIP3 interference weakened the suppressive role of PPX in MPP+-induced neuronal injury. Pramipexole 77-80 microRNA 96 Mus musculus 0-6 33493581-11 2021 miR-96 overexpression or BNIP3 interference weakened the suppressive role of PPX in MPP+-induced neuronal injury. Pramipexole 77-80 BCL2/adenovirus E1B interacting protein 3 Mus musculus 25-30 33493581-15 2021 CONCLUSION: PPX mitigated neuronal injury in MPP+-induced cells and MPTP-induced mice by activating BNIP3-mediated mitophagy via directly decreasing miR-96. Pramipexole 12-15 BCL2/adenovirus E1B interacting protein 3 Mus musculus 100-105 33493581-15 2021 CONCLUSION: PPX mitigated neuronal injury in MPP+-induced cells and MPTP-induced mice by activating BNIP3-mediated mitophagy via directly decreasing miR-96. Pramipexole 12-15 microRNA 96 Mus musculus 149-155 34269222-0 2022 Pramipexole, a dopamine D3/D2 receptor-preferring agonist, attenuates reserpine-induced fibromyalgia-like model in mice. Pramipexole 0-11 dopamine receptor D3 Mus musculus 15-38 34269222-4 2022 Our results showed that pramipexole (PPX) - a dopaminergic D3/D2 receptor agonist - inhibited mechanical allodynia and thermal sensitivity induced by reserpine. Pramipexole 24-35 dopamine receptor D2 Mus musculus 59-73 34269222-4 2022 Our results showed that pramipexole (PPX) - a dopaminergic D3/D2 receptor agonist - inhibited mechanical allodynia and thermal sensitivity induced by reserpine. Pramipexole 37-40 dopamine receptor D2 Mus musculus 59-73 33493581-0 2021 Pramipexole attenuates neuronal injury in Parkinson"s disease by targeting miR-96 to activate BNIP3-mediated mitophagy. Pramipexole 0-11 BCL2/adenovirus E1B interacting protein 3 Mus musculus 94-99 33264473-8 2021 The DRD2 agonist, pramipexole, administered systemically caused colorectal propulsion that was prevented when the pelvic nerves were cut. Pramipexole 18-29 dopamine receptor D2 Homo sapiens 4-8 33891271-0 2021 Pramipexole attenuates 6-OHDA-induced Parkinson"s disease by mediating the Nurr1/NF-kappaB pathway. Pramipexole 0-11 nuclear receptor subfamily 4, group A, member 2 Rattus norvegicus 75-80 33891271-14 2021 PPX improved the motor behavior of PD through mediating the inflammatory response and regulating the Nurr1/NF-kappaB signaling pathway. Pramipexole 0-3 nuclear receptor subfamily 4, group A, member 2 Rattus norvegicus 101-106