PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 26314956-1 2015 Mitochondrial Lon protease (Lon) regulates several mitochondrial functions, and is inhibited by the anticancer molecule triterpenoid 2-cyano-3, 12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO), or by its C-28 methyl ester derivative (CDDO-Me). bardoxolone methyl 232-239 lon peptidase 1, mitochondrial Homo sapiens 14-26 26264646-5 2015 Intravenous delivery of CDDO-Me using poly-lactic-glycolic-acid NP combination with subcutaneous Trp2 vaccine resulted in an increase of antitumor efficacy and apoptotic tumor tissue than Trp2 vaccine alone in B16F10 melanoma. bardoxolone methyl 24-31 tRNA-Pro (anticodon AGG) 2-6 Homo sapiens 97-101 26264646-5 2015 Intravenous delivery of CDDO-Me using poly-lactic-glycolic-acid NP combination with subcutaneous Trp2 vaccine resulted in an increase of antitumor efficacy and apoptotic tumor tissue than Trp2 vaccine alone in B16F10 melanoma. bardoxolone methyl 24-31 tRNA-Pro (anticodon AGG) 2-6 Homo sapiens 188-192 26497549-3 2015 Treatment of K-ras mutant MiaPaCa-2 and K-ras normal BxPC-3 cells with CDDO-Me elicited strong antiproliferative and proapoptopic responses in both cell lines in culture. bardoxolone methyl 71-78 KRAS proto-oncogene, GTPase Homo sapiens 13-18 26497549-3 2015 Treatment of K-ras mutant MiaPaCa-2 and K-ras normal BxPC-3 cells with CDDO-Me elicited strong antiproliferative and proapoptopic responses in both cell lines in culture. bardoxolone methyl 71-78 KRAS proto-oncogene, GTPase Homo sapiens 40-45 26497549-6 2015 Treatment with CDDO-Me significantly inhibited the growth of BxPC-3 xenografts and reduced the levels of p-Akt and p-mTOR in tumor tissue. bardoxolone methyl 15-22 mechanistic target of rapamycin kinase Homo sapiens 117-121 26459563-8 2015 Similar to the activation of Nrf2 by either silencing of Keap1 expression or by the reactive compound 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me), RA839 prevented the induction of both inducible nitric-oxide synthase expression and nitric oxide release in response to lipopolysaccharides in macrophages. bardoxolone methyl 162-169 nuclear factor, erythroid derived 2, like 2 Mus musculus 29-33 26314956-1 2015 Mitochondrial Lon protease (Lon) regulates several mitochondrial functions, and is inhibited by the anticancer molecule triterpenoid 2-cyano-3, 12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO), or by its C-28 methyl ester derivative (CDDO-Me). bardoxolone methyl 232-239 lon peptidase 1, mitochondrial Homo sapiens 14-17 26053096-0 2015 Ca2+ influx-mediated dilation of the endoplasmic reticulum and c-FLIPL downregulation trigger CDDO-Me-induced apoptosis in breast cancer cells. bardoxolone methyl 94-101 CASP8 and FADD like apoptosis regulator Homo sapiens 63-70 26053096-6 2015 In addition, CDDO-Me rapidly reduced the protein levels of c-FLIPL (cellular FLICE-inhibitory protein) and overexpression of c-FLIPL blocked CDDO-Me-induced cell death, but not vacuolation. bardoxolone methyl 13-20 CASP8 and FADD like apoptosis regulator Homo sapiens 59-66 26053096-6 2015 In addition, CDDO-Me rapidly reduced the protein levels of c-FLIPL (cellular FLICE-inhibitory protein) and overexpression of c-FLIPL blocked CDDO-Me-induced cell death, but not vacuolation. bardoxolone methyl 141-148 CASP8 and FADD like apoptosis regulator Homo sapiens 125-132 26053096-7 2015 These results suggest that c-FLIPL downregulation is a key contributor to CDDO-Me-induced apoptotic cell death, independent of ER-derived vacuolation. bardoxolone methyl 74-81 CASP8 and FADD like apoptosis regulator Homo sapiens 27-34 26053096-8 2015 Taken together, our results show that ER-derived vacuolation via Ca2+ influx and ROS generation as well as caspase activation via c-FLIPL downregulation are responsible for the potent anticancer effects of CDDO-Me on breast cancer cells. bardoxolone methyl 206-213 CASP8 and FADD like apoptosis regulator Homo sapiens 130-137 26008822-8 2015 We also found that CDDO-Me inhibited the increases in ROS levels in the A53T flies and improved the neurodegenerative changes by activating the Nrf2/antioxidant response element signaling pathway. bardoxolone methyl 19-26 cap-n-collar Drosophila melanogaster 144-148 25733817-7 2015 Furthermore, CDDO-Me induced autophagy in both Ec109 and KYSE70 cells via suppression of the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway. bardoxolone methyl 13-20 mechanistic target of rapamycin kinase Homo sapiens 136-165 25614226-6 2015 Pretreatment with CDDO-Me significantly ameliorated W/D ratio, lung MPO activity, inflammatory cell infiltration, and inflammatory cytokine production in BALF from the in vivo study. bardoxolone methyl 18-25 myeloperoxidase Mus musculus 68-71 26134508-0 2015 Hsp90 Is a Novel Target Molecule of CDDO-Me in Inhibiting Proliferation of Ovarian Cancer Cells. bardoxolone methyl 36-43 heat shock protein 90 alpha family class A member 1 Homo sapiens 0-5 26134508-3 2015 Here, we demonstrate that CDDO-Me directly interacts with Hsp90 in cells by cellular thermal shift assay. bardoxolone methyl 26-33 heat shock protein 90 alpha family class A member 1 Homo sapiens 58-63 26134508-4 2015 CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. bardoxolone methyl 0-7 heat shock protein family A (Hsp70) member 4 Homo sapiens 43-48 26134508-4 2015 CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. bardoxolone methyl 0-7 heat shock protein 90 alpha family class A member 1 Homo sapiens 68-73 26134508-4 2015 CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. bardoxolone methyl 0-7 erb-b2 receptor tyrosine kinase 2 Homo sapiens 83-88 26134508-4 2015 CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. bardoxolone methyl 0-7 AKT serine/threonine kinase 1 Homo sapiens 93-96 26134508-4 2015 CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. bardoxolone methyl 0-7 heat shock protein 90 alpha family class A member 1 Homo sapiens 128-133 26134508-4 2015 CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. bardoxolone methyl 137-144 heat shock protein 90 alpha family class A member 1 Homo sapiens 68-73 26134508-4 2015 CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. bardoxolone methyl 137-144 erb-b2 receptor tyrosine kinase 2 Homo sapiens 83-88 26134508-4 2015 CDDO-Me treatment leads to upregulation of Hsp70 and degradation of Hsp90 clients (ErbB2 and Akt), indicating the inhibition of Hsp90 by CDDO-Me in cells. bardoxolone methyl 137-144 heat shock protein 90 alpha family class A member 1 Homo sapiens 128-133 26134508-5 2015 Knockdown of Hsp90 significantly inhibits cell proliferation and enhances the anti-proliferation effect of CDDO-Me in H08910 ovarian cancer cells. bardoxolone methyl 107-114 heat shock protein 90 alpha family class A member 1 Homo sapiens 13-18 26134508-6 2015 Dithiothreitol inhibits the interaction of CDDO-Me with Hsp90 in cells and abrogates CDDO-Me induced upregulation of Hsp70, degradation of Akt and cell proliferation inhibition. bardoxolone methyl 43-50 heat shock protein 90 alpha family class A member 1 Homo sapiens 56-61 26134508-6 2015 Dithiothreitol inhibits the interaction of CDDO-Me with Hsp90 in cells and abrogates CDDO-Me induced upregulation of Hsp70, degradation of Akt and cell proliferation inhibition. bardoxolone methyl 85-92 heat shock protein family A (Hsp70) member 4 Homo sapiens 117-122 26134508-6 2015 Dithiothreitol inhibits the interaction of CDDO-Me with Hsp90 in cells and abrogates CDDO-Me induced upregulation of Hsp70, degradation of Akt and cell proliferation inhibition. bardoxolone methyl 85-92 AKT serine/threonine kinase 1 Homo sapiens 139-142 26134508-7 2015 This suggests the anti-ovarian cancer effect of CDDO-Me is possibly mediated by the formation of Michael adducts between CDDO-Me and reactive nucleophiles on Hsp90. bardoxolone methyl 48-55 heat shock protein 90 alpha family class A member 1 Homo sapiens 158-163 26134508-7 2015 This suggests the anti-ovarian cancer effect of CDDO-Me is possibly mediated by the formation of Michael adducts between CDDO-Me and reactive nucleophiles on Hsp90. bardoxolone methyl 121-128 heat shock protein 90 alpha family class A member 1 Homo sapiens 158-163 26134508-8 2015 This study identifies Hsp90 as a novel target protein of CDDO-Me, and provides a novel insight into the mechanism of action of CDDO-Me in ovarian cancer cells. bardoxolone methyl 57-64 heat shock protein 90 alpha family class A member 1 Homo sapiens 22-27 26134508-8 2015 This study identifies Hsp90 as a novel target protein of CDDO-Me, and provides a novel insight into the mechanism of action of CDDO-Me in ovarian cancer cells. bardoxolone methyl 127-134 heat shock protein 90 alpha family class A member 1 Homo sapiens 22-27 25939751-0 2015 Dimethyl fumarate and the oleanane triterpenoids, CDDO-imidazolide and CDDO-methyl ester, both activate the Nrf2 pathway but have opposite effects in the A/J model of lung carcinogenesis. bardoxolone methyl 71-88 nuclear factor, erythroid derived 2, like 2 Mus musculus 108-112 25939751-5 2015 Although the triterpenoids and DMF both activated the Nrf2 pathway, CDDO-Im and CDDO-Me were markedly more potent than DMF. bardoxolone methyl 80-87 nuclear factor, erythroid derived 2, like 2 Mus musculus 54-58 25939751-17 2015 Collectively, these results suggest that although CDDO-Im, CDDO-Me and DMF all activate the Nrf2 pathway, they target distinct genes and signaling pathways, resulting in opposite effects for the prevention of experimental lung cancer. bardoxolone methyl 59-66 nuclear factor, erythroid derived 2, like 2 Mus musculus 92-96 25733817-6 2015 CDDO-Me significantly decreased B-cell lymphoma-extra large (Bcl-xl), B-cell lymphoma 2 (Bcl-2), cleaved caspase-9, and cleaved poly ADP ribose polymerase (PARP) levels but increased the expression level of Bcl-2-associated X (Bax). bardoxolone methyl 0-7 BCL2 like 1 Homo sapiens 61-67 25733817-6 2015 CDDO-Me significantly decreased B-cell lymphoma-extra large (Bcl-xl), B-cell lymphoma 2 (Bcl-2), cleaved caspase-9, and cleaved poly ADP ribose polymerase (PARP) levels but increased the expression level of Bcl-2-associated X (Bax). bardoxolone methyl 0-7 BCL2 apoptosis regulator Homo sapiens 70-87 25733817-6 2015 CDDO-Me significantly decreased B-cell lymphoma-extra large (Bcl-xl), B-cell lymphoma 2 (Bcl-2), cleaved caspase-9, and cleaved poly ADP ribose polymerase (PARP) levels but increased the expression level of Bcl-2-associated X (Bax). bardoxolone methyl 0-7 BCL2 apoptosis regulator Homo sapiens 89-94 25733817-6 2015 CDDO-Me significantly decreased B-cell lymphoma-extra large (Bcl-xl), B-cell lymphoma 2 (Bcl-2), cleaved caspase-9, and cleaved poly ADP ribose polymerase (PARP) levels but increased the expression level of Bcl-2-associated X (Bax). bardoxolone methyl 0-7 caspase 9 Homo sapiens 105-114 25733817-6 2015 CDDO-Me significantly decreased B-cell lymphoma-extra large (Bcl-xl), B-cell lymphoma 2 (Bcl-2), cleaved caspase-9, and cleaved poly ADP ribose polymerase (PARP) levels but increased the expression level of Bcl-2-associated X (Bax). bardoxolone methyl 0-7 poly(ADP-ribose) polymerase 1 Homo sapiens 128-154 25733817-6 2015 CDDO-Me significantly decreased B-cell lymphoma-extra large (Bcl-xl), B-cell lymphoma 2 (Bcl-2), cleaved caspase-9, and cleaved poly ADP ribose polymerase (PARP) levels but increased the expression level of Bcl-2-associated X (Bax). bardoxolone methyl 0-7 poly(ADP-ribose) polymerase 1 Homo sapiens 156-160 25733817-7 2015 Furthermore, CDDO-Me induced autophagy in both Ec109 and KYSE70 cells via suppression of the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway. bardoxolone methyl 13-20 AKT serine/threonine kinase 1 Homo sapiens 172-175 25733817-7 2015 Furthermore, CDDO-Me induced autophagy in both Ec109 and KYSE70 cells via suppression of the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway. bardoxolone methyl 13-20 mechanistic target of rapamycin kinase Homo sapiens 176-180 25733817-9 2015 CDDO-Me also scavenged reactive oxygen species through activation of the nuclear factor (erythroid-derived 2)-related factor 2 (Nrf2) pathway in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 NFE2 like bZIP transcription factor 2 Homo sapiens 128-132 25733817-11 2015 CDDO-Me significantly downregulated E-cadherin but upregulated Snail, Slug, and zinc finger E-box-binding homeobox 1 (TCF-8/ZEB1) in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 cadherin 1 Homo sapiens 36-46 25733817-6 2015 CDDO-Me significantly decreased B-cell lymphoma-extra large (Bcl-xl), B-cell lymphoma 2 (Bcl-2), cleaved caspase-9, and cleaved poly ADP ribose polymerase (PARP) levels but increased the expression level of Bcl-2-associated X (Bax). bardoxolone methyl 0-7 BCL2 associated X, apoptosis regulator Homo sapiens 207-225 25733817-6 2015 CDDO-Me significantly decreased B-cell lymphoma-extra large (Bcl-xl), B-cell lymphoma 2 (Bcl-2), cleaved caspase-9, and cleaved poly ADP ribose polymerase (PARP) levels but increased the expression level of Bcl-2-associated X (Bax). bardoxolone methyl 0-7 BCL2 associated X, apoptosis regulator Homo sapiens 227-230 25733817-11 2015 CDDO-Me significantly downregulated E-cadherin but upregulated Snail, Slug, and zinc finger E-box-binding homeobox 1 (TCF-8/ZEB1) in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 snail family transcriptional repressor 1 Homo sapiens 63-68 25733817-7 2015 Furthermore, CDDO-Me induced autophagy in both Ec109 and KYSE70 cells via suppression of the phosphoinositide 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway. bardoxolone methyl 13-20 protein tyrosine kinase 2 beta Homo sapiens 119-135 25733817-11 2015 CDDO-Me significantly downregulated E-cadherin but upregulated Snail, Slug, and zinc finger E-box-binding homeobox 1 (TCF-8/ZEB1) in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 snail family transcriptional repressor 2 Homo sapiens 70-74 25733817-11 2015 CDDO-Me significantly downregulated E-cadherin but upregulated Snail, Slug, and zinc finger E-box-binding homeobox 1 (TCF-8/ZEB1) in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 zinc finger E-box binding homeobox 1 Homo sapiens 80-116 25733817-11 2015 CDDO-Me significantly downregulated E-cadherin but upregulated Snail, Slug, and zinc finger E-box-binding homeobox 1 (TCF-8/ZEB1) in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 zinc finger E-box binding homeobox 1 Homo sapiens 118-123 25733817-11 2015 CDDO-Me significantly downregulated E-cadherin but upregulated Snail, Slug, and zinc finger E-box-binding homeobox 1 (TCF-8/ZEB1) in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 zinc finger E-box binding homeobox 1 Homo sapiens 124-128 25733817-12 2015 CDDO-Me significantly decreased the expression of octamer-4, sex determining region Y-box 2 (Sox-2), Nanog, and B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1), all markers of cancer cell stemness, in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 SRY-box transcription factor 2 Homo sapiens 61-91 25733817-12 2015 CDDO-Me significantly decreased the expression of octamer-4, sex determining region Y-box 2 (Sox-2), Nanog, and B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1), all markers of cancer cell stemness, in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 SRY-box transcription factor 2 Homo sapiens 93-98 25733817-12 2015 CDDO-Me significantly decreased the expression of octamer-4, sex determining region Y-box 2 (Sox-2), Nanog, and B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1), all markers of cancer cell stemness, in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 Nanog homeobox Homo sapiens 101-106 25733817-12 2015 CDDO-Me significantly decreased the expression of octamer-4, sex determining region Y-box 2 (Sox-2), Nanog, and B lymphoma Mo-MLV insertion region 1 homolog (Bmi-1), all markers of cancer cell stemness, in Ec109 and KYSE70 cells. bardoxolone methyl 0-7 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 158-163 25536195-2 2014 Synthetic triterpenoids, including CDDO-Me, act as anti-inflammatory and antioxidant modulators primarily by inducing the transcription factor Nrf2 to activate downstream genes containing antioxidant response elements (AREs). bardoxolone methyl 35-42 NFE2 like bZIP transcription factor 2 Homo sapiens 143-147 25926128-6 2015 In addition to a higher induction of the key cytoprotective gene heme oxygenase-1 (38.9-fold increase for CDDO-Im and 26.5-fold increase for CDDO-Me), CDDO-Im also induced greater expression of heat shock proteins (5.5-fold increase compared to 2.8-fold for CDDO-Me). bardoxolone methyl 141-148 heme oxygenase 1 Homo sapiens 65-81 25536195-5 2014 CDDO-Me was an effective radioprotector when given ~18 hours before radiation in epithelial cells (average dose modifying factor (DMF) = 1.3), and Nrf2 function was necessary for CDDO-Me to exert these radioprotective effects. bardoxolone methyl 179-186 NFE2 like bZIP transcription factor 2 Homo sapiens 147-151 25536195-8 2014 A therapeutic window exists in which CDDO-Me protects normal cells from radiation by activating the Nrf2 pathway, but does not protect experimentally transformed or cancer cell lines. bardoxolone methyl 37-44 NFE2 like bZIP transcription factor 2 Homo sapiens 100-104 25047252-3 2014 Since the synthetic triterpenoid methyl-2-cyano-3,12-dioxooleana-1,9-dien-28-oate (CDDO-Me) has been shown to inhibit AKT, MEK-1/2, and NF-kappaB, and to induce caspase-mediated apoptosis, we tested the therapeutic potential of this agent in murine lupus nephritis. bardoxolone methyl 83-90 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 136-145 25047252-3 2014 Since the synthetic triterpenoid methyl-2-cyano-3,12-dioxooleana-1,9-dien-28-oate (CDDO-Me) has been shown to inhibit AKT, MEK-1/2, and NF-kappaB, and to induce caspase-mediated apoptosis, we tested the therapeutic potential of this agent in murine lupus nephritis. bardoxolone methyl 83-90 thymoma viral proto-oncogene 1 Mus musculus 118-121 25047252-4 2014 METHODS: The synthetic triterpenoid CDDO-Me or placebo was administered to 2-month-old B6.Sle1.Sle3 mice or MRL/lpr mice, which develop spontaneous lupus. bardoxolone methyl 36-43 systemic lupus erythmatosus susceptibility 1 Mus musculus 90-94 25047252-4 2014 METHODS: The synthetic triterpenoid CDDO-Me or placebo was administered to 2-month-old B6.Sle1.Sle3 mice or MRL/lpr mice, which develop spontaneous lupus. bardoxolone methyl 36-43 systemic lupus erythmatosus susceptibility 3 Mus musculus 95-99 25047252-3 2014 Since the synthetic triterpenoid methyl-2-cyano-3,12-dioxooleana-1,9-dien-28-oate (CDDO-Me) has been shown to inhibit AKT, MEK-1/2, and NF-kappaB, and to induce caspase-mediated apoptosis, we tested the therapeutic potential of this agent in murine lupus nephritis. bardoxolone methyl 83-90 mitogen-activated protein kinase kinase 1 Mus musculus 123-130 25047252-9 2014 At the mechanistic level, CDDO-Me treatment dampened MEK-1/2, ERK, and STAT-3 signaling within lymphocytes and oxidative stress. bardoxolone methyl 26-33 mitogen-activated protein kinase kinase 1 Mus musculus 53-60 25047252-9 2014 At the mechanistic level, CDDO-Me treatment dampened MEK-1/2, ERK, and STAT-3 signaling within lymphocytes and oxidative stress. bardoxolone methyl 26-33 Eph receptor B2 Mus musculus 62-65 25047252-9 2014 At the mechanistic level, CDDO-Me treatment dampened MEK-1/2, ERK, and STAT-3 signaling within lymphocytes and oxidative stress. bardoxolone methyl 26-33 signal transducer and activator of transcription 3 Mus musculus 71-77 25047252-10 2014 Importantly, the NF-E2-related factor 2 pathway was activated after CDDO-Me treatment, indicating that CDDO-Me may modulate renal damage in lupus via the inhibition of oxidative stress. bardoxolone methyl 68-75 nuclear factor, erythroid derived 2, like 2 Mus musculus 17-39 25047252-10 2014 Importantly, the NF-E2-related factor 2 pathway was activated after CDDO-Me treatment, indicating that CDDO-Me may modulate renal damage in lupus via the inhibition of oxidative stress. bardoxolone methyl 103-110 nuclear factor, erythroid derived 2, like 2 Mus musculus 17-39 25105464-0 2014 The effect of ex vivo CDDO-Me activation on nuclear factor erythroid 2-related factor 2 pathway in white blood cells from patients with septic shock. bardoxolone methyl 22-29 nuclear factor, erythroid derived 2, like 2 Mus musculus 44-87 25105464-2 2014 The objective of this study was to test the hypothesis that ex vivo methyl 2-cyano-3,12-dioxoolean-1,9-dien-28-oate (CDDO-Me) activates NRF2-regulated antioxidant genes in white blood cells from patients with septic shock and protects against LPS-induced inflammation and reactive oxidative species production. bardoxolone methyl 117-124 NFE2 like bZIP transcription factor 2 Homo sapiens 136-140 25105464-4 2014 Real-time polymerase chain reaction was used to quantify the expression of NRF2 target genes (NQO1, HO-1, GCLM, and FTL) and IL-6 in peripheral blood mononuclear cells (PBMCs), monocytes, and neutrophils after CDDO-Me treatment alone or after subsequent LPS exposure. bardoxolone methyl 210-217 NFE2 like bZIP transcription factor 2 Homo sapiens 75-79 25105464-6 2014 Treatment with CDDO-Me increased the gene expression of NQO1 (P = 0.04) and decreased the expression of HO-1 (P = 0.03) in PBMCs from patients with septic shock. bardoxolone methyl 15-22 NAD(P)H quinone dehydrogenase 1 Homo sapiens 56-60 25105464-6 2014 Treatment with CDDO-Me increased the gene expression of NQO1 (P = 0.04) and decreased the expression of HO-1 (P = 0.03) in PBMCs from patients with septic shock. bardoxolone methyl 15-22 heme oxygenase 1 Homo sapiens 104-108 25105464-7 2014 Purified monocytes exhibited significant increases in the expression of NQO1 (P = 0.01) and GCLM (P = 0.003) after CDDO-Me treatment. bardoxolone methyl 115-122 NAD(P)H quinone dehydrogenase 1 Homo sapiens 72-76 25105464-7 2014 Purified monocytes exhibited significant increases in the expression of NQO1 (P = 0.01) and GCLM (P = 0.003) after CDDO-Me treatment. bardoxolone methyl 115-122 glutamate-cysteine ligase modifier subunit Homo sapiens 92-96 25105464-9 2014 Peripheral blood mononuclear cells showed a trend toward increased IL-6 gene expression after CDDO-Me treatment, whereas purified monocytes showed a trend toward decreased IL-6. bardoxolone methyl 94-101 interleukin 6 Homo sapiens 67-71 25105464-14 2014 These data suggest that the NRF2 pathway is differentially responsive to CDDO-Me activation in peripheral blood cells from patients with septic shock and results in increased O2 production. bardoxolone methyl 73-80 NFE2 like bZIP transcription factor 2 Homo sapiens 28-32 24859727-15 2014 At a dose shown to evoke maximal Nrf2 induction in the liver, CDDO-me appeared highly selective for known Nrf2-regulated proteins. bardoxolone methyl 62-69 nuclear factor, erythroid derived 2, like 2 Mus musculus 106-110 25364233-5 2014 The role of the Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1 (Keap1)/the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in the therapeutic activities of CDDO-Me will be discussed. bardoxolone methyl 201-208 kelch like ECH associated protein 1 Homo sapiens 98-103 25364233-5 2014 The role of the Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1 (Keap1)/the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in the therapeutic activities of CDDO-Me will be discussed. bardoxolone methyl 201-208 NFE2 like bZIP transcription factor 2 Homo sapiens 109-152 25364233-5 2014 The role of the Kelch-like erythroid cell-derived protein with CNC homology-associated protein 1 (Keap1)/the nuclear factor erythroid 2-related factor 2 (Nrf2) pathway in the therapeutic activities of CDDO-Me will be discussed. bardoxolone methyl 201-208 NFE2 like bZIP transcription factor 2 Homo sapiens 154-158 25364233-6 2014 CDDO-Me contains alpha,beta-unsaturated carbonyl groups on rings A and C that can generate reversible adducts with the thiol groups of Cys residues in target proteins such as Keap1 and IkappaB kinase. bardoxolone methyl 0-7 kelch like ECH associated protein 1 Homo sapiens 175-180 24859727-10 2014 Only two proteins altered by CDDO-me in WT animals were similarly affected in Nrf2((-/-)) mice, demonstrating the high degree of selectivity of CDDO-me for the Nrf2:Keap1 signalling pathway. bardoxolone methyl 144-151 nuclear factor, erythroid derived 2, like 2 Mus musculus 160-164 24859727-10 2014 Only two proteins altered by CDDO-me in WT animals were similarly affected in Nrf2((-/-)) mice, demonstrating the high degree of selectivity of CDDO-me for the Nrf2:Keap1 signalling pathway. bardoxolone methyl 144-151 kelch-like ECH-associated protein 1 Mus musculus 165-170 24859727-12 2014 For example, CDDO-me (bardoxolone methyl) was investigated in clinical trials for the treatment of acute kidney disease, and dimethyl fumarate, recently approved for reducing relapse rate in multiple sclerosis, is a potent Nrf2 inducer. bardoxolone methyl 13-20 nuclear factor, erythroid derived 2, like 2 Mus musculus 223-227 24859727-15 2014 At a dose shown to evoke maximal Nrf2 induction in the liver, CDDO-me appeared highly selective for known Nrf2-regulated proteins. bardoxolone methyl 62-69 nuclear factor, erythroid derived 2, like 2 Mus musculus 33-37 25112275-1 2014 Here we report that leukemia cell lines and primary CD34+ leukemic blasts exposed to platelet rich plasma (PRP) or platelet lysates (PL) display increased resistance to apoptosis induced by mitochondria-targeted agents ABT-737 and CDDO-Me. bardoxolone methyl 231-238 CD34 molecule Homo sapiens 52-56 24837432-6 2014 Oral administration of CDDO-Me to mice with SMAD4-deficient T cells increased survival and suppressed intestinal epithelial neoplasia by decreasing production of inflammatory mediators and increasing expression of 15-PGDH. bardoxolone methyl 23-30 hydroxyprostaglandin dehydrogenase 15 (NAD) Mus musculus 214-221 25056779-5 2014 In the modified LLNA, CDDO-Me showed a significant decrease in lymph node weight and changes in expressions of the immunosuppression-related genes, Zfp459 and Fmo2. bardoxolone methyl 22-29 zinc finger protein 459 Mus musculus 148-154 25056779-5 2014 In the modified LLNA, CDDO-Me showed a significant decrease in lymph node weight and changes in expressions of the immunosuppression-related genes, Zfp459 and Fmo2. bardoxolone methyl 22-29 flavin containing monooxygenase 2 Mus musculus 159-163 24837432-7 2014 Induction of 15-PGDH by CDDO-Me was dose dependent in epithelial cells and was abrogated following treatment with TGF-beta signaling inhibitors in vitro. bardoxolone methyl 24-31 hydroxyprostaglandin dehydrogenase 15 (NAD) Mus musculus 13-20 24837432-8 2014 Furthermore, CDDO-Me-dependent 15-PGDH induction was not observed in Smad3-/- mice. bardoxolone methyl 13-20 hydroxyprostaglandin dehydrogenase 15 (NAD) Mus musculus 31-38 24903467-1 2014 BACKGROUND: Bardoxolone methyl, an Nrf2-activating and nuclear factor-kappaB-inhibiting semisynthetic oleanane triterpenoid compound, was evaluated in a phase 3 trial (BEACON) in patients with type 2 diabetes mellitus (T2DM) and stage 4 chronic kidney disease (CKD). bardoxolone methyl 12-30 NFE2 like bZIP transcription factor 2 Homo sapiens 35-39 25152840-3 2014 The antiproliferative and apoptosis-inducing effects of CDDO-Me were associated with the inhibition of human telomerase reverse transcriptase (hTERT) mRNA, hTERT protein and reduction in hTERT telomerase activity. bardoxolone methyl 56-63 telomerase reverse transcriptase Homo sapiens 143-148 25152840-3 2014 The antiproliferative and apoptosis-inducing effects of CDDO-Me were associated with the inhibition of human telomerase reverse transcriptase (hTERT) mRNA, hTERT protein and reduction in hTERT telomerase activity. bardoxolone methyl 56-63 telomerase reverse transcriptase Homo sapiens 156-161 25152840-3 2014 The antiproliferative and apoptosis-inducing effects of CDDO-Me were associated with the inhibition of human telomerase reverse transcriptase (hTERT) mRNA, hTERT protein and reduction in hTERT telomerase activity. bardoxolone methyl 56-63 telomerase reverse transcriptase Homo sapiens 156-161 25152840-4 2014 CDDO-Me inhibited multiple transcription factors that regulate hTERT expression positively (Sp1, c-Myc and NF-kappaB) and negatively (CTCF, E2F-1 and MAD1). bardoxolone methyl 0-7 telomerase reverse transcriptase Homo sapiens 63-68 25152840-4 2014 CDDO-Me inhibited multiple transcription factors that regulate hTERT expression positively (Sp1, c-Myc and NF-kappaB) and negatively (CTCF, E2F-1 and MAD1). bardoxolone methyl 0-7 MYC proto-oncogene, bHLH transcription factor Homo sapiens 97-102 25152840-4 2014 CDDO-Me inhibited multiple transcription factors that regulate hTERT expression positively (Sp1, c-Myc and NF-kappaB) and negatively (CTCF, E2F-1 and MAD1). bardoxolone methyl 0-7 CCCTC-binding factor Homo sapiens 134-138 25152840-4 2014 CDDO-Me inhibited multiple transcription factors that regulate hTERT expression positively (Sp1, c-Myc and NF-kappaB) and negatively (CTCF, E2F-1 and MAD1). bardoxolone methyl 0-7 E2F transcription factor 1 Homo sapiens 140-145 25152840-4 2014 CDDO-Me inhibited multiple transcription factors that regulate hTERT expression positively (Sp1, c-Myc and NF-kappaB) and negatively (CTCF, E2F-1 and MAD1). bardoxolone methyl 0-7 MAX dimerization protein 1 Homo sapiens 150-154 25152840-5 2014 CDDO-Me inhibited protein levels of DNA methyl transferases DNMT1 and DNMT3a, which also resulted in hypomethylation of hTERT promoter. bardoxolone methyl 0-7 DNA methyltransferase 1 Homo sapiens 60-65 25152840-5 2014 CDDO-Me inhibited protein levels of DNA methyl transferases DNMT1 and DNMT3a, which also resulted in hypomethylation of hTERT promoter. bardoxolone methyl 0-7 DNA methyltransferase 3 alpha Homo sapiens 70-76 25152840-5 2014 CDDO-Me inhibited protein levels of DNA methyl transferases DNMT1 and DNMT3a, which also resulted in hypomethylation of hTERT promoter. bardoxolone methyl 0-7 telomerase reverse transcriptase Homo sapiens 120-125 24903467-1 2014 BACKGROUND: Bardoxolone methyl, an Nrf2-activating and nuclear factor-kappaB-inhibiting semisynthetic oleanane triterpenoid compound, was evaluated in a phase 3 trial (BEACON) in patients with type 2 diabetes mellitus (T2DM) and stage 4 chronic kidney disease (CKD). bardoxolone methyl 12-30 ubiquitin like 5 Homo sapiens 168-174 23020131-3 2013 Based on screening results, five unique genes (GNPAT, SUMO1, SPINT2, FLI1, and SSX1) significantly potentiated the growth inhibitory effects of CDDO-Me and induced apoptosis in A375, a BRAF mutated melanoma line (P < 0.001). bardoxolone methyl 144-151 glyceronephosphate O-acyltransferase Homo sapiens 47-52 23919312-1 2013 Methyl-2-cyano-3,12 dioxoolean-1,9 diene-28-oate (CDDO-Me) is an antioxidative, anti-inflammatory modulator, which activates the nuclear factor-erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway. bardoxolone methyl 50-57 NFE2 like bZIP transcription factor 2 Homo sapiens 129-172 23919312-1 2013 Methyl-2-cyano-3,12 dioxoolean-1,9 diene-28-oate (CDDO-Me) is an antioxidative, anti-inflammatory modulator, which activates the nuclear factor-erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE) pathway. bardoxolone methyl 50-57 NFE2 like bZIP transcription factor 2 Homo sapiens 174-178 23919312-2 2013 While CDDO-Me has radioprotective activity through Nrf2 activation in vitro and in vivo, its ability to mitigate radiation-induced damage when provided after irradiation has not been studied. bardoxolone methyl 6-13 NFE2 like bZIP transcription factor 2 Homo sapiens 51-55 23919312-5 2013 We observed that treatment with CDDO-Me within 30 min after irradiation improved both DNA damage repair and clonogenic survival independently of Nrf2. bardoxolone methyl 32-39 NFE2 like bZIP transcription factor 2 Homo sapiens 145-149 23919312-6 2013 CDDO-Me activates the epidermal growth factor receptor (EGFR) related DNA repair responses. bardoxolone methyl 0-7 epidermal growth factor receptor Homo sapiens 22-54 23919312-6 2013 CDDO-Me activates the epidermal growth factor receptor (EGFR) related DNA repair responses. bardoxolone methyl 0-7 epidermal growth factor receptor Homo sapiens 56-60 23919312-7 2013 In the presence of CDDO-Me, EGFR is phosphorylated and translocates into the nucleus where it interacts with DNA-PKcs. bardoxolone methyl 19-26 epidermal growth factor receptor Homo sapiens 28-32 23919312-7 2013 In the presence of CDDO-Me, EGFR is phosphorylated and translocates into the nucleus where it interacts with DNA-PKcs. bardoxolone methyl 19-26 protein kinase, DNA-activated, catalytic subunit Homo sapiens 109-117 23919312-8 2013 CDDO-Me-mediated mitigation activity can be abrogated through depletion of EGFR, ectopic overexpression of mutant EGFR or inhibition of DNA-PKcs. bardoxolone methyl 0-7 epidermal growth factor receptor Homo sapiens 75-79 23919312-8 2013 CDDO-Me-mediated mitigation activity can be abrogated through depletion of EGFR, ectopic overexpression of mutant EGFR or inhibition of DNA-PKcs. bardoxolone methyl 0-7 epidermal growth factor receptor Homo sapiens 114-118 23919312-8 2013 CDDO-Me-mediated mitigation activity can be abrogated through depletion of EGFR, ectopic overexpression of mutant EGFR or inhibition of DNA-PKcs. bardoxolone methyl 0-7 protein kinase, DNA-activated, catalytic subunit Homo sapiens 136-144 23919312-10 2013 These results suggest that targeting EGFR using CDDO-Me is a promising radiation mitigator with potential utility for first responders to nuclear accidents. bardoxolone methyl 48-55 epidermal growth factor receptor Homo sapiens 37-41 23741300-7 2013 On day 7, CDDO-Me reduced total BALF protein by 50%, alveolar macrophage infiltration by 40%, neutrophil infiltration by 90% (p<=0.01), inhibited production of the inflammatory cytokines KC and IL-6 by over 90% (p<=0.001), and excess production of the pro-fibrotic cytokine TGFbeta by 50%. bardoxolone methyl 10-17 interleukin 6 Homo sapiens 197-201 23741300-7 2013 On day 7, CDDO-Me reduced total BALF protein by 50%, alveolar macrophage infiltration by 40%, neutrophil infiltration by 90% (p<=0.01), inhibited production of the inflammatory cytokines KC and IL-6 by over 90% (p<=0.001), and excess production of the pro-fibrotic cytokine TGFbeta by 50%. bardoxolone methyl 10-17 transforming growth factor beta 1 Homo sapiens 280-287 23741300-8 2013 CDDO-Me also inhibited alpha-smooth muscle actin and fibronectin mRNA by 50% (p<=0.05). bardoxolone methyl 0-7 fibronectin 1 Homo sapiens 53-64 23486104-5 2013 Pretreatment of cells with N-acetylcycsteine, a general purpose antioxidant or overexpression of glutathione peroxidase (GPx) or superoxide dismutase-1 (SOD-1) blocked the telomerase inhibitory activity of CDDO-Me. bardoxolone methyl 206-213 superoxide dismutase 1 Homo sapiens 129-151 23486104-5 2013 Pretreatment of cells with N-acetylcycsteine, a general purpose antioxidant or overexpression of glutathione peroxidase (GPx) or superoxide dismutase-1 (SOD-1) blocked the telomerase inhibitory activity of CDDO-Me. bardoxolone methyl 206-213 superoxide dismutase 1 Homo sapiens 153-158 23486104-6 2013 Furthermore, blocking ROS generation also prevented the inhibition of hTERT gene expression, hTERT protein production and expression of a number of hTERT-regulatory proteins by CDDO-Me (e.g., c-Myc, Sp1, NF-kappaB and p-Akt). bardoxolone methyl 177-184 MYC proto-oncogene, bHLH transcription factor Homo sapiens 192-197 23486104-8 2013 Together, these data suggest that inhibition of telomerase activity by CDDO-Me is mediated through a ROS-dependent mechanism; however, more work is needed to fully understand the role of ROS in down-regulation of hTERT gene and hTERT-regulatory proteins by CDDO-Me. bardoxolone methyl 71-78 telomerase reverse transcriptase Homo sapiens 213-218 23267148-5 2013 Likewise, NAC prevented the CDDO-Me-caused binding of fluorescein isothiocyanate (FITC)-tagged annexin V, cleavage of poly ADP-ribose polymerase-1 (PARP-1), procaspases-3, -8 and -9 and loss of mitochondrial membrane potential. bardoxolone methyl 28-35 annexin A5 Homo sapiens 95-104 23267148-5 2013 Likewise, NAC prevented the CDDO-Me-caused binding of fluorescein isothiocyanate (FITC)-tagged annexin V, cleavage of poly ADP-ribose polymerase-1 (PARP-1), procaspases-3, -8 and -9 and loss of mitochondrial membrane potential. bardoxolone methyl 28-35 poly(ADP-ribose) polymerase 1 Homo sapiens 118-146 23267148-5 2013 Likewise, NAC prevented the CDDO-Me-caused binding of fluorescein isothiocyanate (FITC)-tagged annexin V, cleavage of poly ADP-ribose polymerase-1 (PARP-1), procaspases-3, -8 and -9 and loss of mitochondrial membrane potential. bardoxolone methyl 28-35 poly(ADP-ribose) polymerase 1 Homo sapiens 148-154 23267148-5 2013 Likewise, NAC prevented the CDDO-Me-caused binding of fluorescein isothiocyanate (FITC)-tagged annexin V, cleavage of poly ADP-ribose polymerase-1 (PARP-1), procaspases-3, -8 and -9 and loss of mitochondrial membrane potential. bardoxolone methyl 28-35 caspase 3 Homo sapiens 157-181 23267148-7 2013 Together, these results indicate the pivotal role ROS play in the antiproliferative- and apoptosis-inducing activity of CDDO-Me in ovarian cancer cells; however, the role of ROS in the down-regulation of prosurvival AKT, mTOR, NF-kappaB and antiapoptotic BCL-2, BCL-xL, c-IAP1 and survivin warrants further investigation. bardoxolone methyl 120-127 AKT serine/threonine kinase 1 Homo sapiens 216-219 23267148-7 2013 Together, these results indicate the pivotal role ROS play in the antiproliferative- and apoptosis-inducing activity of CDDO-Me in ovarian cancer cells; however, the role of ROS in the down-regulation of prosurvival AKT, mTOR, NF-kappaB and antiapoptotic BCL-2, BCL-xL, c-IAP1 and survivin warrants further investigation. bardoxolone methyl 120-127 mechanistic target of rapamycin kinase Homo sapiens 221-225 23267148-7 2013 Together, these results indicate the pivotal role ROS play in the antiproliferative- and apoptosis-inducing activity of CDDO-Me in ovarian cancer cells; however, the role of ROS in the down-regulation of prosurvival AKT, mTOR, NF-kappaB and antiapoptotic BCL-2, BCL-xL, c-IAP1 and survivin warrants further investigation. bardoxolone methyl 120-127 nuclear factor kappa B subunit 1 Homo sapiens 227-236 23267148-7 2013 Together, these results indicate the pivotal role ROS play in the antiproliferative- and apoptosis-inducing activity of CDDO-Me in ovarian cancer cells; however, the role of ROS in the down-regulation of prosurvival AKT, mTOR, NF-kappaB and antiapoptotic BCL-2, BCL-xL, c-IAP1 and survivin warrants further investigation. bardoxolone methyl 120-127 BCL2 apoptosis regulator Homo sapiens 255-260 23267148-7 2013 Together, these results indicate the pivotal role ROS play in the antiproliferative- and apoptosis-inducing activity of CDDO-Me in ovarian cancer cells; however, the role of ROS in the down-regulation of prosurvival AKT, mTOR, NF-kappaB and antiapoptotic BCL-2, BCL-xL, c-IAP1 and survivin warrants further investigation. bardoxolone methyl 120-127 BCL2 like 1 Homo sapiens 262-268 23267148-7 2013 Together, these results indicate the pivotal role ROS play in the antiproliferative- and apoptosis-inducing activity of CDDO-Me in ovarian cancer cells; however, the role of ROS in the down-regulation of prosurvival AKT, mTOR, NF-kappaB and antiapoptotic BCL-2, BCL-xL, c-IAP1 and survivin warrants further investigation. bardoxolone methyl 120-127 baculoviral IAP repeat containing 2 Homo sapiens 270-276 23783243-3 2013 An increasing body of evidence supports a vital physiological role for Nrf2 in protection of the kidney against a number of diseases, and the pharmacological induction of Nrf2 by bardoxolone methyl (methyl-2-cyano 3,12-dioxooleano-1,9-dien-28-oate, CDDO-Me) has shown promise for the management of such pathologies. bardoxolone methyl 249-256 NFE2 like bZIP transcription factor 2 Homo sapiens 171-175 23020131-3 2013 Based on screening results, five unique genes (GNPAT, SUMO1, SPINT2, FLI1, and SSX1) significantly potentiated the growth inhibitory effects of CDDO-Me and induced apoptosis in A375, a BRAF mutated melanoma line (P < 0.001). bardoxolone methyl 144-151 small ubiquitin like modifier 1 Homo sapiens 54-59 23020131-3 2013 Based on screening results, five unique genes (GNPAT, SUMO1, SPINT2, FLI1, and SSX1) significantly potentiated the growth inhibitory effects of CDDO-Me and induced apoptosis in A375, a BRAF mutated melanoma line (P < 0.001). bardoxolone methyl 144-151 serine peptidase inhibitor, Kunitz type 2 Homo sapiens 61-67 23020131-3 2013 Based on screening results, five unique genes (GNPAT, SUMO1, SPINT2, FLI1, and SSX1) significantly potentiated the growth inhibitory effects of CDDO-Me and induced apoptosis in A375, a BRAF mutated melanoma line (P < 0.001). bardoxolone methyl 144-151 Fli-1 proto-oncogene, ETS transcription factor Homo sapiens 69-73 23020131-3 2013 Based on screening results, five unique genes (GNPAT, SUMO1, SPINT2, FLI1, and SSX1) significantly potentiated the growth inhibitory effects of CDDO-Me and induced apoptosis in A375, a BRAF mutated melanoma line (P < 0.001). bardoxolone methyl 144-151 SSX family member 1 Homo sapiens 79-83 23020131-3 2013 Based on screening results, five unique genes (GNPAT, SUMO1, SPINT2, FLI1, and SSX1) significantly potentiated the growth inhibitory effects of CDDO-Me and induced apoptosis in A375, a BRAF mutated melanoma line (P < 0.001). bardoxolone methyl 144-151 B-Raf proto-oncogene, serine/threonine kinase Homo sapiens 185-189 23020131-6 2013 Our findings indicate that GNPAT, SUMO1, SPINT2, FLI1, and SSX1 play critical roles in synergy with inflammation pathways in modulating melanoma cell survival and could serve as sensitizing targets to enhance CDDO-Me efficacy in melanoma growth control. bardoxolone methyl 209-216 glyceronephosphate O-acyltransferase Homo sapiens 27-32 22177954-1 2012 Our previous studies have shown that methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me), a oleanane synthetic triterpenoid induces apoptosis in prostate cancer cells by inhibiting the Akt/NF-kappaB/mTOR signaling cascade; however, the mechanism by which CDDO-Me inhibits Akt/NF-kappaB/mTOR signaling has remained undetermined. bardoxolone methyl 91-98 nuclear factor kappa B subunit 1 Homo sapiens 287-296 23519253-0 2012 Telomerase reverse transcriptase (TERT) is a therapeutic target of oleanane triterpenoid CDDO-Me in prostate cancer. bardoxolone methyl 89-96 telomerase reverse transcriptase Homo sapiens 34-38 23519253-2 2012 However, the effect of CDDO-Me on human telomerase reverse transcriptase (hTERT) and its telomerase activity in prostate cancer cells has not been studied. bardoxolone methyl 23-30 telomerase reverse transcriptase Homo sapiens 74-79 23519253-3 2012 We investigated the role of hTERT in mediating the anticancer activity of CDDO-Me in prostate cancer cells in vitro and in vivo. bardoxolone methyl 74-81 telomerase reverse transcriptase Homo sapiens 28-33 23519253-4 2012 The inhibition of cell proliferation and induction of apoptosis by CDDO-Me in LNCaP and PC-3 prostate cancer cell lines was associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT transcriptionally and post-translationally. bardoxolone methyl 67-74 telomerase reverse transcriptase Homo sapiens 158-163 23519253-4 2012 The inhibition of cell proliferation and induction of apoptosis by CDDO-Me in LNCaP and PC-3 prostate cancer cell lines was associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT transcriptionally and post-translationally. bardoxolone methyl 67-74 telomerase reverse transcriptase Homo sapiens 181-186 23519253-4 2012 The inhibition of cell proliferation and induction of apoptosis by CDDO-Me in LNCaP and PC-3 prostate cancer cell lines was associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT transcriptionally and post-translationally. bardoxolone methyl 67-74 telomerase reverse transcriptase Homo sapiens 181-186 23519253-5 2012 Furthermore, ablation of hTERT protein increased the sensitivity of cancer cells to CDDO-Me, whereas its overexpression rendered them resistant to CDDO-Me. bardoxolone methyl 84-91 telomerase reverse transcriptase Homo sapiens 25-30 23519253-5 2012 Furthermore, ablation of hTERT protein increased the sensitivity of cancer cells to CDDO-Me, whereas its overexpression rendered them resistant to CDDO-Me. bardoxolone methyl 147-154 telomerase reverse transcriptase Homo sapiens 25-30 23519253-6 2012 In addition, inhibition of progression of preneoplastic lesions (i.e., low and high-grade prostate intraepithelial neoplasms, PINs) to adenocarcinoma of the prostate by CDDO-Me in TRAMP mice was associated with significant decrease in TERT and its regulatory proteins in the prostate gland. bardoxolone methyl 169-176 tumor necrosis factor receptor superfamily, member 25 Mus musculus 180-185 23519253-6 2012 In addition, inhibition of progression of preneoplastic lesions (i.e., low and high-grade prostate intraepithelial neoplasms, PINs) to adenocarcinoma of the prostate by CDDO-Me in TRAMP mice was associated with significant decrease in TERT and its regulatory proteins in the prostate gland. bardoxolone methyl 169-176 telomerase reverse transcriptase Mus musculus 235-239 22747345-2 2012 Bardoxolone methyl is a potent, orally bioavailable Nrf-2 agonist. bardoxolone methyl 0-18 NFE2 like bZIP transcription factor 2 Homo sapiens 52-57 22634319-1 2012 PURPOSE: Bardoxolone methyl, a novel synthetic triterpenoid and antioxidant inflammation modulator, potently induces Nrf2 and inhibits NF-kappaB and Janus-activated kinase/STAT signaling. bardoxolone methyl 9-27 NFE2 like bZIP transcription factor 2 Homo sapiens 117-121 23045680-5 2012 Here, we report that Nrf2 activation by the synthetic triterpenoids, bardoxolone methyl (BARD) and 2-cyano-3,12-dioxooleana-1,9 (11)-dien-28-oic acid-ethyl amide, protects colonic epithelial cells against IR-induced damage, in part, by enhancing signaling of the DNA damage response. bardoxolone methyl 69-87 nuclear factor, erythroid derived 2, like 2 Mus musculus 21-25 22609405-0 2012 Inhibition of cell proliferation and induction of apoptosis by oleanane triterpenoid (CDDO-Me) in pancreatic cancer cells is associated with the suppression of hTERT gene expression and its telomerase activity. bardoxolone methyl 86-93 telomerase reverse transcriptase Homo sapiens 160-165 22609405-3 2012 Here we report for the first time that inhibition of cell proliferation and induction of apoptosis by CDDO-Me in pancreatic cancer cell lines is associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT expression and activity. bardoxolone methyl 102-109 telomerase reverse transcriptase Homo sapiens 179-184 22609405-3 2012 Here we report for the first time that inhibition of cell proliferation and induction of apoptosis by CDDO-Me in pancreatic cancer cell lines is associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT expression and activity. bardoxolone methyl 102-109 telomerase reverse transcriptase Homo sapiens 202-207 22609405-3 2012 Here we report for the first time that inhibition of cell proliferation and induction of apoptosis by CDDO-Me in pancreatic cancer cell lines is associated with the inhibition of hTERT gene expression, hTERT telomerase activity and a number of proteins that regulate hTERT expression and activity. bardoxolone methyl 102-109 telomerase reverse transcriptase Homo sapiens 202-207 22609405-4 2012 Furthermore, abrogation or overexpression of hTERT protein altered the susceptibility of tumor cells to CDDO-Me. bardoxolone methyl 104-111 telomerase reverse transcriptase Homo sapiens 45-50 22609405-5 2012 These findings suggest that telomerase (hTERT) is a relevant target of CDDO-Me in pancreatic cancer cells. bardoxolone methyl 71-78 telomerase reverse transcriptase Homo sapiens 40-45 22177954-0 2012 Oleanane triterpenoid CDDO-Me inhibits Akt activity without affecting PDK1 kinase or PP2A phosphatase activity in cancer cells. bardoxolone methyl 22-29 AKT serine/threonine kinase 1 Homo sapiens 39-42 22177954-1 2012 Our previous studies have shown that methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me), a oleanane synthetic triterpenoid induces apoptosis in prostate cancer cells by inhibiting the Akt/NF-kappaB/mTOR signaling cascade; however, the mechanism by which CDDO-Me inhibits Akt/NF-kappaB/mTOR signaling has remained undetermined. bardoxolone methyl 91-98 AKT serine/threonine kinase 1 Homo sapiens 196-199 22177954-1 2012 Our previous studies have shown that methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me), a oleanane synthetic triterpenoid induces apoptosis in prostate cancer cells by inhibiting the Akt/NF-kappaB/mTOR signaling cascade; however, the mechanism by which CDDO-Me inhibits Akt/NF-kappaB/mTOR signaling has remained undetermined. bardoxolone methyl 91-98 nuclear factor kappa B subunit 1 Homo sapiens 200-209 22177954-1 2012 Our previous studies have shown that methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me), a oleanane synthetic triterpenoid induces apoptosis in prostate cancer cells by inhibiting the Akt/NF-kappaB/mTOR signaling cascade; however, the mechanism by which CDDO-Me inhibits Akt/NF-kappaB/mTOR signaling has remained undetermined. bardoxolone methyl 91-98 mechanistic target of rapamycin kinase Homo sapiens 210-214 22177954-1 2012 Our previous studies have shown that methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me), a oleanane synthetic triterpenoid induces apoptosis in prostate cancer cells by inhibiting the Akt/NF-kappaB/mTOR signaling cascade; however, the mechanism by which CDDO-Me inhibits Akt/NF-kappaB/mTOR signaling has remained undetermined. bardoxolone methyl 91-98 AKT serine/threonine kinase 1 Homo sapiens 283-286 22177954-1 2012 Our previous studies have shown that methyl-2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate (CDDO-Me), a oleanane synthetic triterpenoid induces apoptosis in prostate cancer cells by inhibiting the Akt/NF-kappaB/mTOR signaling cascade; however, the mechanism by which CDDO-Me inhibits Akt/NF-kappaB/mTOR signaling has remained undetermined. bardoxolone methyl 91-98 mechanistic target of rapamycin kinase Homo sapiens 297-301 22177954-2 2012 Present studies show that Akt plays a critical role in the response of prostate cancer cells to CDDO-Me. bardoxolone methyl 96-103 AKT serine/threonine kinase 1 Homo sapiens 26-29 22177954-3 2012 Silencing of Akt sensitized PC-3 cells to CDDO-Me, whereas its overexpression rendered them resistant to CDDO-Me. bardoxolone methyl 42-49 AKT serine/threonine kinase 1 Homo sapiens 13-16 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 28-35 AKT serine/threonine kinase 1 Homo sapiens 39-42 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 28-35 nuclear factor kappa B subunit 1 Homo sapiens 66-75 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 28-35 mechanistic target of rapamycin kinase Homo sapiens 80-84 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 28-35 AKT serine/threonine kinase 1 Homo sapiens 127-130 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 28-35 AKT serine/threonine kinase 1 Homo sapiens 127-130 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 97-104 AKT serine/threonine kinase 1 Homo sapiens 39-42 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 97-104 nuclear factor kappa B subunit 1 Homo sapiens 66-75 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 97-104 mechanistic target of rapamycin kinase Homo sapiens 80-84 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 97-104 AKT serine/threonine kinase 1 Homo sapiens 127-130 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 97-104 pyruvate dehydrogenase kinase 1 Homo sapiens 229-233 22177954-4 2012 Evaluation of the effect of CDDO-Me on Akt which lies upstream of NF-kappaB and mTOR showed that CDDO-Me directly inhibits the Akt kinase activity in cell-free kinase activity assay and in vivo without modulating the activity of PDK1, the upstream kinase that phosphorylates and activates Akt. bardoxolone methyl 97-104 AKT serine/threonine kinase 1 Homo sapiens 127-130 22177954-6 2012 Further, inhibition of p-Akt by CDDO-Me was not attributable to an increase in the activity of protein phosphatase 2A (PP2A) or PH domain/leucine-rich repeat protein phosphatase1 (PHLPP1) both of which dephosphorylate p-Akt. bardoxolone methyl 32-39 AKT serine/threonine kinase 1 Homo sapiens 25-28 22177954-7 2012 These findings show that Akt is a direct target of CDDO-Me in the Akt/NF-kappaB/mTOR prosurvival signaling axis. bardoxolone methyl 51-58 AKT serine/threonine kinase 1 Homo sapiens 25-28 22177954-7 2012 These findings show that Akt is a direct target of CDDO-Me in the Akt/NF-kappaB/mTOR prosurvival signaling axis. bardoxolone methyl 51-58 AKT serine/threonine kinase 1 Homo sapiens 66-69 22177954-7 2012 These findings show that Akt is a direct target of CDDO-Me in the Akt/NF-kappaB/mTOR prosurvival signaling axis. bardoxolone methyl 51-58 nuclear factor kappa B subunit 1 Homo sapiens 70-79 22177954-7 2012 These findings show that Akt is a direct target of CDDO-Me in the Akt/NF-kappaB/mTOR prosurvival signaling axis. bardoxolone methyl 51-58 mechanistic target of rapamycin kinase Homo sapiens 80-84 22178289-3 2012 In this study, we tested the relative effectiveness and mechanisms of action of two electrophilic peroxisome proliferator-activated receptor gamma (PPARgamma) ligands: cyano-3,12-dioxolean-1,9-dien-28-oic acid-methyl ester (CDDO-Me) and 15-deoxy-Delta(-12,14)-prostaglandin J(2) (15d-PGJ(2)) for inhibiting TGFbeta-induced myofibroblast differentiation in vitro. bardoxolone methyl 224-231 peroxisome proliferator activated receptor gamma Homo sapiens 148-157 21933912-3 2012 Because the methyl ester of the synthetic triterpenoid 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid (CDDO-Me) is a potent chemopreventive agent, we tested its efficacy in a highly relevant mouse model of BRCA1-mutated breast cancer. bardoxolone methyl 106-113 breast cancer 1, early onset Mus musculus 209-214 21933912-5 2012 CDDO-Me significantly (P < 0.05) delayed tumor development in the Brca1-mutated mice by an average of 5.2 weeks. bardoxolone methyl 0-7 breast cancer 1, early onset Mus musculus 69-74 21933912-6 2012 We also observed that levels of ErbB2, p-ErbB2, and cyclin D1 increased in a time-dependent manner in the mammary glands in Brca1-deficient mice, and CDDO-Me inhibited the constitutive phosphorylation of ErbB2 in tumor tissues from these mice. bardoxolone methyl 150-157 erb-b2 receptor tyrosine kinase 2 Mus musculus 32-37 21933912-6 2012 We also observed that levels of ErbB2, p-ErbB2, and cyclin D1 increased in a time-dependent manner in the mammary glands in Brca1-deficient mice, and CDDO-Me inhibited the constitutive phosphorylation of ErbB2 in tumor tissues from these mice. bardoxolone methyl 150-157 erb-b2 receptor tyrosine kinase 2 Mus musculus 41-46 21933912-6 2012 We also observed that levels of ErbB2, p-ErbB2, and cyclin D1 increased in a time-dependent manner in the mammary glands in Brca1-deficient mice, and CDDO-Me inhibited the constitutive phosphorylation of ErbB2 in tumor tissues from these mice. bardoxolone methyl 150-157 erb-b2 receptor tyrosine kinase 2 Mus musculus 41-46 21933912-8 2012 These results suggest that CDDO-Me has the potential to prevent BRCA1-mutated breast cancer. bardoxolone methyl 27-34 breast cancer 1, early onset Mus musculus 64-69 22178289-9 2012 Both electrophilic PPARgamma ligands (CDDO-Me and 15d-PGJ(2)) potently inhibited TGFbeta-induced myofibroblast differentiation, but PPARgamma was only partially required for inhibition of myofibroblast differentiation by either agent. bardoxolone methyl 38-45 peroxisome proliferator activated receptor gamma Homo sapiens 19-28 22178289-9 2012 Both electrophilic PPARgamma ligands (CDDO-Me and 15d-PGJ(2)) potently inhibited TGFbeta-induced myofibroblast differentiation, but PPARgamma was only partially required for inhibition of myofibroblast differentiation by either agent. bardoxolone methyl 38-45 transforming growth factor beta 1 Homo sapiens 81-88 22178289-11 2012 CDDO-Me and 15d-PGJ(2) are strong inhibitors of TGFbeta-induced corneal fibroblast to myofibroblast differentiation in vitro, suggesting this class of agents as potential novel therapies for corneal scarring warranting further study in pre-clinical animal models. bardoxolone methyl 0-7 transforming growth factor beta 1 Homo sapiens 48-55 22946344-5 2012 Pretreatment with N-acetylcysteine (NAC) or overexpression of glutathione peroxidase (GPx) or superoxide dismutase-1 (SOD-1) blocked the antiproliferative effects of CDDO-Me. bardoxolone methyl 166-173 superoxide dismutase 1 Homo sapiens 94-116 22946344-5 2012 Pretreatment with N-acetylcysteine (NAC) or overexpression of glutathione peroxidase (GPx) or superoxide dismutase-1 (SOD-1) blocked the antiproliferative effects of CDDO-Me. bardoxolone methyl 166-173 superoxide dismutase 1 Homo sapiens 118-123 22946344-6 2012 Likewise, NAC prevented the induction of apoptosis (annexin V-FITC binding and cleavage of PARP-1 and procaspases-3,-8 and -9) and reversed the loss of mitochondrial membrane potential and release of cytochrome c from mitochondria by CDDO-Me. bardoxolone methyl 234-241 cytochrome c, somatic Homo sapiens 200-212 22946344-7 2012 CDDO-Me down-regulated p-Akt, p-mTOR and NF-kappaB (p65) but increased the activation of Erk1/2 and NAC blocked the modulation of these cell signaling proteins by CDDO-Me. bardoxolone methyl 0-7 AKT serine/threonine kinase 1 Homo sapiens 25-28 22946344-7 2012 CDDO-Me down-regulated p-Akt, p-mTOR and NF-kappaB (p65) but increased the activation of Erk1/2 and NAC blocked the modulation of these cell signaling proteins by CDDO-Me. bardoxolone methyl 0-7 mechanistic target of rapamycin kinase Homo sapiens 32-36 22946344-7 2012 CDDO-Me down-regulated p-Akt, p-mTOR and NF-kappaB (p65) but increased the activation of Erk1/2 and NAC blocked the modulation of these cell signaling proteins by CDDO-Me. bardoxolone methyl 0-7 RELA proto-oncogene, NF-kB subunit Homo sapiens 52-55 22946344-7 2012 CDDO-Me down-regulated p-Akt, p-mTOR and NF-kappaB (p65) but increased the activation of Erk1/2 and NAC blocked the modulation of these cell signaling proteins by CDDO-Me. bardoxolone methyl 0-7 mitogen-activated protein kinase 3 Homo sapiens 89-95 22946344-7 2012 CDDO-Me down-regulated p-Akt, p-mTOR and NF-kappaB (p65) but increased the activation of Erk1/2 and NAC blocked the modulation of these cell signaling proteins by CDDO-Me. bardoxolone methyl 163-170 mitogen-activated protein kinase 3 Homo sapiens 89-95 22110186-0 2011 Synthetic oleanane triterpenoid, CDDO-Me, induces apoptosis in ovarian cancer cells by inhibiting prosurvival AKT/NF-kappaB/mTOR signaling. bardoxolone methyl 33-40 AKT serine/threonine kinase 1 Homo sapiens 110-113 22110186-0 2011 Synthetic oleanane triterpenoid, CDDO-Me, induces apoptosis in ovarian cancer cells by inhibiting prosurvival AKT/NF-kappaB/mTOR signaling. bardoxolone methyl 33-40 nuclear factor kappa B subunit 1 Homo sapiens 114-123 22110186-6 2011 Abrogation of AKT which regulates both NF-kappaB and mTOR increased the sensitivity of tumor cells to CDDO-Me. bardoxolone methyl 102-109 AKT serine/threonine kinase 1 Homo sapiens 14-17 22110186-0 2011 Synthetic oleanane triterpenoid, CDDO-Me, induces apoptosis in ovarian cancer cells by inhibiting prosurvival AKT/NF-kappaB/mTOR signaling. bardoxolone methyl 33-40 mechanistic target of rapamycin kinase Homo sapiens 124-128 22110186-6 2011 Abrogation of AKT which regulates both NF-kappaB and mTOR increased the sensitivity of tumor cells to CDDO-Me. bardoxolone methyl 102-109 nuclear factor kappa B subunit 1 Homo sapiens 39-48 22110186-6 2011 Abrogation of AKT which regulates both NF-kappaB and mTOR increased the sensitivity of tumor cells to CDDO-Me. bardoxolone methyl 102-109 mechanistic target of rapamycin kinase Homo sapiens 53-57 22110186-3 2011 CDDO-Me strongly inhibited the growth of ovarian cancer cells by inducing apoptosis characterized by increased annexin V binding, cleavage of poly (ADP-ribose) polymerase (PARP-1) and procaspases-3, -8 and -9. bardoxolone methyl 0-7 poly(ADP-ribose) polymerase 1 Homo sapiens 142-170 22110186-7 2011 Thus, these data showing strong growth-inhibitory and apoptosis-inducing activity of CDDO-Me for ovarian cancer cells through the inhibition of AKT/ NF-kappaB/mTOR signaling pathway provide basis for evaluation of CDDO-Me for ovarian cancer. bardoxolone methyl 85-92 AKT serine/threonine kinase 1 Homo sapiens 144-147 22110186-3 2011 CDDO-Me strongly inhibited the growth of ovarian cancer cells by inducing apoptosis characterized by increased annexin V binding, cleavage of poly (ADP-ribose) polymerase (PARP-1) and procaspases-3, -8 and -9. bardoxolone methyl 0-7 caspase 3 Homo sapiens 172-208 22110186-7 2011 Thus, these data showing strong growth-inhibitory and apoptosis-inducing activity of CDDO-Me for ovarian cancer cells through the inhibition of AKT/ NF-kappaB/mTOR signaling pathway provide basis for evaluation of CDDO-Me for ovarian cancer. bardoxolone methyl 85-92 nuclear factor kappa B subunit 1 Homo sapiens 149-158 22110186-7 2011 Thus, these data showing strong growth-inhibitory and apoptosis-inducing activity of CDDO-Me for ovarian cancer cells through the inhibition of AKT/ NF-kappaB/mTOR signaling pathway provide basis for evaluation of CDDO-Me for ovarian cancer. bardoxolone methyl 85-92 mechanistic target of rapamycin kinase Homo sapiens 159-163 21799944-5 2010 CDDO-Me inhibited the growth of both K-ras mutated (MiaPaca2, Panc1 and Capan2) and wild-type K-ras (BxPC3) pancreatic cancer cells at very low concentrations. bardoxolone methyl 0-7 KRAS proto-oncogene, GTPase Homo sapiens 37-42 21961053-0 2011 Prevention of Prostate Cancer with Oleanane Synthetic Triterpenoid CDDO-Me in the TRAMP Mouse Model of Prostate Cancer. bardoxolone methyl 67-74 translocating chain-associating membrane protein 1 Mus musculus 82-87 21961053-3 2011 CDDO-Me inhibited the growth of murine TRAMPC-1 prostate cancer cells by inducing apoptosis through the inhibition of antiapoptotic p-Akt, p-mTOR and NF-kappaB. bardoxolone methyl 0-7 thymoma viral proto-oncogene 1 Mus musculus 134-137 21961053-3 2011 CDDO-Me inhibited the growth of murine TRAMPC-1 prostate cancer cells by inducing apoptosis through the inhibition of antiapoptotic p-Akt, p-mTOR and NF-kappaB. bardoxolone methyl 0-7 mechanistic target of rapamycin kinase Mus musculus 141-145 21961053-4 2011 Early intervention with CDDO-Me (7.5 mg/kg) initiated at five weeks of age for 20 wk inhibited the progression of the preneoplastic lesions (low-grade PIN and high-grade-PIN) to adenocarcinoma in the dorsolateral prostate (DLP) and ventral prostate (VP) lobes of TRAMP mice. bardoxolone methyl 24-31 translocating chain-associating membrane protein 1 Mus musculus 263-268 21961053-7 2011 Treatment with CDDO-Me inhibited the expression of prosurvival p-Akt and NF-kappaB in the prostate and knocking-down Akt in TRAMPC-1 tumor cells sensitized them to CDDO-Me. bardoxolone methyl 15-22 thymoma viral proto-oncogene 1 Mus musculus 65-68 21961053-7 2011 Treatment with CDDO-Me inhibited the expression of prosurvival p-Akt and NF-kappaB in the prostate and knocking-down Akt in TRAMPC-1 tumor cells sensitized them to CDDO-Me. bardoxolone methyl 15-22 thymoma viral proto-oncogene 1 Mus musculus 117-120 21961053-8 2011 These findings indicated that Akt is a target for apoptoxicity in TRAMPC-1 cells in vitro and potentially a target of CDDO-Me for inhibition of prostate cancer in vivo. bardoxolone methyl 118-125 thymoma viral proto-oncogene 1 Mus musculus 30-33 21723306-7 2011 Using NCTC2544 human keratinocytes, we found that both sulforaphane and methyl-2-cyano-3,12-dioxooleana-1,9-dien-28-oate (CDDO-Me) stimulated nuclear localization of Nrf2 and induced expression of the GSH synthesis gene, GCLM. bardoxolone methyl 122-129 NFE2 like bZIP transcription factor 2 Homo sapiens 166-170 21723306-7 2011 Using NCTC2544 human keratinocytes, we found that both sulforaphane and methyl-2-cyano-3,12-dioxooleana-1,9-dien-28-oate (CDDO-Me) stimulated nuclear localization of Nrf2 and induced expression of the GSH synthesis gene, GCLM. bardoxolone methyl 122-129 glutamate-cysteine ligase modifier subunit Homo sapiens 221-225 21545836-7 2011 Treatment with the Nrf2 activator CDDO-Me increased antioxidant gene expression and normalized I/R-induced superoxide in the retina in wild-type but not Nrf2(-/-) mice. bardoxolone methyl 34-41 nuclear factor, erythroid derived 2, like 2 Mus musculus 19-23 21699019-5 2011 CDDO-Me induced apoptosis as demonstrated by the cleavage of PARP-1, activation of procaspases -3, -8, and -9 and mitochondrial depolarization and NAC blocked the activation of these apoptosis related processes. bardoxolone methyl 0-7 poly(ADP-ribose) polymerase 1 Homo sapiens 61-109 21699019-6 2011 Furthermore, induction of apoptosis by CDDO-Me was associated with the inhibition of antiapoptotic/ prosurvival Akt, mTOR and NF-kappaB signaling proteins and the inhibition of these signaling molecules was blocked by NAG. bardoxolone methyl 39-46 AKT serine/threonine kinase 1 Homo sapiens 112-115 21699019-6 2011 Furthermore, induction of apoptosis by CDDO-Me was associated with the inhibition of antiapoptotic/ prosurvival Akt, mTOR and NF-kappaB signaling proteins and the inhibition of these signaling molecules was blocked by NAG. bardoxolone methyl 39-46 mechanistic target of rapamycin kinase Homo sapiens 117-121 20956520-2 2010 Here we report that CDDO-Me ameliorates diabetes in high fat diet-fed type 2 diabetic mice and in Lepr(db/db) mice. bardoxolone methyl 20-27 leptin receptor Mus musculus 98-102 20956520-8 2010 CDDO-Me activates AMP-activated protein kinase (AMPK) and via LKB1 activation in muscle and liver in vivo. bardoxolone methyl 0-7 serine/threonine kinase 11 Mus musculus 62-66 20956520-9 2010 Treatment of isolated hepatocytes with CDDO-Me directly stimulates AMPK activity and LKB1 phosphorylation and decreases acetyl-coA carboxylase activity; it also down-regulates lipogenic gene expression, suppresses gluconeogenesis, and increases glucose uptake. bardoxolone methyl 39-46 serine/threonine kinase 11 Mus musculus 85-89 21799944-5 2010 CDDO-Me inhibited the growth of both K-ras mutated (MiaPaca2, Panc1 and Capan2) and wild-type K-ras (BxPC3) pancreatic cancer cells at very low concentrations. bardoxolone methyl 0-7 KRAS proto-oncogene, GTPase Homo sapiens 94-99 21799944-6 2010 The growth inhibitory activity of CDDO-Me was attributed to the induction of apoptosis characterized by increased annexin-V-FITC binding and cleavage of PARP-1 and procaspases-3, -8 and-9. bardoxolone methyl 34-41 annexin A5 Homo sapiens 114-123 21799944-6 2010 The growth inhibitory activity of CDDO-Me was attributed to the induction of apoptosis characterized by increased annexin-V-FITC binding and cleavage of PARP-1 and procaspases-3, -8 and-9. bardoxolone methyl 34-41 poly(ADP-ribose) polymerase 1 Homo sapiens 153-187 21799944-7 2010 In addition, CDDO-Me induced the loss of mitochondrial membrane potential and release of cytochrome C. bardoxolone methyl 13-20 cytochrome c, somatic Homo sapiens 89-101 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 AKT serine/threonine kinase 1 Homo sapiens 86-89 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 mechanistic target of rapamycin kinase Homo sapiens 105-134 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 mechanistic target of rapamycin kinase Homo sapiens 136-140 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 AKT serine/threonine kinase 1 Homo sapiens 191-194 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 mechanistic target of rapamycin kinase Homo sapiens 199-203 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 forkhead box O3 Homo sapiens 215-221 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 AKT serine/threonine kinase 1 Homo sapiens 191-194 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 eukaryotic translation initiation factor 4E Homo sapiens 242-248 21799944-8 2010 The antitumor activity of CDDO-Me was associated with the inhibition of prosurvival p-Akt, NF-kappaB and mammalian target of rapamycin (mTOR) signaling proteins and the downstream targets of Akt and mTOR, such as p-Foxo3a (Akt) and p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR). bardoxolone methyl 26-33 mechanistic target of rapamycin kinase Homo sapiens 199-203 21799944-9 2010 Silencing of Akt or mTOR with gene specific-siRNA sensitized the pancreatic cancer cells to CDDO-Me, demonstrating Akt and mTOR as molecular targets of CDDO-Me for its growth inhibitory and apoptosis-inducing activity. bardoxolone methyl 92-99 AKT serine/threonine kinase 1 Homo sapiens 13-16 21799944-9 2010 Silencing of Akt or mTOR with gene specific-siRNA sensitized the pancreatic cancer cells to CDDO-Me, demonstrating Akt and mTOR as molecular targets of CDDO-Me for its growth inhibitory and apoptosis-inducing activity. bardoxolone methyl 92-99 mechanistic target of rapamycin kinase Homo sapiens 20-24 21799944-9 2010 Silencing of Akt or mTOR with gene specific-siRNA sensitized the pancreatic cancer cells to CDDO-Me, demonstrating Akt and mTOR as molecular targets of CDDO-Me for its growth inhibitory and apoptosis-inducing activity. bardoxolone methyl 152-159 AKT serine/threonine kinase 1 Homo sapiens 13-16 21799944-9 2010 Silencing of Akt or mTOR with gene specific-siRNA sensitized the pancreatic cancer cells to CDDO-Me, demonstrating Akt and mTOR as molecular targets of CDDO-Me for its growth inhibitory and apoptosis-inducing activity. bardoxolone methyl 152-159 mechanistic target of rapamycin kinase Homo sapiens 20-24 21799944-9 2010 Silencing of Akt or mTOR with gene specific-siRNA sensitized the pancreatic cancer cells to CDDO-Me, demonstrating Akt and mTOR as molecular targets of CDDO-Me for its growth inhibitory and apoptosis-inducing activity. bardoxolone methyl 152-159 AKT serine/threonine kinase 1 Homo sapiens 115-118 21799944-9 2010 Silencing of Akt or mTOR with gene specific-siRNA sensitized the pancreatic cancer cells to CDDO-Me, demonstrating Akt and mTOR as molecular targets of CDDO-Me for its growth inhibitory and apoptosis-inducing activity. bardoxolone methyl 152-159 mechanistic target of rapamycin kinase Homo sapiens 123-127 20566646-4 2010 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO)-Im and CDDO-Me were observed to 1) inhibit the localization of Arp3 and actin at the leading edge of cells, 2) abrogate cell polarity, and 3) inhibit Arp2/3-dependent branched actin polymerization. bardoxolone methyl 60-67 actin related protein 3 Rattus norvegicus 116-120 20488920-3 2010 RNA interference studies indicate that these repressed genes are regulated by specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and Western blot analysis of lysates from pancreatic cancer cells treated with CDDO and CDDO-Me shows for the first time that both compounds decreased the expression of Sp1, Sp3, and Sp4. bardoxolone methyl 232-239 Sp3 transcription factor Homo sapiens 318-321 20230221-9 2010 Treatment with the anti-inflammatory agent CDDO-Me before and after MSC-IFN-beta injections resulted in reduction of MSC in the tumors and reversed the positive effect of tumor inhibition by MSC-IFN-beta alone (P=0.041). bardoxolone methyl 43-50 interferon beta 1 Homo sapiens 72-80 20230221-9 2010 Treatment with the anti-inflammatory agent CDDO-Me before and after MSC-IFN-beta injections resulted in reduction of MSC in the tumors and reversed the positive effect of tumor inhibition by MSC-IFN-beta alone (P=0.041). bardoxolone methyl 43-50 interferon beta 1 Homo sapiens 195-203 20386502-0 2010 Blockage of Stat3 with CDDO-Me inhibits tumor cell growth in chordoma. bardoxolone methyl 23-30 signal transducer and activator of transcription 3 Homo sapiens 12-17 20386502-2 2010 OBJECTIVE: To investigate the activation of Src/Stat3 pathway in chordomas cells and to determine the efficiency of inhibiting this pathway by 2-cyano-3,12-dioxooleana-1,9 (11)-dien-28-oic acid-methyl ester (CDDO-Me) as a potential chemotherapeutic agent for chordoma treatment. bardoxolone methyl 208-215 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 44-47 20386502-9 2010 The expression of key components of the Src/Stat3 signaling cascade and poly (ADP-ribose) polymerase cleavage in these CDDO-Me treated cells were analyzed by Western blot and immunofluorescence. bardoxolone methyl 119-126 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 40-43 20386502-13 2010 Expression of the key components of the Src/Stat3 signaling cascade was inhibited in chordoma cells after treatment with CDDO-Me. bardoxolone methyl 121-128 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 40-43 20386502-13 2010 Expression of the key components of the Src/Stat3 signaling cascade was inhibited in chordoma cells after treatment with CDDO-Me. bardoxolone methyl 121-128 signal transducer and activator of transcription 3 Homo sapiens 44-49 20386502-18 2010 Blockage of Src/Stat3 pathway by CDDO-Me is a potential strategy for chordoma treatment and may be focus for future research. bardoxolone methyl 33-40 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 12-15 20386502-18 2010 Blockage of Src/Stat3 pathway by CDDO-Me is a potential strategy for chordoma treatment and may be focus for future research. bardoxolone methyl 33-40 signal transducer and activator of transcription 3 Homo sapiens 16-21 20488920-2 2010 Treatment of Panc1, Panc28, and L3.6pL pancreatic cancer cells with low micromolar concentrations of CDDO or CDDO-Me resulted in growth inhibition, induction of apoptosis, and down-regulation of cyclin D1, survivin, vascular endothelial growth factor (VEGF), and its receptor (VEGFR2). bardoxolone methyl 109-116 cyclin D1 Homo sapiens 195-204 20488920-2 2010 Treatment of Panc1, Panc28, and L3.6pL pancreatic cancer cells with low micromolar concentrations of CDDO or CDDO-Me resulted in growth inhibition, induction of apoptosis, and down-regulation of cyclin D1, survivin, vascular endothelial growth factor (VEGF), and its receptor (VEGFR2). bardoxolone methyl 109-116 vascular endothelial growth factor A Homo sapiens 216-250 20488920-2 2010 Treatment of Panc1, Panc28, and L3.6pL pancreatic cancer cells with low micromolar concentrations of CDDO or CDDO-Me resulted in growth inhibition, induction of apoptosis, and down-regulation of cyclin D1, survivin, vascular endothelial growth factor (VEGF), and its receptor (VEGFR2). bardoxolone methyl 109-116 vascular endothelial growth factor A Homo sapiens 252-256 20488920-3 2010 RNA interference studies indicate that these repressed genes are regulated by specificity protein (Sp) transcription factors Sp1, Sp3, and Sp4, and Western blot analysis of lysates from pancreatic cancer cells treated with CDDO and CDDO-Me shows for the first time that both compounds decreased the expression of Sp1, Sp3, and Sp4. bardoxolone methyl 232-239 Sp4 transcription factor Homo sapiens 327-330 20488920-2 2010 Treatment of Panc1, Panc28, and L3.6pL pancreatic cancer cells with low micromolar concentrations of CDDO or CDDO-Me resulted in growth inhibition, induction of apoptosis, and down-regulation of cyclin D1, survivin, vascular endothelial growth factor (VEGF), and its receptor (VEGFR2). bardoxolone methyl 109-116 kinase insert domain receptor Homo sapiens 277-283 20488920-4 2010 Moreover, CDDO-Me (7.5 mg/kg/day) also inhibited pancreatic human L3.6pL tumor growth and down-regulated Sp1, Sp3, and Sp4 in tumors using an orthotopic pancreatic cancer model. bardoxolone methyl 10-17 Sp3 transcription factor Homo sapiens 110-113 20488920-4 2010 Moreover, CDDO-Me (7.5 mg/kg/day) also inhibited pancreatic human L3.6pL tumor growth and down-regulated Sp1, Sp3, and Sp4 in tumors using an orthotopic pancreatic cancer model. bardoxolone methyl 10-17 Sp4 transcription factor Homo sapiens 119-122 20488920-5 2010 CDDO-Me also induced reactive oxygen species (ROS) and decreased mitochondrial membrane potential (MMP) in Panc1 and L3.6pL cells, and cotreatment with antioxidants (glutathione and dithiothreitol) blocked the formation of ROS, reversed the loss of MMP, and inhibited down-regulation of Sp1, Sp3, and Sp4. bardoxolone methyl 0-7 Sp3 transcription factor Homo sapiens 292-295 20488920-5 2010 CDDO-Me also induced reactive oxygen species (ROS) and decreased mitochondrial membrane potential (MMP) in Panc1 and L3.6pL cells, and cotreatment with antioxidants (glutathione and dithiothreitol) blocked the formation of ROS, reversed the loss of MMP, and inhibited down-regulation of Sp1, Sp3, and Sp4. bardoxolone methyl 0-7 Sp4 transcription factor Homo sapiens 301-304 20488920-6 2010 Repression of Sp and Sp-dependent genes by CDDO-Me was due to the down-regulation of microRNA-27a and induction of zinc finger and BTB domain containing 10 (ZBTB10), an Sp repressor, and these responses were also reversed by antioxidants. bardoxolone methyl 43-50 zinc finger and BTB domain containing 10 Homo sapiens 157-163 20488920-7 2010 Thus, the anticancer activity of CDDO-Me is due, in part, to activation of ROS, which in turn targets the microRNA-27a:ZBTB10-Sp transcription factor axis. bardoxolone methyl 33-40 zinc finger and BTB domain containing 10 Homo sapiens 119-125 20063378-6 2010 Finally, we evaluated the effect of CDDO-Me on the inhibition of activated Stat3 pathway in osteosarcoma cell lines using MTT assay and Western blot analysis. bardoxolone methyl 36-43 signal transducer and activator of transcription 3 Homo sapiens 75-80 20571062-4 2010 Here, we report that CDDO-Me blocks targeting of NFkappaB to the nucleus by inhibiting IkappaB kinase beta-mediated phosphorylation of IkappaBalpha. bardoxolone methyl 21-28 nuclear factor kappa B subunit 1 Homo sapiens 49-57 20571062-4 2010 Here, we report that CDDO-Me blocks targeting of NFkappaB to the nucleus by inhibiting IkappaB kinase beta-mediated phosphorylation of IkappaBalpha. bardoxolone methyl 21-28 NFKB inhibitor alpha Homo sapiens 135-147 20571062-5 2010 Moreover, CDDO-Me blocked constitutive activation of the signal transducer and activator of transcription 3. bardoxolone methyl 10-17 signal transducer and activator of transcription 3 Homo sapiens 57-107 20571062-6 2010 We report the potent and selective antiproliferative effects of CDDO-Me on FLT3/ITD-positive myeloid leukemia cell lines and primary AML cells. bardoxolone methyl 64-71 fms related receptor tyrosine kinase 3 Homo sapiens 75-79 20571062-7 2010 The present studies show that CDDO-Me treatment results in caspase-3-mediated induction of apoptosis of FLT3/ITD-expressing cells and its antiproliferative effects are synergistic with PKC412, a FLT3-tyrosine kinase inhibitor currently in clinical trials. bardoxolone methyl 30-37 caspase 3 Homo sapiens 59-68 20571062-7 2010 The present studies show that CDDO-Me treatment results in caspase-3-mediated induction of apoptosis of FLT3/ITD-expressing cells and its antiproliferative effects are synergistic with PKC412, a FLT3-tyrosine kinase inhibitor currently in clinical trials. bardoxolone methyl 30-37 fms related receptor tyrosine kinase 3 Homo sapiens 104-108 20571062-7 2010 The present studies show that CDDO-Me treatment results in caspase-3-mediated induction of apoptosis of FLT3/ITD-expressing cells and its antiproliferative effects are synergistic with PKC412, a FLT3-tyrosine kinase inhibitor currently in clinical trials. bardoxolone methyl 30-37 fms related receptor tyrosine kinase 3 Homo sapiens 195-199 20571062-8 2010 Taken together, our studies indicate that CDDO-Me greatly enhanced the efficacy of the FLT3 inhibitor PKC412, suggesting that combining two separate pathway inhibitors might be a viable therapeutic strategy for AML associated with a FLT3/ITD mutation. bardoxolone methyl 42-49 fms related receptor tyrosine kinase 3 Homo sapiens 87-91 20571062-8 2010 Taken together, our studies indicate that CDDO-Me greatly enhanced the efficacy of the FLT3 inhibitor PKC412, suggesting that combining two separate pathway inhibitors might be a viable therapeutic strategy for AML associated with a FLT3/ITD mutation. bardoxolone methyl 42-49 fms related receptor tyrosine kinase 3 Homo sapiens 233-237 20459702-0 2010 Oleanane triterpenoid CDDO-Me induces apoptosis in multidrug resistant osteosarcoma cells through inhibition of Stat3 pathway. bardoxolone methyl 22-29 signal transducer and activator of transcription 3 Homo sapiens 112-117 20626291-7 2010 In contrast, CDDO-Me protection against LPS challenge had no impact on absolute numbers or distribution of splenocyte subsets, despite attenuating in vivo induction of proinflammatory cytokines in an IL-10-independent manner. bardoxolone methyl 13-20 interleukin 10 Mus musculus 200-205 20459702-5 2010 The effect of CDDO-Me on osteosarcoma cell growth was evaluated by MTT and apoptosis by PARP cleavage assay and caspase-3/7 activity. bardoxolone methyl 14-21 poly(ADP-ribose) polymerase 1 Homo sapiens 88-92 20459702-5 2010 The effect of CDDO-Me on osteosarcoma cell growth was evaluated by MTT and apoptosis by PARP cleavage assay and caspase-3/7 activity. bardoxolone methyl 14-21 caspase 3 Homo sapiens 112-121 20459702-8 2010 Treatment with CDDO-Me significantly decreased the level of nuclear translocation and phosphorylation of Stat3. bardoxolone methyl 15-22 signal transducer and activator of transcription 3 Homo sapiens 105-110 20459702-12 2010 CDDO-Me significantly inhibited Stat3 phosphorylation, Stat3 nuclear translocation and induced apoptosis in osteosarcoma. bardoxolone methyl 0-7 signal transducer and activator of transcription 3 Homo sapiens 32-37 20459702-12 2010 CDDO-Me significantly inhibited Stat3 phosphorylation, Stat3 nuclear translocation and induced apoptosis in osteosarcoma. bardoxolone methyl 0-7 signal transducer and activator of transcription 3 Homo sapiens 55-60 18757413-3 2008 CDDO-Me induced caspase-3, caspase-8, and caspase-9 activation; poly(ADP-ribose) polymerase cleavage; internucleosomal DNA fragmentation; and loss of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction in PC3 and DU145 cells. bardoxolone methyl 0-7 caspase 3 Homo sapiens 16-25 19782051-4 2010 Here, we show that CDDO-Me induces ROS generation from both nonmitochondrial and mitochondrial sources, which is associated with the induction of apoptosis as characterized by increased annexin V-binding, cleavage of PARP-1 and procaspases-3, -8, -9, loss of mitochondrial membrane potential and release of cytochrome c. bardoxolone methyl 19-26 annexin A5 Homo sapiens 186-195 19782051-4 2010 Here, we show that CDDO-Me induces ROS generation from both nonmitochondrial and mitochondrial sources, which is associated with the induction of apoptosis as characterized by increased annexin V-binding, cleavage of PARP-1 and procaspases-3, -8, -9, loss of mitochondrial membrane potential and release of cytochrome c. bardoxolone methyl 19-26 poly(ADP-ribose) polymerase 1 Homo sapiens 217-249 19782051-4 2010 Here, we show that CDDO-Me induces ROS generation from both nonmitochondrial and mitochondrial sources, which is associated with the induction of apoptosis as characterized by increased annexin V-binding, cleavage of PARP-1 and procaspases-3, -8, -9, loss of mitochondrial membrane potential and release of cytochrome c. bardoxolone methyl 19-26 cytochrome c, somatic Homo sapiens 307-319 19782051-5 2010 In addition, CDDO-Me inhibited cell survival Akt, NF-kappaB and mTOR signaling proteins. bardoxolone methyl 13-20 AKT serine/threonine kinase 1 Homo sapiens 45-48 19782051-5 2010 In addition, CDDO-Me inhibited cell survival Akt, NF-kappaB and mTOR signaling proteins. bardoxolone methyl 13-20 mechanistic target of rapamycin kinase Homo sapiens 64-68 19782051-6 2010 The inhibition of ROS generation by N-acetylcysteine (NAC) or by overexpression of antioxidant enzymes glutathione peroxidase (GPx) and superoxide dismutase-1 (SOD-1) prevented CDDO-Me-induced apoptosis. bardoxolone methyl 177-184 superoxide dismutase 1 Homo sapiens 136-158 19782051-6 2010 The inhibition of ROS generation by N-acetylcysteine (NAC) or by overexpression of antioxidant enzymes glutathione peroxidase (GPx) and superoxide dismutase-1 (SOD-1) prevented CDDO-Me-induced apoptosis. bardoxolone methyl 177-184 superoxide dismutase 1 Homo sapiens 160-165 19782051-7 2010 Pretreatment with NAC blocked annexin V-binding, cleavage of PARP-1 and procaspases-3, -8, -9, loss of mitochondrial membrane potential and release of cytochrome c by CDDO-Me. bardoxolone methyl 167-174 cytochrome c, somatic Homo sapiens 151-163 19782051-8 2010 NAC also prevented the inhibition of constitutively active Akt, NF-kappaB and mTOR by CDDO-Me. bardoxolone methyl 86-93 AKT serine/threonine kinase 1 Homo sapiens 59-62 18587580-0 2009 CDDO-Me, a synthetic triterpenoid, inhibits expression of IL-6 and Stat3 phosphorylation in multi-drug resistant ovarian cancer cells. bardoxolone methyl 0-7 interleukin 6 Homo sapiens 58-62 18587580-0 2009 CDDO-Me, a synthetic triterpenoid, inhibits expression of IL-6 and Stat3 phosphorylation in multi-drug resistant ovarian cancer cells. bardoxolone methyl 0-7 signal transducer and activator of transcription 3 Homo sapiens 67-72 18587580-3 2009 To explore potential therapeutic strategies for interrupting signaling through this pathway, we assessed the ability of CDDO-Me, a synthetic triterpenoid, to inhibit IL-6 secretion, Stat3 phosphorylation, Stat3 nuclear translocation and paclitaxel sensitivity in several cell line model systems. bardoxolone methyl 120-127 interleukin 6 Homo sapiens 166-170 18587580-3 2009 To explore potential therapeutic strategies for interrupting signaling through this pathway, we assessed the ability of CDDO-Me, a synthetic triterpenoid, to inhibit IL-6 secretion, Stat3 phosphorylation, Stat3 nuclear translocation and paclitaxel sensitivity in several cell line model systems. bardoxolone methyl 120-127 signal transducer and activator of transcription 3 Homo sapiens 182-187 18587580-4 2009 These studies demonstrated that CDDO-Me significantly inhibits IL-6 secretion in paclitaxel-resistant ovarian cancer cells and specifically suppresses IL-6- or oncostatin M-induced Stat3 nuclear translocation. bardoxolone methyl 32-39 interleukin 6 Homo sapiens 63-67 18587580-4 2009 These studies demonstrated that CDDO-Me significantly inhibits IL-6 secretion in paclitaxel-resistant ovarian cancer cells and specifically suppresses IL-6- or oncostatin M-induced Stat3 nuclear translocation. bardoxolone methyl 32-39 interleukin 6 Homo sapiens 151-156 18587580-4 2009 These studies demonstrated that CDDO-Me significantly inhibits IL-6 secretion in paclitaxel-resistant ovarian cancer cells and specifically suppresses IL-6- or oncostatin M-induced Stat3 nuclear translocation. bardoxolone methyl 32-39 signal transducer and activator of transcription 3 Homo sapiens 181-186 18587580-5 2009 Treatment with CDDO-Me significantly decreases the levels of Stat3, Jak2, and Src phosphorylation in ovarian and breast cancer cell lines with constitutively activated Stat3. bardoxolone methyl 15-22 signal transducer and activator of transcription 3 Homo sapiens 61-66 18587580-5 2009 Treatment with CDDO-Me significantly decreases the levels of Stat3, Jak2, and Src phosphorylation in ovarian and breast cancer cell lines with constitutively activated Stat3. bardoxolone methyl 15-22 Janus kinase 2 Homo sapiens 68-72 18587580-5 2009 Treatment with CDDO-Me significantly decreases the levels of Stat3, Jak2, and Src phosphorylation in ovarian and breast cancer cell lines with constitutively activated Stat3. bardoxolone methyl 15-22 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 78-81 18587580-5 2009 Treatment with CDDO-Me significantly decreases the levels of Stat3, Jak2, and Src phosphorylation in ovarian and breast cancer cell lines with constitutively activated Stat3. bardoxolone methyl 15-22 signal transducer and activator of transcription 3 Homo sapiens 168-173 18587580-8 2009 Our data confirm that CDDO-Me interrupts the signaling of multiple kinases involved in the IL-6-Stat3 and Src signaling pathways. bardoxolone methyl 22-29 interleukin 6 Homo sapiens 91-101 18587580-8 2009 Our data confirm that CDDO-Me interrupts the signaling of multiple kinases involved in the IL-6-Stat3 and Src signaling pathways. bardoxolone methyl 22-29 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 106-109 18794136-0 2008 Coupling of endoplasmic reticulum stress to CDDO-Me-induced up-regulation of death receptor 5 via a CHOP-dependent mechanism involving JNK activation. bardoxolone methyl 44-51 TNF receptor superfamily member 10b Homo sapiens 77-93 18794136-0 2008 Coupling of endoplasmic reticulum stress to CDDO-Me-induced up-regulation of death receptor 5 via a CHOP-dependent mechanism involving JNK activation. bardoxolone methyl 44-51 DNA damage inducible transcript 3 Homo sapiens 100-104 18794136-0 2008 Coupling of endoplasmic reticulum stress to CDDO-Me-induced up-regulation of death receptor 5 via a CHOP-dependent mechanism involving JNK activation. bardoxolone methyl 44-51 mitogen-activated protein kinase 8 Homo sapiens 135-138 18794136-2 2008 We previously showed that CDDO-Me induces c-Jun NH(2)-terminal kinase (JNK)-dependent death receptor 5 (DR5) expression and augments death receptor-induced apoptosis. bardoxolone methyl 26-33 mitogen-activated protein kinase 8 Homo sapiens 71-74 18794136-2 2008 We previously showed that CDDO-Me induces c-Jun NH(2)-terminal kinase (JNK)-dependent death receptor 5 (DR5) expression and augments death receptor-induced apoptosis. bardoxolone methyl 26-33 TNF receptor superfamily member 10b Homo sapiens 86-102 18794136-2 2008 We previously showed that CDDO-Me induces c-Jun NH(2)-terminal kinase (JNK)-dependent death receptor 5 (DR5) expression and augments death receptor-induced apoptosis. bardoxolone methyl 26-33 TNF receptor superfamily member 10b Homo sapiens 104-107 18794136-3 2008 The current study focused on addressing how CDDO-Me induces JNK-dependent DR5 expression. bardoxolone methyl 44-51 mitogen-activated protein kinase 8 Homo sapiens 60-63 18794136-3 2008 The current study focused on addressing how CDDO-Me induces JNK-dependent DR5 expression. bardoxolone methyl 44-51 TNF receptor superfamily member 10b Homo sapiens 74-77 18794136-4 2008 Analysis of DR5 promoter regions defines that the CCAAT/enhancer binding protein homologous protein (CHOP) binding site is responsible for CDDO-Me-induced transactivation of the DR5 gene. bardoxolone methyl 139-146 TNF receptor superfamily member 10b Homo sapiens 12-15 18794136-4 2008 Analysis of DR5 promoter regions defines that the CCAAT/enhancer binding protein homologous protein (CHOP) binding site is responsible for CDDO-Me-induced transactivation of the DR5 gene. bardoxolone methyl 139-146 DNA damage inducible transcript 3 Homo sapiens 50-99 18794136-4 2008 Analysis of DR5 promoter regions defines that the CCAAT/enhancer binding protein homologous protein (CHOP) binding site is responsible for CDDO-Me-induced transactivation of the DR5 gene. bardoxolone methyl 139-146 DNA damage inducible transcript 3 Homo sapiens 101-105 18794136-4 2008 Analysis of DR5 promoter regions defines that the CCAAT/enhancer binding protein homologous protein (CHOP) binding site is responsible for CDDO-Me-induced transactivation of the DR5 gene. bardoxolone methyl 139-146 TNF receptor superfamily member 10b Homo sapiens 178-181 18794136-5 2008 Consistently, CDDO-Me induced DR5 expression and parallel CHOP up-regulation. bardoxolone methyl 14-21 TNF receptor superfamily member 10b Homo sapiens 30-33 18794136-5 2008 Consistently, CDDO-Me induced DR5 expression and parallel CHOP up-regulation. bardoxolone methyl 14-21 DNA damage inducible transcript 3 Homo sapiens 58-62 18794136-6 2008 Blockade of CHOP up-regulation also abrogated CDDO-Me-induced DR5 expression. bardoxolone methyl 46-53 DNA damage inducible transcript 3 Homo sapiens 12-16 18794136-6 2008 Blockade of CHOP up-regulation also abrogated CDDO-Me-induced DR5 expression. bardoxolone methyl 46-53 TNF receptor superfamily member 10b Homo sapiens 62-65 18794136-7 2008 These results indicate that CDDO-Me induces CHOP-dependent DR5 up-regulation. bardoxolone methyl 28-35 DNA damage inducible transcript 3 Homo sapiens 44-48 18794136-7 2008 These results indicate that CDDO-Me induces CHOP-dependent DR5 up-regulation. bardoxolone methyl 28-35 TNF receptor superfamily member 10b Homo sapiens 59-62 20372807-5 2010 CDDO-Me, a synthetic triterpenoid, suppressed expression of Spot 14 and FASN genes and likewise inhibited LS cell growth. bardoxolone methyl 0-7 thyroid hormone responsive Homo sapiens 60-67 20372807-5 2010 CDDO-Me, a synthetic triterpenoid, suppressed expression of Spot 14 and FASN genes and likewise inhibited LS cell growth. bardoxolone methyl 0-7 fatty acid synthase Homo sapiens 72-76 20392997-7 2010 Induction of apoptosis by CDDO-Me was associated with the inhibition of pro-survival Akt, NF-kappaB and mTOR signaling proteins and NF-kappaB-regulated anti-apoptotic Bcl-2, Bcl-xL, Bad and survivin. bardoxolone methyl 26-33 AKT serine/threonine kinase 1 Homo sapiens 85-88 20392997-7 2010 Induction of apoptosis by CDDO-Me was associated with the inhibition of pro-survival Akt, NF-kappaB and mTOR signaling proteins and NF-kappaB-regulated anti-apoptotic Bcl-2, Bcl-xL, Bad and survivin. bardoxolone methyl 26-33 mechanistic target of rapamycin kinase Homo sapiens 104-108 20392997-7 2010 Induction of apoptosis by CDDO-Me was associated with the inhibition of pro-survival Akt, NF-kappaB and mTOR signaling proteins and NF-kappaB-regulated anti-apoptotic Bcl-2, Bcl-xL, Bad and survivin. bardoxolone methyl 26-33 BCL2 apoptosis regulator Homo sapiens 167-172 20392997-7 2010 Induction of apoptosis by CDDO-Me was associated with the inhibition of pro-survival Akt, NF-kappaB and mTOR signaling proteins and NF-kappaB-regulated anti-apoptotic Bcl-2, Bcl-xL, Bad and survivin. bardoxolone methyl 26-33 BCL2 like 1 Homo sapiens 174-180 19782051-8 2010 NAC also prevented the inhibition of constitutively active Akt, NF-kappaB and mTOR by CDDO-Me. bardoxolone methyl 86-93 mechanistic target of rapamycin kinase Homo sapiens 78-82 19189297-0 2009 Oleanane triterpenoid CDDO-Me inhibits growth and induces apoptosis in prostate cancer cells by independently targeting pro-survival Akt and mTOR. bardoxolone methyl 22-29 AKT serine/threonine kinase 1 Homo sapiens 133-136 19189297-0 2009 Oleanane triterpenoid CDDO-Me inhibits growth and induces apoptosis in prostate cancer cells by independently targeting pro-survival Akt and mTOR. bardoxolone methyl 22-29 mechanistic target of rapamycin kinase Homo sapiens 141-145 19189297-3 2009 METHODS: The antitumor activity of CDDO-Me for hormone-refractory PC-3 (AR(-)) and C4-2 (AR(+)) prostate cancer cell lines was determined by effects on cell growth and induction of apoptosis, identification of molecular targets, and therapeutic efficacy in vivo in PC-3 xenograft model. bardoxolone methyl 35-42 CDK5 regulatory subunit associated protein 1 Homo sapiens 83-87 19189297-4 2009 RESULTS: CDDO-Me inhibited the growth and induced apoptosis in PC-3 and C4-2 cells at extremely low concentrations. bardoxolone methyl 9-16 CDK5 regulatory subunit associated protein 1 Homo sapiens 72-76 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 AKT serine/threonine kinase 1 Homo sapiens 74-77 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 mechanistic target of rapamycin kinase Homo sapiens 79-108 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 mechanistic target of rapamycin kinase Homo sapiens 110-114 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 nuclear factor kappa B subunit 1 Homo sapiens 121-143 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 nuclear factor kappa B subunit 1 Homo sapiens 145-154 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 AKT serine/threonine kinase 1 Homo sapiens 232-235 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 eukaryotic translation initiation factor 4E Homo sapiens 248-254 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 mechanistic target of rapamycin kinase Homo sapiens 269-273 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 mitochondrially encoded cytochrome c oxidase II Homo sapiens 280-285 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 vascular endothelial growth factor A Homo sapiens 287-291 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 cyclin D1 Homo sapiens 296-305 19189297-5 2009 The antitumor activity of CDDO-Me was associated with the inhibition of p-Akt, mammalian target of rapamycin (mTOR), and nuclear factor kappa B (NF-kappaB) signaling proteins and their downstream targets such as p-Bad and p-Foxo3a (Akt); p-S6K1, p-eIF-4E and p-4E-BP1 (mTOR); and COX-2, VEGF and cyclin D1(NF-kappaB). bardoxolone methyl 26-33 nuclear factor kappa B subunit 1 Homo sapiens 306-315 19189297-6 2009 Silencing of Akt sensitized the PC-3 cells to CDDO-Me, whereas overexpression of Akt induced resistance to CDDO-Me. bardoxolone methyl 46-53 AKT serine/threonine kinase 1 Homo sapiens 13-16 19189297-6 2009 Silencing of Akt sensitized the PC-3 cells to CDDO-Me, whereas overexpression of Akt induced resistance to CDDO-Me. bardoxolone methyl 107-114 AKT serine/threonine kinase 1 Homo sapiens 81-84 19189297-7 2009 Targeted silencing of Akt showed that Akt does not regulate mTOR activation in PC-3 cells, but targeted silencing of mTOR sensitized PC-3 cells to CDDO-Me mediated growth inhibition. bardoxolone methyl 147-154 mechanistic target of rapamycin kinase Homo sapiens 117-121 19189297-10 2009 Data also identified Akt and mTOR as molecular targets of CDDO-Me in prostate cancer cells. bardoxolone methyl 58-65 AKT serine/threonine kinase 1 Homo sapiens 21-24 19189297-10 2009 Data also identified Akt and mTOR as molecular targets of CDDO-Me in prostate cancer cells. bardoxolone methyl 58-65 mechanistic target of rapamycin kinase Homo sapiens 29-33 18794136-8 2008 Moreover, the JNK inhibitor SP600125 abrogated CHOP induction by CDDO-Me, suggesting a JNK-dependent CHOP up-regulation by CDDO-Me as well. bardoxolone methyl 65-72 mitogen-activated protein kinase 8 Homo sapiens 14-17 18794136-8 2008 Moreover, the JNK inhibitor SP600125 abrogated CHOP induction by CDDO-Me, suggesting a JNK-dependent CHOP up-regulation by CDDO-Me as well. bardoxolone methyl 65-72 DNA damage inducible transcript 3 Homo sapiens 47-51 18794136-8 2008 Moreover, the JNK inhibitor SP600125 abrogated CHOP induction by CDDO-Me, suggesting a JNK-dependent CHOP up-regulation by CDDO-Me as well. bardoxolone methyl 65-72 mitogen-activated protein kinase 8 Homo sapiens 87-90 18794136-8 2008 Moreover, the JNK inhibitor SP600125 abrogated CHOP induction by CDDO-Me, suggesting a JNK-dependent CHOP up-regulation by CDDO-Me as well. bardoxolone methyl 65-72 DNA damage inducible transcript 3 Homo sapiens 101-105 18794136-8 2008 Moreover, the JNK inhibitor SP600125 abrogated CHOP induction by CDDO-Me, suggesting a JNK-dependent CHOP up-regulation by CDDO-Me as well. bardoxolone methyl 123-130 mitogen-activated protein kinase 8 Homo sapiens 14-17 18794136-8 2008 Moreover, the JNK inhibitor SP600125 abrogated CHOP induction by CDDO-Me, suggesting a JNK-dependent CHOP up-regulation by CDDO-Me as well. bardoxolone methyl 123-130 DNA damage inducible transcript 3 Homo sapiens 47-51 18794136-8 2008 Moreover, the JNK inhibitor SP600125 abrogated CHOP induction by CDDO-Me, suggesting a JNK-dependent CHOP up-regulation by CDDO-Me as well. bardoxolone methyl 123-130 mitogen-activated protein kinase 8 Homo sapiens 87-90 18794136-8 2008 Moreover, the JNK inhibitor SP600125 abrogated CHOP induction by CDDO-Me, suggesting a JNK-dependent CHOP up-regulation by CDDO-Me as well. bardoxolone methyl 123-130 DNA damage inducible transcript 3 Homo sapiens 101-105 18794136-9 2008 Importantly, knockdown of CHOP attenuated CDDO-Me-induced apoptosis, showing that CHOP induction is involved in CDDO-Me-induced apoptosis. bardoxolone methyl 42-49 DNA damage inducible transcript 3 Homo sapiens 26-30 18794136-9 2008 Importantly, knockdown of CHOP attenuated CDDO-Me-induced apoptosis, showing that CHOP induction is involved in CDDO-Me-induced apoptosis. bardoxolone methyl 42-49 DNA damage inducible transcript 3 Homo sapiens 82-86 18794136-9 2008 Importantly, knockdown of CHOP attenuated CDDO-Me-induced apoptosis, showing that CHOP induction is involved in CDDO-Me-induced apoptosis. bardoxolone methyl 112-119 DNA damage inducible transcript 3 Homo sapiens 26-30 18794136-9 2008 Importantly, knockdown of CHOP attenuated CDDO-Me-induced apoptosis, showing that CHOP induction is involved in CDDO-Me-induced apoptosis. bardoxolone methyl 112-119 DNA damage inducible transcript 3 Homo sapiens 82-86 18794136-10 2008 Additionally, CDDO-Me increased the levels of Bip, phosphorylated eukaryotic translation initiation factor 2alpha, inositol requiring kinase 1alpha, and activating transcription factor 4, all of which are featured changes during endoplasmic reticulum (ER) stress. bardoxolone methyl 14-21 growth differentiation factor 10 Homo sapiens 46-49 18794136-10 2008 Additionally, CDDO-Me increased the levels of Bip, phosphorylated eukaryotic translation initiation factor 2alpha, inositol requiring kinase 1alpha, and activating transcription factor 4, all of which are featured changes during endoplasmic reticulum (ER) stress. bardoxolone methyl 14-21 activating transcription factor 4 Homo sapiens 107-186 18794136-11 2008 Furthermore, salubrinal, an inhibitor of ER stress-induced apoptosis, inhibited JNK activation and up-regulation of CHOP and DR5 by CDDO-Me and protected cells from CDDO-Me-induced apoptosis. bardoxolone methyl 132-139 DNA damage inducible transcript 3 Homo sapiens 116-120 18794136-11 2008 Furthermore, salubrinal, an inhibitor of ER stress-induced apoptosis, inhibited JNK activation and up-regulation of CHOP and DR5 by CDDO-Me and protected cells from CDDO-Me-induced apoptosis. bardoxolone methyl 132-139 TNF receptor superfamily member 10b Homo sapiens 125-128 18794136-12 2008 Thus, ER stress seems to be important for CDDO-Me-induced JNK activation, CHOP and DR5 up-regulation, and apoptosis. bardoxolone methyl 42-49 mitogen-activated protein kinase 8 Homo sapiens 58-61 18794136-12 2008 Thus, ER stress seems to be important for CDDO-Me-induced JNK activation, CHOP and DR5 up-regulation, and apoptosis. bardoxolone methyl 42-49 DNA damage inducible transcript 3 Homo sapiens 74-78 18794136-12 2008 Thus, ER stress seems to be important for CDDO-Me-induced JNK activation, CHOP and DR5 up-regulation, and apoptosis. bardoxolone methyl 42-49 TNF receptor superfamily member 10b Homo sapiens 83-86 18794136-13 2008 Collectively, we conclude that CDDO-Me triggers ER stress, leading to JNK-dependent, CHOP-mediated DR5 up-regulation and apoptosis. bardoxolone methyl 31-38 mitogen-activated protein kinase 8 Homo sapiens 70-73 18794136-13 2008 Collectively, we conclude that CDDO-Me triggers ER stress, leading to JNK-dependent, CHOP-mediated DR5 up-regulation and apoptosis. bardoxolone methyl 31-38 DNA damage inducible transcript 3 Homo sapiens 85-89 18794136-13 2008 Collectively, we conclude that CDDO-Me triggers ER stress, leading to JNK-dependent, CHOP-mediated DR5 up-regulation and apoptosis. bardoxolone methyl 31-38 TNF receptor superfamily member 10b Homo sapiens 99-102 18757413-3 2008 CDDO-Me induced caspase-3, caspase-8, and caspase-9 activation; poly(ADP-ribose) polymerase cleavage; internucleosomal DNA fragmentation; and loss of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction in PC3 and DU145 cells. bardoxolone methyl 0-7 caspase 8 Homo sapiens 27-36 18757413-3 2008 CDDO-Me induced caspase-3, caspase-8, and caspase-9 activation; poly(ADP-ribose) polymerase cleavage; internucleosomal DNA fragmentation; and loss of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction in PC3 and DU145 cells. bardoxolone methyl 0-7 caspase 9 Homo sapiens 42-51 18757413-3 2008 CDDO-Me induced caspase-3, caspase-8, and caspase-9 activation; poly(ADP-ribose) polymerase cleavage; internucleosomal DNA fragmentation; and loss of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide reduction in PC3 and DU145 cells. bardoxolone methyl 0-7 chromobox 8 Homo sapiens 224-227 18757413-5 2008 Interestingly, CDDO-Me induced inactivating phosphorylation at Ser(9) of glycogen synthase kinase 3beta (GSK3beta), a multifunctional kinase that mediates essential events promoting prostate cancer development and acquisition of androgen independence. bardoxolone methyl 15-22 glycogen synthase kinase 3 beta Homo sapiens 73-103 18757413-5 2008 Interestingly, CDDO-Me induced inactivating phosphorylation at Ser(9) of glycogen synthase kinase 3beta (GSK3beta), a multifunctional kinase that mediates essential events promoting prostate cancer development and acquisition of androgen independence. bardoxolone methyl 15-22 glycogen synthase kinase 3 beta Homo sapiens 105-113 18757413-6 2008 The GSK3 inhibitor lithium chloride and, more effectively, GSK3 gene silencing sensitized PC3 and DU145 prostate cancer cells to CDDO-Me cytotoxicity. bardoxolone methyl 129-136 chromobox 8 Homo sapiens 90-93 18757413-7 2008 These data suggest that modulation of GSK3beta activation is involved in the cell death pathway engaged by CDDO-Me in prostate cancer cells. bardoxolone methyl 107-114 glycogen synthase kinase 3 beta Homo sapiens 38-46 18474101-0 2008 The synthetic triterpenoid CDDO-methyl ester modulates microglial activities, inhibits TNF production, and provides dopaminergic neuroprotection. bardoxolone methyl 27-44 tumor necrosis factor Mus musculus 87-90 18628471-0 2008 Prevention and treatment of experimental estrogen receptor-negative mammary carcinogenesis by the synthetic triterpenoid CDDO-methyl Ester and the rexinoid LG100268. bardoxolone methyl 121-138 estrogen receptor 1 (alpha) Mus musculus 41-58 18628471-4 2008 RESULTS: CDDO-Me and 268 significantly delayed the development of ER-negative tumors, with a 14- and 24-week delay, respectively, compared with the control group for the time required to reach 50% tumor incidence. bardoxolone methyl 9-16 estrogen receptor 1 (alpha) Mus musculus 66-68 18628471-8 2008 CDDO-Me inhibited constitutive STAT3 phosphorylation and the degradation of IKBalpha in ER-negative breast cancer cells, whereas 268 blocked IKBalpha degradation and the release of interleukin-6 in RAW264.7 macrophage-like cells, inhibited the ability of endothelial cells to organize into networks, and blocked angiogenesis in vivo. bardoxolone methyl 0-7 signal transducer and activator of transcription 3 Mus musculus 31-36 18628471-8 2008 CDDO-Me inhibited constitutive STAT3 phosphorylation and the degradation of IKBalpha in ER-negative breast cancer cells, whereas 268 blocked IKBalpha degradation and the release of interleukin-6 in RAW264.7 macrophage-like cells, inhibited the ability of endothelial cells to organize into networks, and blocked angiogenesis in vivo. bardoxolone methyl 0-7 nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor, alpha Mus musculus 76-84 18474101-8 2008 RESULTS: We found that at low nanomolar concentrations, treatment of rat primary mesencephalon neuron/glia cultures with CDDO-Me resulted in attenuated LPS-, TNF- or fibrillar amyloid beta 1-42 (A beta 1-42) peptide-induced increases in reactive microglia and inflammatory gene expression without an overall effect on cell viability. bardoxolone methyl 121-128 tumor necrosis factor Mus musculus 158-161 18474101-9 2008 In functional assays CDDO-Me blocked death in the dopaminergic neuron-like cell line MN9D induced by conditioned media (CM) of LPS-stimulated BV2 microglia, but did not block cell death induced by addition of TNF to MN9D cells, suggesting that dopaminergic neuroprotection by CDDO-Me involved inhibition of microglial-derived cytokine production and not direct inhibition of TNF-dependent pro-apoptotic pathways. bardoxolone methyl 21-28 tumor necrosis factor Mus musculus 375-378 18474101-10 2008 Multiplexed immunoassays of CM from LPS-stimulated microglia confirmed that CDDO-Me-treated BV2 cells produced decreased levels of specific subsets of cytokines, in particular TNF. bardoxolone methyl 76-83 tumor necrosis factor Mus musculus 176-179 18253090-0 2007 c-FLIP downregulation contributes to apoptosis induction by the novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me) in human lung cancer cells. bardoxolone methyl 145-152 CASP8 and FADD like apoptosis regulator Homo sapiens 0-6 18483301-4 2008 In addition, CDDO-Me rapidly decreased the viability of murine lymphoid Ba/F3 cells expressing wild-type p210 as well as the imatinib-resistant E255K and T315I mutations of bcr-abl. bardoxolone methyl 13-20 envoplakin Mus musculus 105-109 18413761-0 2008 Triterpenoid CDDO-methyl ester inhibits the Janus-activated kinase-1 (JAK1)-->signal transducer and activator of transcription-3 (STAT3) pathway by direct inhibition of JAK1 and STAT3. bardoxolone methyl 13-30 signal transducer and activator of transcription 3 Homo sapiens 181-186 18413761-3 2008 The present studies show that CDDO-Me blocks interleukin-6 (IL-6)-induced and constitutive activation of the Janus-activated kinase 1 (JAK1) in cells. bardoxolone methyl 30-37 interleukin 6 Homo sapiens 45-58 18413761-3 2008 The present studies show that CDDO-Me blocks interleukin-6 (IL-6)-induced and constitutive activation of the Janus-activated kinase 1 (JAK1) in cells. bardoxolone methyl 30-37 interleukin 6 Homo sapiens 60-64 18413761-3 2008 The present studies show that CDDO-Me blocks interleukin-6 (IL-6)-induced and constitutive activation of the Janus-activated kinase 1 (JAK1) in cells. bardoxolone methyl 30-37 Janus kinase 1 Homo sapiens 109-133 18413761-3 2008 The present studies show that CDDO-Me blocks interleukin-6 (IL-6)-induced and constitutive activation of the Janus-activated kinase 1 (JAK1) in cells. bardoxolone methyl 30-37 Janus kinase 1 Homo sapiens 135-139 18413761-4 2008 In support of a direct mechanism, CDDO-Me forms adducts with JAK1 at Cys(1077) in the kinase domain and inhibits JAK1 activity. bardoxolone methyl 34-41 Janus kinase 1 Homo sapiens 61-65 18413761-4 2008 In support of a direct mechanism, CDDO-Me forms adducts with JAK1 at Cys(1077) in the kinase domain and inhibits JAK1 activity. bardoxolone methyl 34-41 Janus kinase 1 Homo sapiens 113-117 18413761-5 2008 In concert with these results, CDDO-Me blocked IL-6-induced and constitutive activation of signal transducer and activator of transcription 3 (STAT3). bardoxolone methyl 31-38 interleukin 6 Homo sapiens 47-51 18413761-5 2008 In concert with these results, CDDO-Me blocked IL-6-induced and constitutive activation of signal transducer and activator of transcription 3 (STAT3). bardoxolone methyl 31-38 signal transducer and activator of transcription 3 Homo sapiens 91-141 18413761-5 2008 In concert with these results, CDDO-Me blocked IL-6-induced and constitutive activation of signal transducer and activator of transcription 3 (STAT3). bardoxolone methyl 31-38 signal transducer and activator of transcription 3 Homo sapiens 143-148 18413761-6 2008 Moreover, we show that CDDO-Me (a) binds directly to STAT3 by a mechanism dependent on the alkylation of Cys(259) and (b) inhibits the formation of STAT3 dimers. bardoxolone methyl 23-30 signal transducer and activator of transcription 3 Homo sapiens 53-58 18413761-6 2008 Moreover, we show that CDDO-Me (a) binds directly to STAT3 by a mechanism dependent on the alkylation of Cys(259) and (b) inhibits the formation of STAT3 dimers. bardoxolone methyl 23-30 signal transducer and activator of transcription 3 Homo sapiens 148-153 18413761-7 2008 These findings indicate that CDDO-Me inhibits activation of the JAK1-->STAT3 pathway by forming adducts with both JAK1 and STAT3. bardoxolone methyl 29-36 Janus kinase 1 Homo sapiens 64-68 18413761-7 2008 These findings indicate that CDDO-Me inhibits activation of the JAK1-->STAT3 pathway by forming adducts with both JAK1 and STAT3. bardoxolone methyl 29-36 signal transducer and activator of transcription 3 Homo sapiens 74-79 18413761-7 2008 These findings indicate that CDDO-Me inhibits activation of the JAK1-->STAT3 pathway by forming adducts with both JAK1 and STAT3. bardoxolone methyl 29-36 Janus kinase 1 Homo sapiens 117-121 18413761-7 2008 These findings indicate that CDDO-Me inhibits activation of the JAK1-->STAT3 pathway by forming adducts with both JAK1 and STAT3. bardoxolone methyl 29-36 signal transducer and activator of transcription 3 Homo sapiens 126-131 18472640-0 2008 CDDO-Me inhibits proliferation, induces apoptosis, down-regulates Akt, mTOR, NF-kappaB and NF-kappaB-regulated antiapoptotic and proangiogenic proteins in TRAMP prostate cancer cells. bardoxolone methyl 0-7 thymoma viral proto-oncogene 1 Mus musculus 66-69 18472640-0 2008 CDDO-Me inhibits proliferation, induces apoptosis, down-regulates Akt, mTOR, NF-kappaB and NF-kappaB-regulated antiapoptotic and proangiogenic proteins in TRAMP prostate cancer cells. bardoxolone methyl 0-7 mechanistic target of rapamycin kinase Mus musculus 71-75 18472640-0 2008 CDDO-Me inhibits proliferation, induces apoptosis, down-regulates Akt, mTOR, NF-kappaB and NF-kappaB-regulated antiapoptotic and proangiogenic proteins in TRAMP prostate cancer cells. bardoxolone methyl 0-7 tumor necrosis factor receptor superfamily, member 25 Mus musculus 155-160 18472640-7 2008 Further, CDDO-Me inhibited NF-kappaB-regulated antiapoptotic Bcl-2, Bcl-xL, and XIAP and proangiogenic VEGF. bardoxolone methyl 9-16 B cell leukemia/lymphoma 2 Mus musculus 61-66 18472640-7 2008 Further, CDDO-Me inhibited NF-kappaB-regulated antiapoptotic Bcl-2, Bcl-xL, and XIAP and proangiogenic VEGF. bardoxolone methyl 9-16 BCL2-like 1 Mus musculus 68-74 18472640-7 2008 Further, CDDO-Me inhibited NF-kappaB-regulated antiapoptotic Bcl-2, Bcl-xL, and XIAP and proangiogenic VEGF. bardoxolone methyl 9-16 X-linked inhibitor of apoptosis Mus musculus 80-84 18472640-7 2008 Further, CDDO-Me inhibited NF-kappaB-regulated antiapoptotic Bcl-2, Bcl-xL, and XIAP and proangiogenic VEGF. bardoxolone methyl 9-16 vascular endothelial growth factor A Mus musculus 103-107 18065492-6 2007 We also show antiangiogenic effects of CDDO-Me on xenografts of Kaposi"s sarcoma cells in immunocompromised mice, using CD31 as a marker. bardoxolone methyl 39-46 platelet/endothelial cell adhesion molecule 1 Mus musculus 120-124 18065492-7 2007 Several known individual molecular targets of CDDO-Me and related triterpenoids that are relevant to all of these findings include nuclear factor-kappaB signaling, signal transducers and activators of transcription signaling, and transforming growth factor-beta signaling, as well as Keap1, the endogenous inhibitor of the transcription factor Nrf2. bardoxolone methyl 46-53 nuclear factor, erythroid derived 2, like 2 Mus musculus 344-348 17822364-10 2007 In conclusion, these results demonstrate that activation of the Nrf2-dependent antioxidative pathway by CDDO-Im or CDDO-Me protects against the LPS-induced inflammatory response and suggest that they can be potential therapeutic candidates for intervening sepsis syndrome. bardoxolone methyl 115-122 NFE2 like bZIP transcription factor 2 Homo sapiens 64-68 18277094-4 2008 After treatment of human neuroblastoma cells with CDDO-Me, cell cycle studies show depletion of the S-phase, while apoptosis studies show conformational activation and mitochondrial translocation of Bax protein, as well as activation of caspases -3 and -8. bardoxolone methyl 50-57 BCL2 associated X, apoptosis regulator Homo sapiens 199-202 18277094-4 2008 After treatment of human neuroblastoma cells with CDDO-Me, cell cycle studies show depletion of the S-phase, while apoptosis studies show conformational activation and mitochondrial translocation of Bax protein, as well as activation of caspases -3 and -8. bardoxolone methyl 50-57 caspase 3 Homo sapiens 237-255 18413761-0 2008 Triterpenoid CDDO-methyl ester inhibits the Janus-activated kinase-1 (JAK1)-->signal transducer and activator of transcription-3 (STAT3) pathway by direct inhibition of JAK1 and STAT3. bardoxolone methyl 13-30 Janus kinase 1 Homo sapiens 70-74 18413761-0 2008 Triterpenoid CDDO-methyl ester inhibits the Janus-activated kinase-1 (JAK1)-->signal transducer and activator of transcription-3 (STAT3) pathway by direct inhibition of JAK1 and STAT3. bardoxolone methyl 13-30 signal transducer and activator of transcription 3 Homo sapiens 81-131 18413761-0 2008 Triterpenoid CDDO-methyl ester inhibits the Janus-activated kinase-1 (JAK1)-->signal transducer and activator of transcription-3 (STAT3) pathway by direct inhibition of JAK1 and STAT3. bardoxolone methyl 13-30 signal transducer and activator of transcription 3 Homo sapiens 133-138 18413761-0 2008 Triterpenoid CDDO-methyl ester inhibits the Janus-activated kinase-1 (JAK1)-->signal transducer and activator of transcription-3 (STAT3) pathway by direct inhibition of JAK1 and STAT3. bardoxolone methyl 13-30 Janus kinase 1 Homo sapiens 172-176 18608528-4 2008 The suppression of these cell-mediated immune responses by CDDO-Me was associated with the inhibition of NF-kappaB transcription factor. bardoxolone methyl 59-66 nuclear factor kappa B subunit 1 Homo sapiens 105-114 17822364-0 2007 Preclinical evaluation of targeting the Nrf2 pathway by triterpenoids (CDDO-Im and CDDO-Me) for protection from LPS-induced inflammatory response and reactive oxygen species in human peripheral blood mononuclear cells and neutrophils. bardoxolone methyl 83-90 NFE2 like bZIP transcription factor 2 Homo sapiens 40-44 18253090-1 2007 The novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me) induces apoptosis of cancer cells, enhances tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis, and exhibits potent anticancer activity in animal models with a favorable pharmacokinetic profile. bardoxolone methyl 85-92 TNF superfamily member 10 Homo sapiens 138-193 18253090-1 2007 The novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me) induces apoptosis of cancer cells, enhances tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-induced apoptosis, and exhibits potent anticancer activity in animal models with a favorable pharmacokinetic profile. bardoxolone methyl 85-92 TNF superfamily member 10 Homo sapiens 195-200 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 50-57 mitogen-activated protein kinase 8 Homo sapiens 186-209 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 50-57 mitogen-activated protein kinase 8 Homo sapiens 211-214 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 50-57 TNF receptor superfamily member 10b Homo sapiens 242-258 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 50-57 TNF receptor superfamily member 10b Homo sapiens 260-263 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 150-157 mitogen-activated protein kinase 8 Homo sapiens 186-209 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 150-157 mitogen-activated protein kinase 8 Homo sapiens 211-214 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 150-157 TNF receptor superfamily member 10b Homo sapiens 242-258 18253090-3 2007 In an effort to understand the mechanism by which CDDO-Me induces apoptosis, particularly in human lung cancer cells, we previously demonstrated that CDDO-Me induces apoptosis involving c-Jun N-terminal kinase (JNK)-dependent upregulation of death receptor 5 (DR5) expression. bardoxolone methyl 150-157 TNF receptor superfamily member 10b Homo sapiens 260-263 18253090-4 2007 In the current work, we determined the modulatory effects of CDDO-Me on the levels of c-FLIP, a major inhibitor of death receptor-mediated caspase-8 activation, and its impact on CDDO-Me-induced apoptosis and enhancement of TRAIL-induced apoptosis in human lung cancer cells. bardoxolone methyl 61-68 CASP8 and FADD like apoptosis regulator Homo sapiens 86-92 18253090-4 2007 In the current work, we determined the modulatory effects of CDDO-Me on the levels of c-FLIP, a major inhibitor of death receptor-mediated caspase-8 activation, and its impact on CDDO-Me-induced apoptosis and enhancement of TRAIL-induced apoptosis in human lung cancer cells. bardoxolone methyl 61-68 caspase 8 Homo sapiens 139-148 18253090-4 2007 In the current work, we determined the modulatory effects of CDDO-Me on the levels of c-FLIP, a major inhibitor of death receptor-mediated caspase-8 activation, and its impact on CDDO-Me-induced apoptosis and enhancement of TRAIL-induced apoptosis in human lung cancer cells. bardoxolone methyl 61-68 TNF superfamily member 10 Homo sapiens 224-229 18253090-4 2007 In the current work, we determined the modulatory effects of CDDO-Me on the levels of c-FLIP, a major inhibitor of death receptor-mediated caspase-8 activation, and its impact on CDDO-Me-induced apoptosis and enhancement of TRAIL-induced apoptosis in human lung cancer cells. bardoxolone methyl 179-186 TNF superfamily member 10 Homo sapiens 224-229 18253090-5 2007 CDDO-Me rapidly and potently decreased c-FLIP levels including both long (FLIP(L)) and short (FLIP(S)) forms of c-FLIP in multiple human lung cancer cell lines. bardoxolone methyl 0-7 CASP8 and FADD like apoptosis regulator Homo sapiens 39-45 18253090-5 2007 CDDO-Me rapidly and potently decreased c-FLIP levels including both long (FLIP(L)) and short (FLIP(S)) forms of c-FLIP in multiple human lung cancer cell lines. bardoxolone methyl 0-7 CASP8 and FADD like apoptosis regulator Homo sapiens 112-118 18253090-7 2007 Moreover, CDDO-Me increased ubiquitination of c-FLIP. bardoxolone methyl 10-17 CASP8 and FADD like apoptosis regulator Homo sapiens 46-52 18253090-8 2007 Thus, CDDO-Me induces ubiquitin/proteasome-dependent c-FLIP degradation independently of JNK activation. bardoxolone methyl 6-13 CASP8 and FADD like apoptosis regulator Homo sapiens 53-59 18253090-9 2007 Importantly, overexpression of c-FLIP (e.g., FLIP(L)) protected cells not only from CDDO-Me-induced apoptosis, but also from induction of apoptosis by the combination of CDDO-Me and TRAIL. bardoxolone methyl 84-91 CASP8 and FADD like apoptosis regulator Homo sapiens 31-37 18253090-9 2007 Importantly, overexpression of c-FLIP (e.g., FLIP(L)) protected cells not only from CDDO-Me-induced apoptosis, but also from induction of apoptosis by the combination of CDDO-Me and TRAIL. bardoxolone methyl 170-177 CASP8 and FADD like apoptosis regulator Homo sapiens 31-37 18253090-10 2007 Accordingly, silencing of c-FLIP with c-FLIP siRNA sensitized cancer cells to CDDO-Me. bardoxolone methyl 78-85 CASP8 and FADD like apoptosis regulator Homo sapiens 26-32 18253090-10 2007 Accordingly, silencing of c-FLIP with c-FLIP siRNA sensitized cancer cells to CDDO-Me. bardoxolone methyl 78-85 CASP8 and FADD like apoptosis regulator Homo sapiens 38-44 18253090-11 2007 Collectively, these results indicate that c-FLIP downregulation contributes to CDDO-Me-initiated apoptosis and also to enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 79-86 CASP8 and FADD like apoptosis regulator Homo sapiens 42-48 18253090-11 2007 Collectively, these results indicate that c-FLIP downregulation contributes to CDDO-Me-initiated apoptosis and also to enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 161-168 CASP8 and FADD like apoptosis regulator Homo sapiens 42-48 18253090-11 2007 Collectively, these results indicate that c-FLIP downregulation contributes to CDDO-Me-initiated apoptosis and also to enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 161-168 TNF superfamily member 10 Homo sapiens 134-139 16998237-0 2006 Triterpenoid CDDO-Me blocks the NF-kappaB pathway by direct inhibition of IKKbeta on Cys-179. bardoxolone methyl 13-20 inhibitor of nuclear factor kappa B kinase subunit beta Homo sapiens 74-81 17970042-0 2007 CDDO-me induces apoptosis and inhibits Akt, mTOR and NF-kappaB signaling proteins in prostate cancer cells. bardoxolone methyl 0-7 AKT serine/threonine kinase 1 Homo sapiens 39-42 17970042-0 2007 CDDO-me induces apoptosis and inhibits Akt, mTOR and NF-kappaB signaling proteins in prostate cancer cells. bardoxolone methyl 0-7 mechanistic target of rapamycin kinase Homo sapiens 44-48 17970042-0 2007 CDDO-me induces apoptosis and inhibits Akt, mTOR and NF-kappaB signaling proteins in prostate cancer cells. bardoxolone methyl 0-7 nuclear factor kappa B subunit 1 Homo sapiens 53-62 17970042-6 2007 Furthermore, CDDO-Me inhibited the levels of anti-apoptotic Akt, mTOR and NF-kappaB (p65) signaling molecules. bardoxolone methyl 13-20 AKT serine/threonine kinase 1 Homo sapiens 60-63 17970042-6 2007 Furthermore, CDDO-Me inhibited the levels of anti-apoptotic Akt, mTOR and NF-kappaB (p65) signaling molecules. bardoxolone methyl 13-20 mechanistic target of rapamycin kinase Homo sapiens 65-69 17970042-6 2007 Furthermore, CDDO-Me inhibited the levels of anti-apoptotic Akt, mTOR and NF-kappaB (p65) signaling molecules. bardoxolone methyl 13-20 nuclear factor kappa B subunit 1 Homo sapiens 74-83 17970042-6 2007 Furthermore, CDDO-Me inhibited the levels of anti-apoptotic Akt, mTOR and NF-kappaB (p65) signaling molecules. bardoxolone methyl 13-20 RELA proto-oncogene, NF-kB subunit Homo sapiens 85-88 17483332-7 2007 In vitro, after treatment of 4T1 cells with 500 nmol/L CDDO-Me for 2 h, we found (a) inactivation of STAT3, (b) inactivation of Src and Akt, (c) 4-fold reduction of c-Myc mRNA levels, (d) accumulation of cells in G(2)-M cell cycle phase, (e) abrogation of invasive growth of 4T1 cells, and (f) lack of apoptosis induction. bardoxolone methyl 55-62 signal transducer and activator of transcription 3 Mus musculus 101-106 17483332-7 2007 In vitro, after treatment of 4T1 cells with 500 nmol/L CDDO-Me for 2 h, we found (a) inactivation of STAT3, (b) inactivation of Src and Akt, (c) 4-fold reduction of c-Myc mRNA levels, (d) accumulation of cells in G(2)-M cell cycle phase, (e) abrogation of invasive growth of 4T1 cells, and (f) lack of apoptosis induction. bardoxolone methyl 55-62 Rous sarcoma oncogene Mus musculus 128-131 17483332-7 2007 In vitro, after treatment of 4T1 cells with 500 nmol/L CDDO-Me for 2 h, we found (a) inactivation of STAT3, (b) inactivation of Src and Akt, (c) 4-fold reduction of c-Myc mRNA levels, (d) accumulation of cells in G(2)-M cell cycle phase, (e) abrogation of invasive growth of 4T1 cells, and (f) lack of apoptosis induction. bardoxolone methyl 55-62 thymoma viral proto-oncogene 1 Mus musculus 136-139 17483332-10 2007 In summary, these data suggest that CDDO-Me may have therapeutic potential in breast cancer therapy, in part, through inactivation of STAT3. bardoxolone methyl 36-43 signal transducer and activator of transcription 3 Mus musculus 134-139 17361329-10 2007 Furthermore, CDDO-Me inhibited the levels of anti-apoptotic and prosurvival p-Akt, NF-kappaB (p65) and Notch1 signaling molecules. bardoxolone methyl 13-20 AKT serine/threonine kinase 1 Homo sapiens 78-81 17361329-10 2007 Furthermore, CDDO-Me inhibited the levels of anti-apoptotic and prosurvival p-Akt, NF-kappaB (p65) and Notch1 signaling molecules. bardoxolone methyl 13-20 RELA proto-oncogene, NF-kB subunit Homo sapiens 94-97 17361329-10 2007 Furthermore, CDDO-Me inhibited the levels of anti-apoptotic and prosurvival p-Akt, NF-kappaB (p65) and Notch1 signaling molecules. bardoxolone methyl 13-20 notch receptor 1 Homo sapiens 103-109 16998237-2 2006 The present studies show that CDDO and CDDO-Me block tumor necrosis factoralpha-induced targeting of NF-kappaB p65 to the nucleus. bardoxolone methyl 39-46 RELA proto-oncogene, NF-kB subunit Homo sapiens 111-114 16998237-3 2006 CDDO-Me also blocked tumor necrosis factor alpha-induced phosphorylation of IkappaBalpha. bardoxolone methyl 0-7 tumor necrosis factor Homo sapiens 21-48 16998237-3 2006 CDDO-Me also blocked tumor necrosis factor alpha-induced phosphorylation of IkappaBalpha. bardoxolone methyl 0-7 NFKB inhibitor alpha Homo sapiens 76-88 16998237-4 2006 In concert with these results, we found that CDDO-Me inhibits IkappaBalpha kinasebeta (IKKbeta) activity in cells. bardoxolone methyl 45-52 NFKB inhibitor alpha Homo sapiens 62-74 16998237-4 2006 In concert with these results, we found that CDDO-Me inhibits IkappaBalpha kinasebeta (IKKbeta) activity in cells. bardoxolone methyl 45-52 inhibitor of nuclear factor kappa B kinase subunit beta Homo sapiens 87-94 16998237-5 2006 In support of a direct mechanism, CDDO-Me inhibited recombinant IKKbeta activity in vitro. bardoxolone methyl 34-41 inhibitor of nuclear factor kappa B kinase subunit beta Homo sapiens 64-71 16998237-6 2006 The results also demonstrate that (i) CDDO and CDDO-Me form adducts with IKKbeta, but not IKKbeta with mutation of Cys-179 to Ala, and (ii) CDDO-Me inhibits IKKbeta by a mechanism dependent on oxidation of Cys-179. bardoxolone methyl 47-54 inhibitor of nuclear factor kappa B kinase subunit beta Homo sapiens 73-80 16998237-7 2006 These findings indicate that CDDO and CDDO-Me directly block IKKbeta activity and thereby the NF-kappaB pathway by interacting with Cys-179 in the IKKbeta activation loop. bardoxolone methyl 38-45 inhibitor of nuclear factor kappa B kinase subunit beta Homo sapiens 61-68 16998237-7 2006 These findings indicate that CDDO and CDDO-Me directly block IKKbeta activity and thereby the NF-kappaB pathway by interacting with Cys-179 in the IKKbeta activation loop. bardoxolone methyl 38-45 inhibitor of nuclear factor kappa B kinase subunit beta Homo sapiens 147-154 16582599-0 2006 Depletion of intracellular glutathione contributes to JNK-mediated death receptor 5 upregulation and apoptosis induction by the novel synthetic triterpenoid methyl-2-cyano-3, 12-dioxooleana-1, 9-dien-28-oate (CDDO-Me). bardoxolone methyl 209-216 mitogen-activated protein kinase 8 Homo sapiens 54-57 16582599-5 2006 Thiol antioxidants, including N-acetyl-L-cycteine (NAC), glutathione (GSH) and dithiothrietol (DTT), abrogated CDDO-Me-induced apoptosis. bardoxolone methyl 111-118 X-linked Kx blood group Homo sapiens 30-49 16582599-2 2006 We previously demonstrated that CDDO-Me induces a c-Jun N-terminal kinase (JNK)-mediated DR5 expression and apoptosis. bardoxolone methyl 32-39 mitogen-activated protein kinase 8 Homo sapiens 50-73 16582599-5 2006 Thiol antioxidants, including N-acetyl-L-cycteine (NAC), glutathione (GSH) and dithiothrietol (DTT), abrogated CDDO-Me-induced apoptosis. bardoxolone methyl 111-118 X-linked Kx blood group Homo sapiens 51-54 16582599-2 2006 We previously demonstrated that CDDO-Me induces a c-Jun N-terminal kinase (JNK)-mediated DR5 expression and apoptosis. bardoxolone methyl 32-39 mitogen-activated protein kinase 8 Homo sapiens 75-78 16582599-2 2006 We previously demonstrated that CDDO-Me induces a c-Jun N-terminal kinase (JNK)-mediated DR5 expression and apoptosis. bardoxolone methyl 32-39 TNF receptor superfamily member 10b Homo sapiens 89-92 16582599-7 2006 Accordingly, only thiol antioxidants blocked JNK activation induced by CDDO-Me. bardoxolone methyl 71-78 mitogen-activated protein kinase 8 Homo sapiens 45-48 16582599-3 2006 This study revealed the mechanism by which CDDO-Me induces JNK activation and subsequent DR5 upregulation and apoptosis. bardoxolone methyl 43-50 mitogen-activated protein kinase 8 Homo sapiens 59-62 16582599-10 2006 These results suggest that depletion of intracellular GSH, but not ROS generation, contributes to CDDO-Me-induced JNK activation and apoptosis, at least in our systems. bardoxolone methyl 98-105 mitogen-activated protein kinase 8 Homo sapiens 114-117 16582599-3 2006 This study revealed the mechanism by which CDDO-Me induces JNK activation and subsequent DR5 upregulation and apoptosis. bardoxolone methyl 43-50 TNF receptor superfamily member 10b Homo sapiens 89-92 16582599-4 2006 To determine whether CDDO-Me activates JNK and induces DR5 expression and apoptosis via oxidative stress by inducing the generation of reactive oxygen species (ROS), we examined the effects of various antioxidants on JNK activation, DR5 upregulation, and apoptosis induction by CDDO-Me. bardoxolone methyl 21-28 mitogen-activated protein kinase 8 Homo sapiens 39-42 16582599-4 2006 To determine whether CDDO-Me activates JNK and induces DR5 expression and apoptosis via oxidative stress by inducing the generation of reactive oxygen species (ROS), we examined the effects of various antioxidants on JNK activation, DR5 upregulation, and apoptosis induction by CDDO-Me. bardoxolone methyl 21-28 TNF receptor superfamily member 10b Homo sapiens 55-58 16551868-0 2006 A synthetic triterpenoid, CDDO-Me, inhibits IkappaBalpha kinase and enhances apoptosis induced by TNF and chemotherapeutic agents through down-regulation of expression of nuclear factor kappaB-regulated gene products in human leukemic cells. bardoxolone methyl 26-33 NFKB inhibitor alpha Homo sapiens 44-56 16582599-11 2006 Furthermore, these thiol antioxidants abrogated CDDO-Me-induced DR5 expression, whereas the GSH-depleting agent diethylmaleate also upregulated DR5 expression at concentrations that deplete intracellular GSH, demonstrating that GSH depletion can cause DR5 upregulation. bardoxolone methyl 48-55 TNF receptor superfamily member 10b Homo sapiens 64-67 16582599-12 2006 Collectively, we conclude that CDDO-Me activates the JNK pathway via depletion of intracellular GSH, leading to DR5 upregulation and induction of apoptosis. bardoxolone methyl 31-38 mitogen-activated protein kinase 8 Homo sapiens 53-56 16582599-12 2006 Collectively, we conclude that CDDO-Me activates the JNK pathway via depletion of intracellular GSH, leading to DR5 upregulation and induction of apoptosis. bardoxolone methyl 31-38 TNF receptor superfamily member 10b Homo sapiens 112-115 16410408-7 2006 Finally, transmission electron microscopy revealed that CDDO-Me rapidly induced caspase-independent mitochondrial swelling and loss of inner membrane structure before the release of cytochrome c. bardoxolone methyl 56-63 cytochrome c, somatic Homo sapiens 182-194 16551868-0 2006 A synthetic triterpenoid, CDDO-Me, inhibits IkappaBalpha kinase and enhances apoptosis induced by TNF and chemotherapeutic agents through down-regulation of expression of nuclear factor kappaB-regulated gene products in human leukemic cells. bardoxolone methyl 26-33 tumor necrosis factor Homo sapiens 98-101 16551868-2 2006 Because the nuclear transcription factor nuclear factor kappaB (NF-kappaB) has been shown to suppress apoptosis and promote proliferation and is linked with inflammation and differentiation, we postulated that CDDO-Me modulates NF-kappaB activity and NF-kappaB-regulated gene expression. bardoxolone methyl 210-217 nuclear factor kappa B subunit 1 Homo sapiens 64-73 16551868-2 2006 Because the nuclear transcription factor nuclear factor kappaB (NF-kappaB) has been shown to suppress apoptosis and promote proliferation and is linked with inflammation and differentiation, we postulated that CDDO-Me modulates NF-kappaB activity and NF-kappaB-regulated gene expression. bardoxolone methyl 210-217 nuclear factor kappa B subunit 1 Homo sapiens 228-237 16551868-2 2006 Because the nuclear transcription factor nuclear factor kappaB (NF-kappaB) has been shown to suppress apoptosis and promote proliferation and is linked with inflammation and differentiation, we postulated that CDDO-Me modulates NF-kappaB activity and NF-kappaB-regulated gene expression. bardoxolone methyl 210-217 nuclear factor kappa B subunit 1 Homo sapiens 228-237 16551868-3 2006 Using human leukemia cell lines and patient samples, we show that CDDO-Me potently inhibits both constitutive and inducible NF-kappaB activated by tumor necrosis factor (TNF), interleukin (IL)-1beta, phorbol ester, okadaic acid, hydrogen peroxide, lipopolysaccharide, and cigarette smoke. bardoxolone methyl 66-73 nuclear factor kappa B subunit 1 Homo sapiens 124-133 16551868-3 2006 Using human leukemia cell lines and patient samples, we show that CDDO-Me potently inhibits both constitutive and inducible NF-kappaB activated by tumor necrosis factor (TNF), interleukin (IL)-1beta, phorbol ester, okadaic acid, hydrogen peroxide, lipopolysaccharide, and cigarette smoke. bardoxolone methyl 66-73 tumor necrosis factor Homo sapiens 147-168 16551868-3 2006 Using human leukemia cell lines and patient samples, we show that CDDO-Me potently inhibits both constitutive and inducible NF-kappaB activated by tumor necrosis factor (TNF), interleukin (IL)-1beta, phorbol ester, okadaic acid, hydrogen peroxide, lipopolysaccharide, and cigarette smoke. bardoxolone methyl 66-73 tumor necrosis factor Homo sapiens 170-173 16551868-3 2006 Using human leukemia cell lines and patient samples, we show that CDDO-Me potently inhibits both constitutive and inducible NF-kappaB activated by tumor necrosis factor (TNF), interleukin (IL)-1beta, phorbol ester, okadaic acid, hydrogen peroxide, lipopolysaccharide, and cigarette smoke. bardoxolone methyl 66-73 interleukin 1 beta Homo sapiens 176-198 16551868-7 2006 CDDO-Me also potentiated the cytotoxic effects of TNF and chemotherapeutic agents. bardoxolone methyl 0-7 tumor necrosis factor Homo sapiens 50-53 16551868-8 2006 Overall, our results suggest that CDDO-Me inhibits NF-kappaB through inhibition of IkappaBalpha kinase, leading to the suppression of expression of NF-kappaB-regulated gene products and enhancement of apoptosis induced by TNF and chemotherapeutic agents. bardoxolone methyl 34-41 nuclear factor kappa B subunit 1 Homo sapiens 51-60 16551868-8 2006 Overall, our results suggest that CDDO-Me inhibits NF-kappaB through inhibition of IkappaBalpha kinase, leading to the suppression of expression of NF-kappaB-regulated gene products and enhancement of apoptosis induced by TNF and chemotherapeutic agents. bardoxolone methyl 34-41 NFKB inhibitor alpha Homo sapiens 83-95 16551868-8 2006 Overall, our results suggest that CDDO-Me inhibits NF-kappaB through inhibition of IkappaBalpha kinase, leading to the suppression of expression of NF-kappaB-regulated gene products and enhancement of apoptosis induced by TNF and chemotherapeutic agents. bardoxolone methyl 34-41 nuclear factor kappa B subunit 1 Homo sapiens 148-157 16551868-8 2006 Overall, our results suggest that CDDO-Me inhibits NF-kappaB through inhibition of IkappaBalpha kinase, leading to the suppression of expression of NF-kappaB-regulated gene products and enhancement of apoptosis induced by TNF and chemotherapeutic agents. bardoxolone methyl 34-41 tumor necrosis factor Homo sapiens 222-225 12467212-11 2002 In addition, no correlation was found between cell sensitivity to CDDO-Me and p53 status, suggesting that CDDO-Me induce a p53-independent apoptosis. bardoxolone methyl 106-113 tumor protein p53 Homo sapiens 123-126 15931262-0 2005 The novel triterpenoid CDDO-Me suppresses MAPK pathways and promotes p38 activation in acute myeloid leukemia cells. bardoxolone methyl 23-30 mitogen-activated protein kinase 3 Homo sapiens 42-46 15931262-0 2005 The novel triterpenoid CDDO-Me suppresses MAPK pathways and promotes p38 activation in acute myeloid leukemia cells. bardoxolone methyl 23-30 mitogen-activated protein kinase 1 Homo sapiens 69-72 15931262-3 2005 In this report, the effects of CDDO-Me on CD34(+) AML progenitor cells in vitro were examined. bardoxolone methyl 31-38 CD34 molecule Homo sapiens 42-46 15931262-5 2005 CDDO-Me is known to inhibit the activation of ERK1/2. bardoxolone methyl 0-7 mitogen-activated protein kinase 3 Homo sapiens 46-52 15931262-6 2005 In this series of primary AML samples, ERK was expressed and phosphorylated in all patient samples studied and CDDO-Me inhibited ERK phosphorylation in five of 10 samples. bardoxolone methyl 111-118 mitogen-activated protein kinase 1 Homo sapiens 129-132 15931262-8 2005 CDDO-Me induced phosphorylation of p38 in AML-derived U937 cells. bardoxolone methyl 0-7 mitogen-activated protein kinase 1 Homo sapiens 35-38 15931262-9 2005 Pretreatment of U937 cells with a p38 inhibitor protected cells from the cyto-toxic effects of CDDO-Me. bardoxolone methyl 95-102 mitogen-activated protein kinase 1 Homo sapiens 34-37 15931262-10 2005 These findings suggest a role for p38 in CDDO-Me-induced apoptosis. bardoxolone methyl 41-48 mitogen-activated protein kinase 1 Homo sapiens 34-37 15931262-11 2005 In preliminary studies, CDDO-Me induced p38 phosphorylation in seven of eight primary AML samples. bardoxolone methyl 24-31 mitogen-activated protein kinase 1 Homo sapiens 40-43 15798084-1 2005 2-Cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO) and the corresponding methyl (CDDO-Me) and imidazole (CDDO-Im) esters induce peroxisome proliferator-activated receptor gamma (PPARgamma)-dependent transactivation in SW-480 colon cancer cells, and these responses were inhibited by small inhibitory RNA for PPARgamma. bardoxolone methyl 82-89 peroxisome proliferator activated receptor gamma Homo sapiens 129-177 15798084-1 2005 2-Cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO) and the corresponding methyl (CDDO-Me) and imidazole (CDDO-Im) esters induce peroxisome proliferator-activated receptor gamma (PPARgamma)-dependent transactivation in SW-480 colon cancer cells, and these responses were inhibited by small inhibitory RNA for PPARgamma. bardoxolone methyl 82-89 peroxisome proliferator activated receptor gamma Homo sapiens 179-188 15798084-1 2005 2-Cyano-3,12-dioxoolean-1,9-dien-28-oic acid (CDDO) and the corresponding methyl (CDDO-Me) and imidazole (CDDO-Im) esters induce peroxisome proliferator-activated receptor gamma (PPARgamma)-dependent transactivation in SW-480 colon cancer cells, and these responses were inhibited by small inhibitory RNA for PPARgamma. bardoxolone methyl 82-89 peroxisome proliferator activated receptor gamma Homo sapiens 309-318 15798084-6 2005 Induction of caveolin-1 by CDDO, CDDO-Me, and CDDO-Im was inhibited by the PPARgamma antagonist N-(4"-aminopyridyl-2-chloro-5-nitrobenzamide (T007), whereas higher doses induced apoptosis [poly(ADP-ribose) polymerase cleavage], which was not inhibited by T007. bardoxolone methyl 33-40 caveolin 1 Homo sapiens 13-23 15798084-6 2005 Induction of caveolin-1 by CDDO, CDDO-Me, and CDDO-Im was inhibited by the PPARgamma antagonist N-(4"-aminopyridyl-2-chloro-5-nitrobenzamide (T007), whereas higher doses induced apoptosis [poly(ADP-ribose) polymerase cleavage], which was not inhibited by T007. bardoxolone methyl 33-40 peroxisome proliferator activated receptor gamma Homo sapiens 75-84 15798084-7 2005 These results illustrate that CDDO-, CDDO-Me, and CDDO-Im induce both PPARgamma-dependent and -independent responses in colon cancer cells, and activation of these pathways are separable and concentration-dependent for all three compounds. bardoxolone methyl 37-44 peroxisome proliferator activated receptor gamma Homo sapiens 70-79 15492284-8 2004 Moreover, application of the JNK-specific inhibitor SP600125 blocked CDDO-Me-induced increases in JNK activation, DR up-regulation, caspase-8 activation, and DNA fragmentation. bardoxolone methyl 69-76 caspase 8 Homo sapiens 132-141 15492284-9 2004 These results show that activation of JNK pathway results in CDDO-Me-induced DR up-regulation, caspase-8 activation, and apoptosis. bardoxolone methyl 61-68 mitogen-activated protein kinase 8 Homo sapiens 38-41 15492284-10 2004 Collectively, we conclude that CDDO-Me induces apoptosis via the JNK-mediated DR up-regulation in human lung cancer cells. bardoxolone methyl 31-38 mitogen-activated protein kinase 8 Homo sapiens 65-68 15492284-4 2004 We found that CDDO-Me not only activated caspase-8 but also induced expression of DRs, particularly DR5, in a p53-independent mechanism. bardoxolone methyl 14-21 caspase 8 Homo sapiens 41-50 15492284-4 2004 We found that CDDO-Me not only activated caspase-8 but also induced expression of DRs, particularly DR5, in a p53-independent mechanism. bardoxolone methyl 14-21 TNF receptor superfamily member 10b Homo sapiens 100-103 15492284-4 2004 We found that CDDO-Me not only activated caspase-8 but also induced expression of DRs, particularly DR5, in a p53-independent mechanism. bardoxolone methyl 14-21 tumor protein p53 Homo sapiens 110-113 15492284-5 2004 Correspondingly, CDDO-Me augmented TRAIL-induced apoptosis in these cells regardless of p53 status as evidenced by enhanced DNA fragmentation and activation of caspase cascades, suggesting that CDDO-Me-induced DRs are functionally active. bardoxolone methyl 17-24 TNF superfamily member 10 Homo sapiens 35-40 15492284-5 2004 Correspondingly, CDDO-Me augmented TRAIL-induced apoptosis in these cells regardless of p53 status as evidenced by enhanced DNA fragmentation and activation of caspase cascades, suggesting that CDDO-Me-induced DRs are functionally active. bardoxolone methyl 194-201 TNF superfamily member 10 Homo sapiens 35-40 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 98-105 TNF receptor superfamily member 10b Homo sapiens 23-26 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 98-105 TNF superfamily member 10 Homo sapiens 145-150 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 98-105 TNF superfamily member 10 Homo sapiens 259-264 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF receptor superfamily member 10b Homo sapiens 23-26 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF superfamily member 10 Homo sapiens 259-264 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF receptor superfamily member 10b Homo sapiens 23-26 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF superfamily member 10 Homo sapiens 259-264 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF receptor superfamily member 10b Homo sapiens 23-26 15492284-6 2004 Moreover, silencing of DR5 expression using small interfering RNA suppressed apoptosis induced by CDDO-Me alone or by combination of CDDO-Me and TRAIL, indicating that DR5 up-regulation is required for induction of apoptosis by CDDO-Me and for enhancement of TRAIL-induced apoptosis by CDDO-Me. bardoxolone methyl 133-140 TNF superfamily member 10 Homo sapiens 259-264 15492284-7 2004 CDDO-Me rapidly activated c-Jun NH(2)-terminal kinase (JNK) before DR up-regulation and caspase-8 activation. bardoxolone methyl 0-7 mitogen-activated protein kinase 8 Homo sapiens 55-58 15492284-7 2004 CDDO-Me rapidly activated c-Jun NH(2)-terminal kinase (JNK) before DR up-regulation and caspase-8 activation. bardoxolone methyl 0-7 caspase 8 Homo sapiens 88-97 15492284-8 2004 Moreover, application of the JNK-specific inhibitor SP600125 blocked CDDO-Me-induced increases in JNK activation, DR up-regulation, caspase-8 activation, and DNA fragmentation. bardoxolone methyl 69-76 mitogen-activated protein kinase 8 Homo sapiens 29-32 15492284-8 2004 Moreover, application of the JNK-specific inhibitor SP600125 blocked CDDO-Me-induced increases in JNK activation, DR up-regulation, caspase-8 activation, and DNA fragmentation. bardoxolone methyl 69-76 mitogen-activated protein kinase 8 Homo sapiens 98-101 11756188-3 2002 CDDO-Me decreased the viability of leukemic cell lines, including multidrug resistant (MDR)-1-overexpressing, p53(null) HL-60-Dox and of primary AML cells, and it was 3- to 5-fold more active than CDDO. bardoxolone methyl 0-7 tumor protein p53 Homo sapiens 110-113 11756188-4 2002 CDDO-Me induced a loss of mitochondrial membrane potential, induction of caspase-3 cleavage, increase in annexin V binding and DNA fragmentation, suggesting the induction of apoptosis. bardoxolone methyl 0-7 caspase 3 Homo sapiens 73-82 11756188-4 2002 CDDO-Me induced a loss of mitochondrial membrane potential, induction of caspase-3 cleavage, increase in annexin V binding and DNA fragmentation, suggesting the induction of apoptosis. bardoxolone methyl 0-7 annexin A5 Homo sapiens 105-114 11756188-5 2002 CDDO-Me induced pro-apoptotic Bax protein that preceded caspase activation. bardoxolone methyl 0-7 BCL2 associated X, apoptosis regulator Homo sapiens 30-33 11756188-6 2002 Furthermore, CDDO-Me inhibited the activation of ERK1/2, as determined by the inhibition of mitochondrial ERK1/2 phosphorylation, and it blocked Bcl-2 phosphorylation, rendering Bcl-2 less anti-apoptotic. bardoxolone methyl 13-20 mitogen-activated protein kinase 3 Homo sapiens 49-55 11756188-6 2002 Furthermore, CDDO-Me inhibited the activation of ERK1/2, as determined by the inhibition of mitochondrial ERK1/2 phosphorylation, and it blocked Bcl-2 phosphorylation, rendering Bcl-2 less anti-apoptotic. bardoxolone methyl 13-20 mitogen-activated protein kinase 3 Homo sapiens 106-112 11756188-6 2002 Furthermore, CDDO-Me inhibited the activation of ERK1/2, as determined by the inhibition of mitochondrial ERK1/2 phosphorylation, and it blocked Bcl-2 phosphorylation, rendering Bcl-2 less anti-apoptotic. bardoxolone methyl 13-20 BCL2 apoptosis regulator Homo sapiens 145-150 11756188-6 2002 Furthermore, CDDO-Me inhibited the activation of ERK1/2, as determined by the inhibition of mitochondrial ERK1/2 phosphorylation, and it blocked Bcl-2 phosphorylation, rendering Bcl-2 less anti-apoptotic. bardoxolone methyl 13-20 BCL2 apoptosis regulator Homo sapiens 178-183 11756188-9 2002 Combinations with ATRA or the RXR-specific ligand LG100268 enhanced the effects of CDDO-Me on cell viability and terminal differentiation of myeloid leukemic cell lines. bardoxolone methyl 83-90 retinoid X receptor alpha Homo sapiens 30-33 11756188-10 2002 In conclusion, CDDO-Me is an MDR-1- and a p53-independent compound that exerts strong antiproliferative, apoptotic, and differentiating effects in myeloid leukemic cell lines and in primary AML samples when given in submicromolar concentrations. bardoxolone methyl 15-22 ATP binding cassette subfamily B member 1 Homo sapiens 29-34 11756188-10 2002 In conclusion, CDDO-Me is an MDR-1- and a p53-independent compound that exerts strong antiproliferative, apoptotic, and differentiating effects in myeloid leukemic cell lines and in primary AML samples when given in submicromolar concentrations. bardoxolone methyl 15-22 tumor protein p53 Homo sapiens 42-45 12467212-7 2002 These results indicate that CDDO-Me induced apoptosis in human NSCLC cells via a cytochrome c-triggered caspase activation pathway. bardoxolone methyl 28-35 cytochrome c, somatic Homo sapiens 81-93 35512420-2 2022 Herein, we reported a novel strategy to prepare Cathepsin B (CTSB) cleavable and improved water-soluble prodrugs of CDDO-Me. bardoxolone methyl 116-123 cathepsin B Homo sapiens 48-59 11043571-4 2000 The methyl ester of CDDO, CDDO-Me, binds to PPARgamma with similar affinity, but is an antagonist. bardoxolone methyl 26-33 peroxisome proliferator activated receptor gamma Mus musculus 44-53 11043571-6 2000 The partial agonism of CDDO and the antagonism of CDDO-Me reflect the differences in their capacity to recruit or displace cofactors of transcriptional regulation; CDDO and rosiglitazone both release the nuclear receptor corepressor, NCoR, from PPARgamma, while CDDO-Me does not. bardoxolone methyl 50-57 nuclear receptor co-repressor 1 Mus musculus 234-238 34573098-3 2021 Bardoxolone methyl (methyl 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate; CDDO-Me; RTA 402) is a semisynthetic triterpenoid with effects against antioxidative stress and inflammation in neurodegeneration and kidney disease that activates the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. bardoxolone methyl 74-81 NFE2 like bZIP transcription factor 2 Homo sapiens 242-285 34573098-3 2021 Bardoxolone methyl (methyl 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate; CDDO-Me; RTA 402) is a semisynthetic triterpenoid with effects against antioxidative stress and inflammation in neurodegeneration and kidney disease that activates the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. bardoxolone methyl 74-81 NFE2 like bZIP transcription factor 2 Homo sapiens 287-291 34573098-3 2021 Bardoxolone methyl (methyl 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate; CDDO-Me; RTA 402) is a semisynthetic triterpenoid with effects against antioxidative stress and inflammation in neurodegeneration and kidney disease that activates the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. bardoxolone methyl 83-90 NFE2 like bZIP transcription factor 2 Homo sapiens 242-285 34573098-3 2021 Bardoxolone methyl (methyl 2-cyano-3,12-dioxooleana-1,9(11)-dien-28-oate; CDDO-Me; RTA 402) is a semisynthetic triterpenoid with effects against antioxidative stress and inflammation in neurodegeneration and kidney disease that activates the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway. bardoxolone methyl 83-90 NFE2 like bZIP transcription factor 2 Homo sapiens 287-291 34573098-8 2021 Treatment with RTA402 results in antiapoptotic, antioxidative stress, anti-inflammatory, and myelin-preserving effects on retinal ganglion cell (RGC) survival and visual function via regulation of NQO1 and HO-1, reduced IL-6 and Iba1 expression in macrophages, and promoted microglial expression of TGF-beta and Ym1 + 2 in the retina and optic nerve. bardoxolone methyl 15-21 NAD(P)H quinone dehydrogenase 1 Homo sapiens 197-201 34573098-8 2021 Treatment with RTA402 results in antiapoptotic, antioxidative stress, anti-inflammatory, and myelin-preserving effects on retinal ganglion cell (RGC) survival and visual function via regulation of NQO1 and HO-1, reduced IL-6 and Iba1 expression in macrophages, and promoted microglial expression of TGF-beta and Ym1 + 2 in the retina and optic nerve. bardoxolone methyl 15-21 heme oxygenase 1 Homo sapiens 206-210 34573098-8 2021 Treatment with RTA402 results in antiapoptotic, antioxidative stress, anti-inflammatory, and myelin-preserving effects on retinal ganglion cell (RGC) survival and visual function via regulation of NQO1 and HO-1, reduced IL-6 and Iba1 expression in macrophages, and promoted microglial expression of TGF-beta and Ym1 + 2 in the retina and optic nerve. bardoxolone methyl 15-21 interleukin 6 Homo sapiens 220-224 34573098-8 2021 Treatment with RTA402 results in antiapoptotic, antioxidative stress, anti-inflammatory, and myelin-preserving effects on retinal ganglion cell (RGC) survival and visual function via regulation of NQO1 and HO-1, reduced IL-6 and Iba1 expression in macrophages, and promoted microglial expression of TGF-beta and Ym1 + 2 in the retina and optic nerve. bardoxolone methyl 15-21 allograft inflammatory factor 1 Homo sapiens 229-233 34573098-8 2021 Treatment with RTA402 results in antiapoptotic, antioxidative stress, anti-inflammatory, and myelin-preserving effects on retinal ganglion cell (RGC) survival and visual function via regulation of NQO1 and HO-1, reduced IL-6 and Iba1 expression in macrophages, and promoted microglial expression of TGF-beta and Ym1 + 2 in the retina and optic nerve. bardoxolone methyl 15-21 transforming growth factor alpha Homo sapiens 299-307 34573098-10 2021 Our results provide explicit evidence that RTA 402 modulates the Nrf2 and NFkappaB signaling pathways to protect RGCs from apoptosis and maintain the visual function in an rAION model. bardoxolone methyl 43-50 NFE2 like bZIP transcription factor 2 Homo sapiens 65-69 34440619-4 2021 CDDO-Me (CDDO methyl ester, an Nrf2 inducer) administration attenuated PD-like neurodegeneration mainly through Nrf2 activation in astrocytes by activating the antioxidant signaling pathway and enhancing autophagy in the substantia nigra and striatum. bardoxolone methyl 0-7 NFE2 like bZIP transcription factor 2 Rattus norvegicus 31-35 34440619-4 2021 CDDO-Me (CDDO methyl ester, an Nrf2 inducer) administration attenuated PD-like neurodegeneration mainly through Nrf2 activation in astrocytes by activating the antioxidant signaling pathway and enhancing autophagy in the substantia nigra and striatum. bardoxolone methyl 0-7 NFE2 like bZIP transcription factor 2 Rattus norvegicus 112-116 34440619-4 2021 CDDO-Me (CDDO methyl ester, an Nrf2 inducer) administration attenuated PD-like neurodegeneration mainly through Nrf2 activation in astrocytes by activating the antioxidant signaling pathway and enhancing autophagy in the substantia nigra and striatum. bardoxolone methyl 9-26 NFE2 like bZIP transcription factor 2 Rattus norvegicus 31-35 34440619-4 2021 CDDO-Me (CDDO methyl ester, an Nrf2 inducer) administration attenuated PD-like neurodegeneration mainly through Nrf2 activation in astrocytes by activating the antioxidant signaling pathway and enhancing autophagy in the substantia nigra and striatum. bardoxolone methyl 9-26 NFE2 like bZIP transcription factor 2 Rattus norvegicus 112-116 34440619-10 2021 Treatment with CDDO-Me attenuated the Nrf2-knockdown-induced degeneration in the flies through the activation of the antioxidant signaling pathway and increased autophagy. bardoxolone methyl 15-22 Keap1 Drosophila melanogaster 38-42 11043571-6 2000 The partial agonism of CDDO and the antagonism of CDDO-Me reflect the differences in their capacity to recruit or displace cofactors of transcriptional regulation; CDDO and rosiglitazone both release the nuclear receptor corepressor, NCoR, from PPARgamma, while CDDO-Me does not. bardoxolone methyl 262-269 nuclear receptor co-repressor 1 Mus musculus 234-238 34480604-5 2021 Bardoxolone-methyl (CDDO-Me) induced oxidative stress caused similarly increased expression of both the wild-type and eGFP-tagged HMOX1 at the mRNA and protein level. bardoxolone methyl 0-18 heme oxygenase 1 Homo sapiens 130-135 34480604-5 2021 Bardoxolone-methyl (CDDO-Me) induced oxidative stress caused similarly increased expression of both the wild-type and eGFP-tagged HMOX1 at the mRNA and protein level. bardoxolone methyl 20-27 heme oxygenase 1 Homo sapiens 130-135 35512420-2 2022 Herein, we reported a novel strategy to prepare Cathepsin B (CTSB) cleavable and improved water-soluble prodrugs of CDDO-Me. bardoxolone methyl 116-123 cathepsin B Homo sapiens 61-65 35512420-3 2022 CTSB linkers connection to the highly active alpha-cyano-alpha, beta-unsaturated ketone (CUK) part of CDDO-Me with the incorporation of polyethylene glycol (PEG) moieties, provided a series of prodrugs of CDDO-Me without CUK part exposure. bardoxolone methyl 102-109 cathepsin B Homo sapiens 0-4 35512420-3 2022 CTSB linkers connection to the highly active alpha-cyano-alpha, beta-unsaturated ketone (CUK) part of CDDO-Me with the incorporation of polyethylene glycol (PEG) moieties, provided a series of prodrugs of CDDO-Me without CUK part exposure. bardoxolone methyl 205-212 cathepsin B Homo sapiens 0-4 35512420-4 2022 Theoretically, these prodrugs shielding CUK part can be stably circulated and finally cleaved by CTSB in lysosomes to release CDDO-Me. bardoxolone methyl 126-133 cathepsin B Homo sapiens 97-101 35512420-7 2022 More importantly, prodrug 20 showed relatively lower human ether-a-go-go-related gene (hERG) inhibitory activity compared with CDDO-Me, which demonstrated prodrug 20 might be safer than CDDO-Me. bardoxolone methyl 127-134 ETS transcription factor ERG Homo sapiens 87-91 35512420-7 2022 More importantly, prodrug 20 showed relatively lower human ether-a-go-go-related gene (hERG) inhibitory activity compared with CDDO-Me, which demonstrated prodrug 20 might be safer than CDDO-Me. bardoxolone methyl 186-193 ETS transcription factor ERG Homo sapiens 87-91 35512420-8 2022 In conclusion, the novel strategy of shielding CUK part with CTSB linkers provided a new idea for solving the limitations of CDDO-Me in clinical application. bardoxolone methyl 125-132 cathepsin B Homo sapiens 61-65 35395074-6 2022 Targeting the Rab13-mediated BCSC niche with bardoxolone-methyl (CDDO-Me) prevented BCSC stemness in vitro and in vivo. bardoxolone methyl 45-63 RAB13, member RAS oncogene family Homo sapiens 14-19 35395074-6 2022 Targeting the Rab13-mediated BCSC niche with bardoxolone-methyl (CDDO-Me) prevented BCSC stemness in vitro and in vivo. bardoxolone methyl 65-72 RAB13, member RAS oncogene family Homo sapiens 14-19 35626147-5 2022 The siRNA-mediated knockdown of NFE2L2, the gene encoding for NRF2, or activation of the NRF2 pathway through siRNA-mediated knockdown of KEAP1 or via chemical induction with the NRF2-activator CDDO-Me confirmed that NRF2 is a regulator of KYNU expression in LUAD. bardoxolone methyl 194-201 NFE2 like bZIP transcription factor 2 Homo sapiens 32-38 35626147-5 2022 The siRNA-mediated knockdown of NFE2L2, the gene encoding for NRF2, or activation of the NRF2 pathway through siRNA-mediated knockdown of KEAP1 or via chemical induction with the NRF2-activator CDDO-Me confirmed that NRF2 is a regulator of KYNU expression in LUAD. bardoxolone methyl 194-201 NFE2 like bZIP transcription factor 2 Homo sapiens 62-66 35626147-5 2022 The siRNA-mediated knockdown of NFE2L2, the gene encoding for NRF2, or activation of the NRF2 pathway through siRNA-mediated knockdown of KEAP1 or via chemical induction with the NRF2-activator CDDO-Me confirmed that NRF2 is a regulator of KYNU expression in LUAD. bardoxolone methyl 194-201 NFE2 like bZIP transcription factor 2 Homo sapiens 89-93 35626147-5 2022 The siRNA-mediated knockdown of NFE2L2, the gene encoding for NRF2, or activation of the NRF2 pathway through siRNA-mediated knockdown of KEAP1 or via chemical induction with the NRF2-activator CDDO-Me confirmed that NRF2 is a regulator of KYNU expression in LUAD. bardoxolone methyl 194-201 kelch like ECH associated protein 1 Homo sapiens 138-143 35624848-0 2022 CDDO-Me Attenuates CA1 Neuronal Death by Facilitating RalBP1-Mediated Mitochondrial Fission and 4-HNE Efflux in the Rat Hippocampus Following Status Epilepticus. bardoxolone methyl 0-7 carbonic anhydrase 1 Rattus norvegicus 19-22 35624848-0 2022 CDDO-Me Attenuates CA1 Neuronal Death by Facilitating RalBP1-Mediated Mitochondrial Fission and 4-HNE Efflux in the Rat Hippocampus Following Status Epilepticus. bardoxolone methyl 0-7 ralA binding protein 1 Rattus norvegicus 54-60 35624848-2 2022 In the present study, we found that 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me) attenuated CA1 neuronal death and aberrant mitochondrial elongations in these neurons coupled with enhanced RalBP1 expression and reduced 4-HNE levels following status epilepticus (SE). bardoxolone methyl 101-108 carbonic anhydrase 1 Rattus norvegicus 121-124 35624848-2 2022 In the present study, we found that 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me) attenuated CA1 neuronal death and aberrant mitochondrial elongations in these neurons coupled with enhanced RalBP1 expression and reduced 4-HNE levels following status epilepticus (SE). bardoxolone methyl 101-108 ralA binding protein 1 Rattus norvegicus 218-224 35624848-4 2022 Following SE, however, cotreatment of RalBP1 siRNA diminished the effect of CDDO-Me on 4-HNE levels, mitochondrial hyperfusion in CA1 neurons, and CA1 neuronal death. bardoxolone methyl 76-83 ralA binding protein 1 Rattus norvegicus 38-44 35624848-4 2022 Following SE, however, cotreatment of RalBP1 siRNA diminished the effect of CDDO-Me on 4-HNE levels, mitochondrial hyperfusion in CA1 neurons, and CA1 neuronal death. bardoxolone methyl 76-83 carbonic anhydrase 1 Rattus norvegicus 130-133 35624848-5 2022 These findings indicate that CDDO-Me may ameliorate CA1 neuronal death by facilitating RalBP1-mediated 4-HNE efflux and mitochondrial fission following SE. bardoxolone methyl 29-36 carbonic anhydrase 1 Rattus norvegicus 52-55 35624848-5 2022 These findings indicate that CDDO-Me may ameliorate CA1 neuronal death by facilitating RalBP1-mediated 4-HNE efflux and mitochondrial fission following SE. bardoxolone methyl 29-36 ralA binding protein 1 Rattus norvegicus 87-93 35562960-0 2022 CDDO-Me Attenuates Clasmatodendrosis in CA1 Astrocyte by Inhibiting HSP25-AKT Mediated DRP1-S637 Phosphorylation in Chronic Epilepsy Rats. bardoxolone methyl 0-7 heat shock protein family B (small) member 1 Rattus norvegicus 68-73 35620281-2 2022 In this study, transgenic Sickle Cell Anemia mice (SS mice) treated with CDDO-Methyl (CDDO-Me), a potent Nrf2 activator, showed reduced progression of hemolytic anemia with aging, but surprisingly also showed reduced endothelial function. bardoxolone methyl 86-93 nuclear factor, erythroid derived 2, like 2 Mus musculus 105-109 35620281-4 2022 It is unclear what molecular mechanism underly the vascular impairment, however, our in vitro assays revealed that CDDO-Me induced the expression of the endothelin receptor (ETA and ETB) in vascular smooth muscle cells. bardoxolone methyl 115-122 endothelin receptor type B Mus musculus 182-185 35192689-7 2022 Human islets transplanted under the kidney capsule of immunocompromised mice and treated systemically with CDDO-Me, an Nrf2 activator, display increased beta-cell proliferation. bardoxolone methyl 107-114 nuclear factor, erythroid derived 2, like 2 Mus musculus 119-123 35562960-5 2022 2-Cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me; bardoxolone methyl or RTA 402) abrogated abnormal mitochondrial elongation by reducing HSP25 upregulation, AKT S473- and DRP1 S637 phosphorylations. bardoxolone methyl 65-72 heat shock protein family B (small) member 1 Rattus norvegicus 161-166 35562960-5 2022 2-Cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me; bardoxolone methyl or RTA 402) abrogated abnormal mitochondrial elongation by reducing HSP25 upregulation, AKT S473- and DRP1 S637 phosphorylations. bardoxolone methyl 65-72 AKT serine/threonine kinase 1 Rattus norvegicus 181-184 35562960-0 2022 CDDO-Me Attenuates Clasmatodendrosis in CA1 Astrocyte by Inhibiting HSP25-AKT Mediated DRP1-S637 Phosphorylation in Chronic Epilepsy Rats. bardoxolone methyl 0-7 AKT serine/threonine kinase 1 Rattus norvegicus 74-77 35562960-5 2022 2-Cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me; bardoxolone methyl or RTA 402) abrogated abnormal mitochondrial elongation by reducing HSP25 upregulation, AKT S473- and DRP1 S637 phosphorylations. bardoxolone methyl 65-72 dynamin 1-like Rattus norvegicus 195-199 35562960-0 2022 CDDO-Me Attenuates Clasmatodendrosis in CA1 Astrocyte by Inhibiting HSP25-AKT Mediated DRP1-S637 Phosphorylation in Chronic Epilepsy Rats. bardoxolone methyl 0-7 dynamin 1-like Rattus norvegicus 87-91 34973124-11 2022 The antioxidant enzymes, HO1, NQO1, GPX1, and CAT were upregulated by CDDO-Me treatment. bardoxolone methyl 70-77 heme oxygenase 1 Homo sapiens 25-28 35456042-6 2022 Exposure of worms to triterpenoid CDDO-Me, an inhibitor of human LonP1, stimulated only UPRcyt responses. bardoxolone methyl 34-41 lon peptidase 1, mitochondrial Homo sapiens 65-70 34973124-11 2022 The antioxidant enzymes, HO1, NQO1, GPX1, and CAT were upregulated by CDDO-Me treatment. bardoxolone methyl 70-77 NAD(P)H quinone dehydrogenase 1 Homo sapiens 30-34 34973124-11 2022 The antioxidant enzymes, HO1, NQO1, GPX1, and CAT were upregulated by CDDO-Me treatment. bardoxolone methyl 70-77 glutathione peroxidase 1 Homo sapiens 36-40 34973124-11 2022 The antioxidant enzymes, HO1, NQO1, GPX1, and CAT were upregulated by CDDO-Me treatment. bardoxolone methyl 70-77 catalase Homo sapiens 46-49 35265066-5 2022 We also demonstrated that addition of CDDO-Me to tri-cultures enhanced T cell-mediated reductions in CCL2, VEGF and IL-6 production in a contact-independent manner. bardoxolone methyl 38-45 C-C motif chemokine ligand 2 Homo sapiens 101-105 35265066-5 2022 We also demonstrated that addition of CDDO-Me to tri-cultures enhanced T cell-mediated reductions in CCL2, VEGF and IL-6 production in a contact-independent manner. bardoxolone methyl 38-45 vascular endothelial growth factor A Homo sapiens 107-111 35265066-5 2022 We also demonstrated that addition of CDDO-Me to tri-cultures enhanced T cell-mediated reductions in CCL2, VEGF and IL-6 production in a contact-independent manner. bardoxolone methyl 38-45 interleukin 6 Homo sapiens 116-120 35265066-7 2022 Our results indicated that CDDO-Me inhibited phosphorylation of STAT3, a known inducer of TAM activation. bardoxolone methyl 27-34 signal transducer and activator of transcription 3 Homo sapiens 64-69 35204223-8 2022 Furthermore, although activated in cells that survived standalone FUS, pre-sensitization with CDDO-me and/or nelfinavir suppressed both total and activated (phosphorylated) NF-kappaB and Akt protein levels. bardoxolone methyl 94-101 nuclear factor kappa B subunit 1 Homo sapiens 173-182 35204223-8 2022 Furthermore, although activated in cells that survived standalone FUS, pre-sensitization with CDDO-me and/or nelfinavir suppressed both total and activated (phosphorylated) NF-kappaB and Akt protein levels. bardoxolone methyl 94-101 AKT serine/threonine kinase 1 Homo sapiens 187-190 33872408-3 2021 APPROACH & RESULTS: Wild type and Nrf2 null mice were administered bardoxolone methyl (CDDO-Me), a potent activator of Nrf2 that has entered clinical development, and then subjected to two-thirds partial hepatectomy. bardoxolone methyl 67-85 nuclear factor, erythroid derived 2, like 2 Mus musculus 34-38 33922531-0 2021 CDDO-Me Attenuates Astroglial Autophagy via Nrf2-, ERK1/2-SP1- and Src-CK2-PTEN-PI3K/AKT-Mediated Signaling Pathways in the Hippocampus of Chronic Epilepsy Rats. bardoxolone methyl 0-7 NFE2 like bZIP transcription factor 2 Rattus norvegicus 44-48 33922531-0 2021 CDDO-Me Attenuates Astroglial Autophagy via Nrf2-, ERK1/2-SP1- and Src-CK2-PTEN-PI3K/AKT-Mediated Signaling Pathways in the Hippocampus of Chronic Epilepsy Rats. bardoxolone methyl 0-7 mitogen activated protein kinase 3 Rattus norvegicus 51-57 33922531-0 2021 CDDO-Me Attenuates Astroglial Autophagy via Nrf2-, ERK1/2-SP1- and Src-CK2-PTEN-PI3K/AKT-Mediated Signaling Pathways in the Hippocampus of Chronic Epilepsy Rats. bardoxolone methyl 0-7 SRC proto-oncogene, non-receptor tyrosine kinase Rattus norvegicus 67-70 33922531-0 2021 CDDO-Me Attenuates Astroglial Autophagy via Nrf2-, ERK1/2-SP1- and Src-CK2-PTEN-PI3K/AKT-Mediated Signaling Pathways in the Hippocampus of Chronic Epilepsy Rats. bardoxolone methyl 0-7 casein kinase 2 beta Rattus norvegicus 71-74 33922531-0 2021 CDDO-Me Attenuates Astroglial Autophagy via Nrf2-, ERK1/2-SP1- and Src-CK2-PTEN-PI3K/AKT-Mediated Signaling Pathways in the Hippocampus of Chronic Epilepsy Rats. bardoxolone methyl 0-7 phosphatase and tensin homolog Rattus norvegicus 75-79 33922531-0 2021 CDDO-Me Attenuates Astroglial Autophagy via Nrf2-, ERK1/2-SP1- and Src-CK2-PTEN-PI3K/AKT-Mediated Signaling Pathways in the Hippocampus of Chronic Epilepsy Rats. bardoxolone methyl 0-7 AKT serine/threonine kinase 1 Rattus norvegicus 85-88 33922531-4 2021 The 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is one of the activators for nuclear factor-erythroid 2-related factor 2 (Nrf2) that is a redox-sensitive transcription factor. bardoxolone methyl 69-76 NFE2 like bZIP transcription factor 2 Rattus norvegicus 116-159 33922531-4 2021 The 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is one of the activators for nuclear factor-erythroid 2-related factor 2 (Nrf2) that is a redox-sensitive transcription factor. bardoxolone methyl 69-76 NFE2 like bZIP transcription factor 2 Rattus norvegicus 161-165 33922531-4 2021 The 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is one of the activators for nuclear factor-erythroid 2-related factor 2 (Nrf2) that is a redox-sensitive transcription factor. bardoxolone methyl 78-85 NFE2 like bZIP transcription factor 2 Rattus norvegicus 116-159 33922531-4 2021 The 2-cyano-3,12-dioxo-oleana-1,9(11)-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is one of the activators for nuclear factor-erythroid 2-related factor 2 (Nrf2) that is a redox-sensitive transcription factor. bardoxolone methyl 78-85 NFE2 like bZIP transcription factor 2 Rattus norvegicus 161-165 33922531-6 2021 In the present study, clasmatodendritic astrocytes showed reduced Nrf2 expression and its nuclear accumulation, which were restored by CDDO-Me. bardoxolone methyl 135-142 NFE2 like bZIP transcription factor 2 Rattus norvegicus 66-70 35073378-3 2022 In this study, we aimed to disentangle the effects of these factors and clarify whether bardoxolone methyl (CDDO-Me), a novel nuclear factor erythroid 2-related factor 2 (Nrf2) activator, protects against glucose spike-induced endothelial dysfunction. bardoxolone methyl 108-115 NFE2 like bZIP transcription factor 2 Rattus norvegicus 126-169 35073378-3 2022 In this study, we aimed to disentangle the effects of these factors and clarify whether bardoxolone methyl (CDDO-Me), a novel nuclear factor erythroid 2-related factor 2 (Nrf2) activator, protects against glucose spike-induced endothelial dysfunction. bardoxolone methyl 108-115 NFE2 like bZIP transcription factor 2 Rattus norvegicus 171-175 33714136-4 2021 CDDO Methyl Ester (CDDO-ME) or ML385 treatment aggravated or alleviated rPT cells injury induced by Cd respectively, indicating that Nrf2 nucleus translocation played a negative role during Cd-induced rPT cells injury. bardoxolone methyl 0-17 NFE2 like bZIP transcription factor 2 Rattus norvegicus 133-137 33714136-4 2021 CDDO Methyl Ester (CDDO-ME) or ML385 treatment aggravated or alleviated rPT cells injury induced by Cd respectively, indicating that Nrf2 nucleus translocation played a negative role during Cd-induced rPT cells injury. bardoxolone methyl 19-26 NFE2 like bZIP transcription factor 2 Rattus norvegicus 133-137 33714136-6 2021 Dephosphorylation of AMPK induced by Cd were facilitated or restored by CDDO-ME or ML385 treatment, which confirmed AMPK is a downstream factor of Nrf2. bardoxolone methyl 72-79 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 21-25 33714136-6 2021 Dephosphorylation of AMPK induced by Cd were facilitated or restored by CDDO-ME or ML385 treatment, which confirmed AMPK is a downstream factor of Nrf2. bardoxolone methyl 72-79 protein kinase AMP-activated catalytic subunit alpha 2 Rattus norvegicus 116-120 33714136-6 2021 Dephosphorylation of AMPK induced by Cd were facilitated or restored by CDDO-ME or ML385 treatment, which confirmed AMPK is a downstream factor of Nrf2. bardoxolone methyl 72-79 NFE2 like bZIP transcription factor 2 Rattus norvegicus 147-151 33714136-7 2021 Simultaneously, CDDO-ME further enhanced Phosphorylation of mTOR and AKT which increased during Cd exposure. bardoxolone methyl 16-23 mechanistic target of rapamycin kinase Rattus norvegicus 60-64 33714136-7 2021 Simultaneously, CDDO-ME further enhanced Phosphorylation of mTOR and AKT which increased during Cd exposure. bardoxolone methyl 16-23 AKT serine/threonine kinase 1 Rattus norvegicus 69-72 33872408-3 2021 APPROACH & RESULTS: Wild type and Nrf2 null mice were administered bardoxolone methyl (CDDO-Me), a potent activator of Nrf2 that has entered clinical development, and then subjected to two-thirds partial hepatectomy. bardoxolone methyl 67-85 nuclear factor, erythroid derived 2, like 2 Mus musculus 119-123 33872408-3 2021 APPROACH & RESULTS: Wild type and Nrf2 null mice were administered bardoxolone methyl (CDDO-Me), a potent activator of Nrf2 that has entered clinical development, and then subjected to two-thirds partial hepatectomy. bardoxolone methyl 87-94 nuclear factor, erythroid derived 2, like 2 Mus musculus 34-38 33872408-3 2021 APPROACH & RESULTS: Wild type and Nrf2 null mice were administered bardoxolone methyl (CDDO-Me), a potent activator of Nrf2 that has entered clinical development, and then subjected to two-thirds partial hepatectomy. bardoxolone methyl 87-94 nuclear factor, erythroid derived 2, like 2 Mus musculus 119-123 31471671-0 2021 Beneficial effects of bardoxolone methyl, an Nrf2 activator, on crush-related acute kidney injury in rats. bardoxolone methyl 22-40 NFE2 like bZIP transcription factor 2 Rattus norvegicus 45-49 33924360-12 2021 In particular, CDDO-Me administration reduced the histopathological signs of endometriosis and inflammatory cells recruitment into the lesions, as shown by toluidine blue staining and myeloperoxidase (MPO) activity. bardoxolone methyl 15-22 myeloperoxidase Rattus norvegicus 184-199 33924360-12 2021 In particular, CDDO-Me administration reduced the histopathological signs of endometriosis and inflammatory cells recruitment into the lesions, as shown by toluidine blue staining and myeloperoxidase (MPO) activity. bardoxolone methyl 15-22 myeloperoxidase Rattus norvegicus 201-204 33924360-13 2021 CDDO-Me strongly suppressed alpha-SMA and fibronectin expression and collagen deposition, reducing endometriosis-associated fibrosis. bardoxolone methyl 0-7 fibronectin 1 Rattus norvegicus 42-53 33924360-14 2021 In conclusion, CDDO-Me treatment resulted in a coordinated and effective suppression of endometriosis by modulating the Nrf2 and NFkB pathways. bardoxolone methyl 15-22 NFE2 like bZIP transcription factor 2 Rattus norvegicus 120-124 33924360-14 2021 In conclusion, CDDO-Me treatment resulted in a coordinated and effective suppression of endometriosis by modulating the Nrf2 and NFkB pathways. bardoxolone methyl 15-22 nuclear factor kappa B subunit 1 Rattus norvegicus 129-133 33924360-7 2021 CDDO-Me displayed antioxidant activity by activating the Nfr2 pathway and the expression of antioxidant mediators such as NQO-1 and HO-1. bardoxolone methyl 0-7 NAD(P)H quinone dehydrogenase 1 Rattus norvegicus 122-127 33924360-7 2021 CDDO-Me displayed antioxidant activity by activating the Nfr2 pathway and the expression of antioxidant mediators such as NQO-1 and HO-1. bardoxolone methyl 0-7 heme oxygenase 1 Rattus norvegicus 132-136 33924360-9 2021 CDDO-Me also showed anti-inflammatory activity by decreasing the expression of pro-inflammatory cytokines in peritoneal fluids and NFkB activation. bardoxolone methyl 0-7 nuclear factor kappa B subunit 1 Rattus norvegicus 131-135 31471671-1 2021 PURPOSE: The purpose of this study was to investigate the effects of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, on acute kidney injury in a rat model of crush syndrome model. bardoxolone methyl 69-87 NFE2 like bZIP transcription factor 2 Rattus norvegicus 96-139 31471671-1 2021 PURPOSE: The purpose of this study was to investigate the effects of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, on acute kidney injury in a rat model of crush syndrome model. bardoxolone methyl 69-87 NFE2 like bZIP transcription factor 2 Rattus norvegicus 141-145 31471671-1 2021 PURPOSE: The purpose of this study was to investigate the effects of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, on acute kidney injury in a rat model of crush syndrome model. bardoxolone methyl 89-91 NFE2 like bZIP transcription factor 2 Rattus norvegicus 96-139 31471671-1 2021 PURPOSE: The purpose of this study was to investigate the effects of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator, on acute kidney injury in a rat model of crush syndrome model. bardoxolone methyl 89-91 NFE2 like bZIP transcription factor 2 Rattus norvegicus 141-145 33096818-0 2020 CDDO-Me Distinctly Regulates Regional Specific Astroglial Responses to Status Epilepticus via ERK1/2-Nrf2, PTEN-PI3K-AKT and NFkappaB Signaling Pathways. bardoxolone methyl 0-7 mitogen activated protein kinase 3 Rattus norvegicus 94-100 33096818-0 2020 CDDO-Me Distinctly Regulates Regional Specific Astroglial Responses to Status Epilepticus via ERK1/2-Nrf2, PTEN-PI3K-AKT and NFkappaB Signaling Pathways. bardoxolone methyl 0-7 NFE2 like bZIP transcription factor 2 Rattus norvegicus 101-105 33096818-0 2020 CDDO-Me Distinctly Regulates Regional Specific Astroglial Responses to Status Epilepticus via ERK1/2-Nrf2, PTEN-PI3K-AKT and NFkappaB Signaling Pathways. bardoxolone methyl 0-7 phosphatase and tensin homolog Rattus norvegicus 107-111 33096818-0 2020 CDDO-Me Distinctly Regulates Regional Specific Astroglial Responses to Status Epilepticus via ERK1/2-Nrf2, PTEN-PI3K-AKT and NFkappaB Signaling Pathways. bardoxolone methyl 0-7 AKT serine/threonine kinase 1 Rattus norvegicus 117-120 33096818-3 2020 Furthermore, CDDO-Me has antioxidant properties, since it activates nuclear factor-erythroid 2-related factor 2 (Nrf2), which is a key player of redox homeostasis. bardoxolone methyl 13-20 NFE2 like bZIP transcription factor 2 Rattus norvegicus 68-111 33096818-3 2020 Furthermore, CDDO-Me has antioxidant properties, since it activates nuclear factor-erythroid 2-related factor 2 (Nrf2), which is a key player of redox homeostasis. bardoxolone methyl 13-20 NFE2 like bZIP transcription factor 2 Rattus norvegicus 113-117 33096818-5 2020 Under physiological conditions, CDDO-Me increased Nrf2 expression in the hippocampus without altering activities (phosphorylations) of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), phosphatidylinositol-3-kinase (PI3K), and AKT. bardoxolone methyl 32-39 NFE2 like bZIP transcription factor 2 Rattus norvegicus 50-54 33096818-5 2020 Under physiological conditions, CDDO-Me increased Nrf2 expression in the hippocampus without altering activities (phosphorylations) of phosphatase and tensin homolog deleted on chromosome 10 (PTEN), phosphatidylinositol-3-kinase (PI3K), and AKT. bardoxolone methyl 32-39 AKT serine/threonine kinase 1 Rattus norvegicus 241-244 33096818-7 2020 However, CDDO-Me ameliorated reduced astroglial Nrf2 expression in the CA1 region and the molecular layer of the dentate gyrus (ML), and attenuated reactive astrogliosis and ML astroglial apoptosis following SE. bardoxolone methyl 9-16 NFE2 like bZIP transcription factor 2 Rattus norvegicus 48-52 33096818-7 2020 However, CDDO-Me ameliorated reduced astroglial Nrf2 expression in the CA1 region and the molecular layer of the dentate gyrus (ML), and attenuated reactive astrogliosis and ML astroglial apoptosis following SE. bardoxolone methyl 9-16 carbonic anhydrase 1 Rattus norvegicus 71-74 33096818-8 2020 In CA1 astrocytes, CDDO-Me inhibited the PI3K/AKT pathway by activating PTEN. bardoxolone methyl 19-26 carbonic anhydrase 1 Rattus norvegicus 3-6 33096818-8 2020 In CA1 astrocytes, CDDO-Me inhibited the PI3K/AKT pathway by activating PTEN. bardoxolone methyl 19-26 AKT serine/threonine kinase 1 Rattus norvegicus 46-49 33096818-8 2020 In CA1 astrocytes, CDDO-Me inhibited the PI3K/AKT pathway by activating PTEN. bardoxolone methyl 19-26 phosphatase and tensin homolog Rattus norvegicus 72-76 33096818-9 2020 In contrast, CDDO-ME resulted in extracellular signal-related kinases 1/2 (ERK1/2)-mediated Nrf2 upregulation in ML astrocytes. bardoxolone methyl 13-20 mitogen activated protein kinase 3 Rattus norvegicus 75-81 33096818-9 2020 In contrast, CDDO-ME resulted in extracellular signal-related kinases 1/2 (ERK1/2)-mediated Nrf2 upregulation in ML astrocytes. bardoxolone methyl 13-20 NFE2 like bZIP transcription factor 2 Rattus norvegicus 92-96 33096818-10 2020 Furthermore, CDDO-Me decreased nuclear factor-kappaB (NFkappaB) phosphorylation in both CA1 and ML astrocytes. bardoxolone methyl 13-20 carbonic anhydrase 1 Rattus norvegicus 88-91 33096818-11 2020 Therefore, our findings suggest that CDDO-Me may attenuate SE-induced reactive astrogliosis and astroglial apoptosis via regulation of ERK1/2-Nrf2, PTEN-PI3K-AKT, and NFkappaB signaling pathways. bardoxolone methyl 37-44 mitogen activated protein kinase 3 Rattus norvegicus 135-141 33096818-11 2020 Therefore, our findings suggest that CDDO-Me may attenuate SE-induced reactive astrogliosis and astroglial apoptosis via regulation of ERK1/2-Nrf2, PTEN-PI3K-AKT, and NFkappaB signaling pathways. bardoxolone methyl 37-44 NFE2 like bZIP transcription factor 2 Rattus norvegicus 142-146 33096818-11 2020 Therefore, our findings suggest that CDDO-Me may attenuate SE-induced reactive astrogliosis and astroglial apoptosis via regulation of ERK1/2-Nrf2, PTEN-PI3K-AKT, and NFkappaB signaling pathways. bardoxolone methyl 37-44 phosphatase and tensin homolog Rattus norvegicus 148-152 33096818-11 2020 Therefore, our findings suggest that CDDO-Me may attenuate SE-induced reactive astrogliosis and astroglial apoptosis via regulation of ERK1/2-Nrf2, PTEN-PI3K-AKT, and NFkappaB signaling pathways. bardoxolone methyl 37-44 AKT serine/threonine kinase 1 Rattus norvegicus 158-161 32300202-0 2020 CDDO-Me Alters the Tumor Microenvironment in Estrogen Receptor Negative Breast Cancer. bardoxolone methyl 0-7 estrogen receptor 1 (alpha) Mus musculus 45-62 32802194-20 2020 Quantitative evaluation of the anti-inflammatory effect of CDDO-Me on neuroinflammation was successfully monitored with TSPO-targeting [18F]CB251-PET/MR and BLI. bardoxolone methyl 59-66 translocator protein Mus musculus 120-124 32766432-6 2020 Protein levels of multidrug resistance-associated protein 2, multidrug resistance-associated protein 3, and sodium-taurocholate cotransporter were higher in the IR90 group relative to those in the sham or IR60 groups, wherein the difference was notable only when BARD was administered. bardoxolone methyl 263-267 ATP binding cassette subfamily C member 2 Rattus norvegicus 18-59 32484788-2 2020 Bardoxolone-methyl (BARD) is a NF-E2 p45-related factor 2 (Nrf2) agonist. bardoxolone methyl 0-18 NFE2 like bZIP transcription factor 2 Homo sapiens 31-57 32484788-2 2020 Bardoxolone-methyl (BARD) is a NF-E2 p45-related factor 2 (Nrf2) agonist. bardoxolone methyl 0-18 NFE2 like bZIP transcription factor 2 Homo sapiens 59-63 32484788-2 2020 Bardoxolone-methyl (BARD) is a NF-E2 p45-related factor 2 (Nrf2) agonist. bardoxolone methyl 20-24 NFE2 like bZIP transcription factor 2 Homo sapiens 31-57 32484788-2 2020 Bardoxolone-methyl (BARD) is a NF-E2 p45-related factor 2 (Nrf2) agonist. bardoxolone methyl 20-24 NFE2 like bZIP transcription factor 2 Homo sapiens 59-63 32370011-0 2020 CDDO-Me Inhibits Microglial Activation and Monocyte Infiltration by Abrogating NFkappaB- and p38 MAPK-Mediated Signaling Pathways Following Status Epilepticus. bardoxolone methyl 0-7 nuclear factor kappa B subunit 1 Homo sapiens 79-87 32370011-0 2020 CDDO-Me Inhibits Microglial Activation and Monocyte Infiltration by Abrogating NFkappaB- and p38 MAPK-Mediated Signaling Pathways Following Status Epilepticus. bardoxolone methyl 0-7 mitogen-activated protein kinase 14 Homo sapiens 93-101 32370011-3 2020 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2), which regulates intracellular redox homeostasis. bardoxolone methyl 0-58 NFE2 like bZIP transcription factor 2 Homo sapiens 97-140 32370011-3 2020 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2), which regulates intracellular redox homeostasis. bardoxolone methyl 0-58 NFE2 like bZIP transcription factor 2 Homo sapiens 142-146 32370011-3 2020 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2), which regulates intracellular redox homeostasis. bardoxolone methyl 60-67 NFE2 like bZIP transcription factor 2 Homo sapiens 97-140 32370011-3 2020 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2), which regulates intracellular redox homeostasis. bardoxolone methyl 60-67 NFE2 like bZIP transcription factor 2 Homo sapiens 142-146 32370011-3 2020 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2), which regulates intracellular redox homeostasis. bardoxolone methyl 69-76 NFE2 like bZIP transcription factor 2 Homo sapiens 97-140 32370011-3 2020 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid methyl ester (CDDO-Me; RTA 402) is an activator of nuclear factor-erythroid 2-related factor 2 (Nrf2), which regulates intracellular redox homeostasis. bardoxolone methyl 69-76 NFE2 like bZIP transcription factor 2 Homo sapiens 142-146 32370011-5 2020 In the present study, CDDO-Me ameliorated monocyte infiltration without vasogenic edema formation in the frontoparietal cortex (FPC) following SE, accompanied by abrogating monocyte chemotactic protein-1 (MCP-1)/tumor necrosis factor-alpha (TNF-alpha) expressions and p38 mitogen-activated protein kinase (p38 MAPK) phosphorylation. bardoxolone methyl 22-29 C-C motif chemokine ligand 2 Homo sapiens 173-203 32370011-5 2020 In the present study, CDDO-Me ameliorated monocyte infiltration without vasogenic edema formation in the frontoparietal cortex (FPC) following SE, accompanied by abrogating monocyte chemotactic protein-1 (MCP-1)/tumor necrosis factor-alpha (TNF-alpha) expressions and p38 mitogen-activated protein kinase (p38 MAPK) phosphorylation. bardoxolone methyl 22-29 C-C motif chemokine ligand 2 Homo sapiens 205-210 32370011-5 2020 In the present study, CDDO-Me ameliorated monocyte infiltration without vasogenic edema formation in the frontoparietal cortex (FPC) following SE, accompanied by abrogating monocyte chemotactic protein-1 (MCP-1)/tumor necrosis factor-alpha (TNF-alpha) expressions and p38 mitogen-activated protein kinase (p38 MAPK) phosphorylation. bardoxolone methyl 22-29 tumor necrosis factor Homo sapiens 212-239 32370011-5 2020 In the present study, CDDO-Me ameliorated monocyte infiltration without vasogenic edema formation in the frontoparietal cortex (FPC) following SE, accompanied by abrogating monocyte chemotactic protein-1 (MCP-1)/tumor necrosis factor-alpha (TNF-alpha) expressions and p38 mitogen-activated protein kinase (p38 MAPK) phosphorylation. bardoxolone methyl 22-29 tumor necrosis factor Homo sapiens 241-250 32370011-5 2020 In the present study, CDDO-Me ameliorated monocyte infiltration without vasogenic edema formation in the frontoparietal cortex (FPC) following SE, accompanied by abrogating monocyte chemotactic protein-1 (MCP-1)/tumor necrosis factor-alpha (TNF-alpha) expressions and p38 mitogen-activated protein kinase (p38 MAPK) phosphorylation. bardoxolone methyl 22-29 mitogen-activated protein kinase 14 Homo sapiens 268-304 32370011-5 2020 In the present study, CDDO-Me ameliorated monocyte infiltration without vasogenic edema formation in the frontoparietal cortex (FPC) following SE, accompanied by abrogating monocyte chemotactic protein-1 (MCP-1)/tumor necrosis factor-alpha (TNF-alpha) expressions and p38 mitogen-activated protein kinase (p38 MAPK) phosphorylation. bardoxolone methyl 22-29 mitogen-activated protein kinase 14 Homo sapiens 306-314 32370011-6 2020 Furthermore, CDDO-Me inhibited nuclear factor-kappaB (NFkappaB)-S276 phosphorylation and microglial transformation, independent of Nrf2 expression. bardoxolone methyl 13-20 nuclear factor kappa B subunit 1 Homo sapiens 54-62 32370011-8 2020 Therefore, these findings suggest, for the first time, that CDDO-Me may attenuate microglia/monocyte-mediated neuroinflammation via modulating NFkappaB- and p38 MAPK-MCP-1 signaling pathways following SE. bardoxolone methyl 60-67 nuclear factor kappa B subunit 1 Homo sapiens 143-151 32370011-8 2020 Therefore, these findings suggest, for the first time, that CDDO-Me may attenuate microglia/monocyte-mediated neuroinflammation via modulating NFkappaB- and p38 MAPK-MCP-1 signaling pathways following SE. bardoxolone methyl 60-67 mitogen-activated protein kinase 14 Homo sapiens 157-165 32370011-8 2020 Therefore, these findings suggest, for the first time, that CDDO-Me may attenuate microglia/monocyte-mediated neuroinflammation via modulating NFkappaB- and p38 MAPK-MCP-1 signaling pathways following SE. bardoxolone methyl 60-67 C-C motif chemokine ligand 2 Homo sapiens 166-171 32787104-1 2020 Synthetic triterpenoids including CDDO, its methyl ester (CDDO-Me, bardoxolone methyl), and its imidazolide (CDDO-Im) enhance Nrf2-mediated anti-oxidant and anti-inflammatory activity in many diseases by reacting with thiols on the adaptor protein, Keap1. bardoxolone methyl 58-65 NFE2 like bZIP transcription factor 2 Homo sapiens 126-130 32556916-1 2020 Bardoxolone methyl (Bard), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator regulates multiple oxidative and inflammatory diseases. bardoxolone methyl 0-18 NFE2 like bZIP transcription factor 2 Rattus norvegicus 29-72 32556916-1 2020 Bardoxolone methyl (Bard), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator regulates multiple oxidative and inflammatory diseases. bardoxolone methyl 0-18 NFE2 like bZIP transcription factor 2 Rattus norvegicus 74-78 32556916-1 2020 Bardoxolone methyl (Bard), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator regulates multiple oxidative and inflammatory diseases. bardoxolone methyl 0-4 NFE2 like bZIP transcription factor 2 Rattus norvegicus 29-72 32556916-1 2020 Bardoxolone methyl (Bard), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator regulates multiple oxidative and inflammatory diseases. bardoxolone methyl 0-4 NFE2 like bZIP transcription factor 2 Rattus norvegicus 74-78 32040844-4 2020 Direct binding and inhibition of PKM2 activity by CDDO-Me was demonstrated by pull-down and activity assays. bardoxolone methyl 50-57 pyruvate kinase M1/2 Homo sapiens 33-37 32040844-8 2020 CDDO-Me binds directly and specifically to recombinant PKM2, leading to a reduction of its catalytic activity. bardoxolone methyl 0-13 pyruvate kinase M1/2 Homo sapiens 55-59 32300202-5 2020 We demonstrate that CDDO-Me significantly attenuates IL-10 and VEGF expression but stimulates TNF production, and reduces surface expression of CD206 and CD115, markers of immunosuppressive TAMs. bardoxolone methyl 20-27 interleukin 10 Mus musculus 53-58 32300202-5 2020 We demonstrate that CDDO-Me significantly attenuates IL-10 and VEGF expression but stimulates TNF production, and reduces surface expression of CD206 and CD115, markers of immunosuppressive TAMs. bardoxolone methyl 20-27 vascular endothelial growth factor A Mus musculus 63-67 32300202-5 2020 We demonstrate that CDDO-Me significantly attenuates IL-10 and VEGF expression but stimulates TNF production, and reduces surface expression of CD206 and CD115, markers of immunosuppressive TAMs. bardoxolone methyl 20-27 tumor necrosis factor Mus musculus 94-97 32300202-5 2020 We demonstrate that CDDO-Me significantly attenuates IL-10 and VEGF expression but stimulates TNF production, and reduces surface expression of CD206 and CD115, markers of immunosuppressive TAMs. bardoxolone methyl 20-27 mannose receptor, C type 1 Mus musculus 144-149 32300202-5 2020 We demonstrate that CDDO-Me significantly attenuates IL-10 and VEGF expression but stimulates TNF production, and reduces surface expression of CD206 and CD115, markers of immunosuppressive TAMs. bardoxolone methyl 20-27 colony stimulating factor 1 receptor Mus musculus 154-159 32300202-6 2020 CDDO-Me treatment redirects the TAM transcriptional profile, inducing signaling pathways associated with immune stimulation, and inhibits TAM tumor infiltration, consistent with decreased expression of CCL2. bardoxolone methyl 0-7 chemokine (C-C motif) ligand 2 Mus musculus 202-206 32300202-7 2020 In CDDO-Me-treated mice, both the absolute number and proportion of splenic CD4+ T cells were reduced, while the proportion of CD8+ T cells was significantly increased in both tumors and spleen. bardoxolone methyl 3-10 CD4 antigen Mus musculus 76-79 32300202-8 2020 Moreover, mice fed CDDO-Me demonstrated significant reductions in numbers of CD4+ Foxp3+ regulatory T cells within tumors. bardoxolone methyl 19-26 CD4 antigen Mus musculus 77-80 32300202-8 2020 Moreover, mice fed CDDO-Me demonstrated significant reductions in numbers of CD4+ Foxp3+ regulatory T cells within tumors. bardoxolone methyl 19-26 forkhead box P3 Mus musculus 82-87 31541463-6 2020 As a downstream gene of Nrf2, heme oxygenase-1 expression decreased significantly with the cotreatment of erastin and APAP, which could be rescued by BM. bardoxolone methyl 150-152 NFE2 like bZIP transcription factor 2 Homo sapiens 24-28 32032580-13 2020 Moreover, bardoxolone methyl (CDDO-Me), a potent Nrf2 activator, ameliorated BLM-induced skin fibrosis after topical administration. bardoxolone methyl 10-28 nuclear factor, erythroid derived 2, like 2 Mus musculus 49-53 32032580-13 2020 Moreover, bardoxolone methyl (CDDO-Me), a potent Nrf2 activator, ameliorated BLM-induced skin fibrosis after topical administration. bardoxolone methyl 30-37 nuclear factor, erythroid derived 2, like 2 Mus musculus 49-53 31541463-6 2020 As a downstream gene of Nrf2, heme oxygenase-1 expression decreased significantly with the cotreatment of erastin and APAP, which could be rescued by BM. bardoxolone methyl 150-152 heme oxygenase 1 Homo sapiens 30-46 31421166-3 2019 Here we report the identification of the compound Bardoxolone methyl (CDDO-me) as a potent inhibitor of the Hrd1 ubiquitin ligase-mediated ERAD, which possesses a broad-spectrum activity against both DENV and ZIKV. bardoxolone methyl 50-68 synoviolin 1 Homo sapiens 108-112 32015160-0 2020 CDDO-Me elicits anti-breast cancer activity by targeting LRP6 and FZD7 receptor complex. bardoxolone methyl 0-15 LDL receptor related protein 6 Homo sapiens 57-61 32015160-0 2020 CDDO-Me elicits anti-breast cancer activity by targeting LRP6 and FZD7 receptor complex. bardoxolone methyl 0-15 frizzled class receptor 7 Homo sapiens 66-70 32015160-3 2020 In this study, we found that CDDO-Me could inhibit Wnt/beta-catenin signaling mainly through targeting the LRP6 and FZD7 receptor complex. bardoxolone methyl 29-42 catenin beta 1 Homo sapiens 55-67 32015160-3 2020 In this study, we found that CDDO-Me could inhibit Wnt/beta-catenin signaling mainly through targeting the LRP6 and FZD7 receptor complex. bardoxolone methyl 29-42 LDL receptor related protein 6 Homo sapiens 107-111 32015160-3 2020 In this study, we found that CDDO-Me could inhibit Wnt/beta-catenin signaling mainly through targeting the LRP6 and FZD7 receptor complex. bardoxolone methyl 29-42 frizzled class receptor 7 Homo sapiens 116-120 32015160-5 2020 We further showed CDDO-Me mediated degradation of FZD7 in a LRP6 ectodomain-dependent manner. bardoxolone methyl 18-25 frizzled class receptor 7 Homo sapiens 50-54 32015160-5 2020 We further showed CDDO-Me mediated degradation of FZD7 in a LRP6 ectodomain-dependent manner. bardoxolone methyl 18-25 LDL receptor related protein 6 Homo sapiens 60-64 32015160-6 2020 In breast cancer cells, treatment with CDDO-Me increased the degradation of LRP6 and Fzd7, reduced the levels of phosphorylated DVL2 and active beta-catenin, resulting in downregulation of Wnt target genes and some cancer stem cell (CSC) marker genes. bardoxolone methyl 39-46 LDL receptor related protein 6 Homo sapiens 76-80 32015160-6 2020 In breast cancer cells, treatment with CDDO-Me increased the degradation of LRP6 and Fzd7, reduced the levels of phosphorylated DVL2 and active beta-catenin, resulting in downregulation of Wnt target genes and some cancer stem cell (CSC) marker genes. bardoxolone methyl 39-46 frizzled class receptor 7 Homo sapiens 85-89 32015160-6 2020 In breast cancer cells, treatment with CDDO-Me increased the degradation of LRP6 and Fzd7, reduced the levels of phosphorylated DVL2 and active beta-catenin, resulting in downregulation of Wnt target genes and some cancer stem cell (CSC) marker genes. bardoxolone methyl 39-46 dishevelled segment polarity protein 2 Homo sapiens 128-132 32015160-6 2020 In breast cancer cells, treatment with CDDO-Me increased the degradation of LRP6 and Fzd7, reduced the levels of phosphorylated DVL2 and active beta-catenin, resulting in downregulation of Wnt target genes and some cancer stem cell (CSC) marker genes. bardoxolone methyl 39-46 catenin beta 1 Homo sapiens 144-156 32015160-7 2020 In a murine xenograft bearing MMTV-Wnt1-driven mammary tumor, administration of CDDO-Me significantly inhibited tumor growth, accompanied by reduced expression of phosphorylated and total LRP6, phosphorylated and unphosphorylated DVL2, active beta-catenin, several Wnt target genes and CSC marker genes. bardoxolone methyl 80-87 LDL receptor related protein 6 Homo sapiens 188-192 32015160-7 2020 In a murine xenograft bearing MMTV-Wnt1-driven mammary tumor, administration of CDDO-Me significantly inhibited tumor growth, accompanied by reduced expression of phosphorylated and total LRP6, phosphorylated and unphosphorylated DVL2, active beta-catenin, several Wnt target genes and CSC marker genes. bardoxolone methyl 80-87 dishevelled segment polarity protein 2 Homo sapiens 230-234 32015160-7 2020 In a murine xenograft bearing MMTV-Wnt1-driven mammary tumor, administration of CDDO-Me significantly inhibited tumor growth, accompanied by reduced expression of phosphorylated and total LRP6, phosphorylated and unphosphorylated DVL2, active beta-catenin, several Wnt target genes and CSC marker genes. bardoxolone methyl 80-87 catenin beta 1 Homo sapiens 243-255 32015160-8 2020 Collectively, our results demonstrated that CDDO-Me is a potent Wnt/beta-catenin signaling inhibitor and so may be a promising therapeutic agent against breast cancer. bardoxolone methyl 44-51 catenin beta 1 Homo sapiens 68-80 31940946-9 2020 The anti-oxidant N-acetyl cysteine (NAC) could overcome this AR-suppressive effect of CDDO-Me. Co-exposure of PC cells to CDDO-Me enhanced the efficacy of a clinically approved anti-androgen, enzalutamide (ENZ), as evident by decreased cell-viability along with migration and colony forming ability of PC cells. bardoxolone methyl 86-93 androgen receptor Homo sapiens 61-63 31940946-0 2020 Bardoxolone-Methyl (CDDO-Me) Suppresses Androgen Receptor and Its Splice-Variant AR-V7 and Enhances Efficacy of Enzalutamide in Prostate Cancer Cells. bardoxolone methyl 0-28 androgen receptor Homo sapiens 40-57 31940946-4 2020 We examined whether a triterpenoid antioxidant drug, Bardoxolone-methyl, known as CDDO-Me or RTA 402, can decrease AR-FL and AR-V7 expression in PC cells. bardoxolone methyl 53-71 androgen receptor Homo sapiens 115-120 31940946-4 2020 We examined whether a triterpenoid antioxidant drug, Bardoxolone-methyl, known as CDDO-Me or RTA 402, can decrease AR-FL and AR-V7 expression in PC cells. bardoxolone methyl 53-71 androgen receptor Homo sapiens 115-117 31940946-4 2020 We examined whether a triterpenoid antioxidant drug, Bardoxolone-methyl, known as CDDO-Me or RTA 402, can decrease AR-FL and AR-V7 expression in PC cells. bardoxolone methyl 82-96 androgen receptor Homo sapiens 115-120 31940946-4 2020 We examined whether a triterpenoid antioxidant drug, Bardoxolone-methyl, known as CDDO-Me or RTA 402, can decrease AR-FL and AR-V7 expression in PC cells. bardoxolone methyl 82-96 androgen receptor Homo sapiens 115-117 31940946-5 2020 Nanomolar (nM) concentrations of CDDO-Me rapidly downregulated AR-FL in LNCaP and C4-2B cells, and both AR-FL and AR-V7 in CWR22Rv1 (22Rv1) cells. bardoxolone methyl 33-48 androgen receptor Homo sapiens 63-68 31940946-5 2020 Nanomolar (nM) concentrations of CDDO-Me rapidly downregulated AR-FL in LNCaP and C4-2B cells, and both AR-FL and AR-V7 in CWR22Rv1 (22Rv1) cells. bardoxolone methyl 33-48 androgen receptor Homo sapiens 63-65 31940946-6 2020 The AR-suppressive effect of CDDO-Me was evident at both the mRNA and protein levels. bardoxolone methyl 29-36 androgen receptor Homo sapiens 4-6 31940946-9 2020 The anti-oxidant N-acetyl cysteine (NAC) could overcome this AR-suppressive effect of CDDO-Me. Co-exposure of PC cells to CDDO-Me enhanced the efficacy of a clinically approved anti-androgen, enzalutamide (ENZ), as evident by decreased cell-viability along with migration and colony forming ability of PC cells. bardoxolone methyl 86-94 androgen receptor Homo sapiens 61-63 31601682-4 2019 Here, we investigated the beneficial hemodynamic effects of bardoxolone methyl (2-Cyano-3,12-dioxooleana-1,9(11)-dien-28-oic acid methyl ester, CDDO-Me), a pharmacological activator of Nrf2, in a rodent model of CHF. bardoxolone methyl 80-142 NFE2 like bZIP transcription factor 2 Rattus norvegicus 185-189 31601682-9 2019 Molecular studies revealed that CDDO-Me-induced cardiac functional improvement was attributed to an increase of both Nrf2 transcription and translation, and a decrease of oxidative stress in the noninfarcted areas of the heart. bardoxolone methyl 32-39 NFE2 like bZIP transcription factor 2 Rattus norvegicus 117-121 31601682-10 2019 Furthermore, CDDO-Me reduced NF-kappaB binding and increased Nrf2 binding to the CREB-binding protein, which may contribute to the selective increase of Nrf2 downstream targets, including NADPH Oxidase Quinone 1, Heme Oxygenase 1, Catalase, and Glutamate-Cysteine Ligase Catalytic Subunit, and the attenuation of myocardial inflammation in CHF rats. bardoxolone methyl 13-20 NFE2 like bZIP transcription factor 2 Rattus norvegicus 61-65 31601682-10 2019 Furthermore, CDDO-Me reduced NF-kappaB binding and increased Nrf2 binding to the CREB-binding protein, which may contribute to the selective increase of Nrf2 downstream targets, including NADPH Oxidase Quinone 1, Heme Oxygenase 1, Catalase, and Glutamate-Cysteine Ligase Catalytic Subunit, and the attenuation of myocardial inflammation in CHF rats. bardoxolone methyl 13-20 CREB binding protein Rattus norvegicus 81-101 31601682-10 2019 Furthermore, CDDO-Me reduced NF-kappaB binding and increased Nrf2 binding to the CREB-binding protein, which may contribute to the selective increase of Nrf2 downstream targets, including NADPH Oxidase Quinone 1, Heme Oxygenase 1, Catalase, and Glutamate-Cysteine Ligase Catalytic Subunit, and the attenuation of myocardial inflammation in CHF rats. bardoxolone methyl 13-20 NFE2 like bZIP transcription factor 2 Rattus norvegicus 153-157 31601682-10 2019 Furthermore, CDDO-Me reduced NF-kappaB binding and increased Nrf2 binding to the CREB-binding protein, which may contribute to the selective increase of Nrf2 downstream targets, including NADPH Oxidase Quinone 1, Heme Oxygenase 1, Catalase, and Glutamate-Cysteine Ligase Catalytic Subunit, and the attenuation of myocardial inflammation in CHF rats. bardoxolone methyl 13-20 heme oxygenase 1 Rattus norvegicus 213-229 31601682-10 2019 Furthermore, CDDO-Me reduced NF-kappaB binding and increased Nrf2 binding to the CREB-binding protein, which may contribute to the selective increase of Nrf2 downstream targets, including NADPH Oxidase Quinone 1, Heme Oxygenase 1, Catalase, and Glutamate-Cysteine Ligase Catalytic Subunit, and the attenuation of myocardial inflammation in CHF rats. bardoxolone methyl 13-20 catalase Rattus norvegicus 231-239 31601682-10 2019 Furthermore, CDDO-Me reduced NF-kappaB binding and increased Nrf2 binding to the CREB-binding protein, which may contribute to the selective increase of Nrf2 downstream targets, including NADPH Oxidase Quinone 1, Heme Oxygenase 1, Catalase, and Glutamate-Cysteine Ligase Catalytic Subunit, and the attenuation of myocardial inflammation in CHF rats. bardoxolone methyl 13-20 glutamate-cysteine ligase, catalytic subunit Rattus norvegicus 245-288 31421166-8 2019 Inhibition of grp94 may contribute to the antiviral activity of CDDO-me. bardoxolone methyl 64-71 heat shock protein 90 beta family member 1 Homo sapiens 14-19 31421166-3 2019 Here we report the identification of the compound Bardoxolone methyl (CDDO-me) as a potent inhibitor of the Hrd1 ubiquitin ligase-mediated ERAD, which possesses a broad-spectrum activity against both DENV and ZIKV. bardoxolone methyl 70-77 synoviolin 1 Homo sapiens 108-112 31421166-4 2019 Cellular thermal shift assay (CETSA) suggested that CDDO-me binds to grp94, a key component of the Hrd1 pathway, at a low nanomolar concentration, whereas interaction was not detected with its paralog Hsp90. bardoxolone methyl 52-59 heat shock protein 90 beta family member 1 Homo sapiens 69-74 31421166-4 2019 Cellular thermal shift assay (CETSA) suggested that CDDO-me binds to grp94, a key component of the Hrd1 pathway, at a low nanomolar concentration, whereas interaction was not detected with its paralog Hsp90. bardoxolone methyl 52-59 synoviolin 1 Homo sapiens 99-103 31543152-0 2019 Increased albuminuria in bardoxolone methyl-treated type 2 diabetes patients: mere reflection of eGFR improvement? bardoxolone methyl 25-43 epidermal growth factor receptor Homo sapiens 97-101 31543152-2 2019 Because bardoxolone methyl also increases albuminuria, Rossing and colleagues performed a retrospective analysis of the Bardoxolone Methyl Evaluation in Patients With Chronic Kidney Disease and Type 2 Diabetes (BEACON) study and attributed increased albuminuria to increased estimated glomerular filtration rate. bardoxolone methyl 8-26 ubiquitin like 5 Homo sapiens 211-217 31543152-2 2019 Because bardoxolone methyl also increases albuminuria, Rossing and colleagues performed a retrospective analysis of the Bardoxolone Methyl Evaluation in Patients With Chronic Kidney Disease and Type 2 Diabetes (BEACON) study and attributed increased albuminuria to increased estimated glomerular filtration rate. bardoxolone methyl 120-138 ubiquitin like 5 Homo sapiens 211-217 31574956-5 2019 CDDO-Me also abolished NF-kappaB threonine 435 phosphorylation in endothelial cells and TNF-alpha-mediated-phosphatidylinositol-3-kinase (PI3K)/AKT/endothelial nitric oxide synthase (eNOS) signaling cascades, which trigger vasogenic edema following SE. bardoxolone methyl 0-7 tumor necrosis factor Rattus norvegicus 88-97 31356822-4 2019 In this study, cell viability, reactive oxygen species (ROS) production, and several proteins" expressions of human embryonic kidney 293T (HEK293T) cells in response to UA, OTA, and/or Lonp1 inhibitor CDDO-me treatment were detected to reveal the protective mechanism of UA against OTA-induced renal cytotoxicity. bardoxolone methyl 201-208 lon peptidase 1, mitochondrial Homo sapiens 185-190 31356822-7 2019 The protein expressions of Lonp1, Aco2 and Hsp75 were inhibited by the addition of CDDO-me. bardoxolone methyl 83-90 lon peptidase 1, mitochondrial Homo sapiens 27-32 31356822-7 2019 The protein expressions of Lonp1, Aco2 and Hsp75 were inhibited by the addition of CDDO-me. bardoxolone methyl 83-90 aconitase 2 Homo sapiens 34-38 31356822-7 2019 The protein expressions of Lonp1, Aco2 and Hsp75 were inhibited by the addition of CDDO-me. bardoxolone methyl 83-90 TNF receptor associated protein 1 Homo sapiens 43-48 31574956-0 2019 CDDO-Me Attenuates Vasogenic Edema and Astroglial Death by Regulating NF-kappaB p65 Phosphorylations and Nrf2 Expression Following Status Epilepticus. bardoxolone methyl 0-7 NFE2 like bZIP transcription factor 2 Rattus norvegicus 105-109 31574956-4 2019 CDDO-Me abrogated tumor necrosis factor-alpha (TNF-alpha) synthesis in activated microglia by inhibiting nuclear factor-kappaB (NF-kappaB) p65 serine 276 phosphorylation. bardoxolone methyl 0-7 tumor necrosis factor Rattus norvegicus 47-56 31574956-5 2019 CDDO-Me also abolished NF-kappaB threonine 435 phosphorylation in endothelial cells and TNF-alpha-mediated-phosphatidylinositol-3-kinase (PI3K)/AKT/endothelial nitric oxide synthase (eNOS) signaling cascades, which trigger vasogenic edema following SE. bardoxolone methyl 0-7 AKT serine/threonine kinase 1 Rattus norvegicus 144-147 31574956-5 2019 CDDO-Me also abolished NF-kappaB threonine 435 phosphorylation in endothelial cells and TNF-alpha-mediated-phosphatidylinositol-3-kinase (PI3K)/AKT/endothelial nitric oxide synthase (eNOS) signaling cascades, which trigger vasogenic edema following SE. bardoxolone methyl 0-7 nitric oxide synthase 3 Rattus norvegicus 183-187 31574956-4 2019 CDDO-Me abrogated tumor necrosis factor-alpha (TNF-alpha) synthesis in activated microglia by inhibiting nuclear factor-kappaB (NF-kappaB) p65 serine 276 phosphorylation. bardoxolone methyl 0-7 synaptotagmin 1 Rattus norvegicus 139-142 31574956-6 2019 Furthermore, CDDO-Me increased astroglial viability via the up-regulation of nuclear factor-erythroid 2-related factor 2 (Nrf2) expression. bardoxolone methyl 13-20 NFE2 like bZIP transcription factor 2 Rattus norvegicus 77-120 31574956-6 2019 Furthermore, CDDO-Me increased astroglial viability via the up-regulation of nuclear factor-erythroid 2-related factor 2 (Nrf2) expression. bardoxolone methyl 13-20 NFE2 like bZIP transcription factor 2 Rattus norvegicus 122-126 31574956-7 2019 Therefore, our findings suggest that CDDO-Me may ameliorate SE-induced vasogenic edema formation by regulating NF-kappaB p65 phosphorylations in microglia as well as endothelial cells and enhancing Nrf2 expression in astrocytes, respectively. bardoxolone methyl 37-44 NFE2 like bZIP transcription factor 2 Rattus norvegicus 198-202 31580543-6 2019 Bardoxolone exerts antioxidant activity by promoting the activation of Nrf2 (Nuclear factor erythroid2-derivative - 2) and the downregulation of the proinflammatory NF-kB (Nuclear factor kappa-light-chain-enhancer of activated B cells) signaling. bardoxolone methyl 0-11 NFE2 like bZIP transcription factor 2 Homo sapiens 71-75 30079536-4 2019 OA derivatives, such as CDDO-Im and CDDO-Me, are even more potent Nrf2 activators. bardoxolone methyl 36-43 NFE2 like bZIP transcription factor 2 Homo sapiens 66-70 31387295-0 2019 CDDO-Me Selectively Attenuates CA1 Neuronal Death Induced by Status Epilepticus via Facilitating Mitochondrial Fission Independent of LONP1. bardoxolone methyl 0-7 carbonic anhydrase 1 Homo sapiens 31-34 31387295-0 2019 CDDO-Me Selectively Attenuates CA1 Neuronal Death Induced by Status Epilepticus via Facilitating Mitochondrial Fission Independent of LONP1. bardoxolone methyl 0-7 lon peptidase 1, mitochondrial Homo sapiens 134-139 30826055-0 2019 CDDO-Me, Sulforaphane and tBHQ attenuate the RANKL-induced osteoclast differentiation via activating the NRF2-mediated antioxidant response. bardoxolone methyl 0-7 TNF superfamily member 11 Homo sapiens 45-50 30826055-0 2019 CDDO-Me, Sulforaphane and tBHQ attenuate the RANKL-induced osteoclast differentiation via activating the NRF2-mediated antioxidant response. bardoxolone methyl 0-7 NFE2 like bZIP transcription factor 2 Homo sapiens 105-109 30826055-8 2019 Taken together, these results suggest that CDDO-Me, SFN and tBHQ attenuate RANKL-induced osteoclastogenesis via activation of NRF2-mediated antioxidant response. bardoxolone methyl 43-50 TNF superfamily member 11 Homo sapiens 75-80 30826055-8 2019 Taken together, these results suggest that CDDO-Me, SFN and tBHQ attenuate RANKL-induced osteoclastogenesis via activation of NRF2-mediated antioxidant response. bardoxolone methyl 43-50 NFE2 like bZIP transcription factor 2 Homo sapiens 126-130 31387295-2 2019 In addition, CDDO-Me affects cellular differentiation and cell cycle arrest, and irreversibly inhibits Lon protease-1 (LONP1). bardoxolone methyl 13-20 lon peptidase 1, mitochondrial Homo sapiens 103-117 31387295-2 2019 In addition, CDDO-Me affects cellular differentiation and cell cycle arrest, and irreversibly inhibits Lon protease-1 (LONP1). bardoxolone methyl 13-20 lon peptidase 1, mitochondrial Homo sapiens 119-124 31387295-4 2019 CDDO-Me increased dynamin-related proteins 1 (DRP1)-serine 616 phosphorylation via activating extracellular-signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK), but not protein kinase A (PKA) or protein phosphatases (PPs). bardoxolone methyl 0-7 dynamin 1 like Homo sapiens 18-44 31387295-4 2019 CDDO-Me increased dynamin-related proteins 1 (DRP1)-serine 616 phosphorylation via activating extracellular-signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK), but not protein kinase A (PKA) or protein phosphatases (PPs). bardoxolone methyl 0-7 dynamin 1 like Homo sapiens 46-50 31387295-4 2019 CDDO-Me increased dynamin-related proteins 1 (DRP1)-serine 616 phosphorylation via activating extracellular-signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK), but not protein kinase A (PKA) or protein phosphatases (PPs). bardoxolone methyl 0-7 mitogen-activated protein kinase 1 Homo sapiens 94-135 31387295-4 2019 CDDO-Me increased dynamin-related proteins 1 (DRP1)-serine 616 phosphorylation via activating extracellular-signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK), but not protein kinase A (PKA) or protein phosphatases (PPs). bardoxolone methyl 0-7 mitogen-activated protein kinase 3 Homo sapiens 137-143 31387295-4 2019 CDDO-Me increased dynamin-related proteins 1 (DRP1)-serine 616 phosphorylation via activating extracellular-signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK), but not protein kinase A (PKA) or protein phosphatases (PPs). bardoxolone methyl 0-7 mitogen-activated protein kinase 8 Homo sapiens 149-172 31387295-4 2019 CDDO-Me increased dynamin-related proteins 1 (DRP1)-serine 616 phosphorylation via activating extracellular-signal-regulated kinase 1/2 (ERK1/2) and c-Jun N-terminal kinase (JNK), but not protein kinase A (PKA) or protein phosphatases (PPs). bardoxolone methyl 0-7 mitogen-activated protein kinase 8 Homo sapiens 174-177 31387295-5 2019 In addition, CDDO-Me facilitated DRP1-mediated mitochondrial fissions, which selectively attenuated SE-induced CA1 neuronal death. bardoxolone methyl 13-20 dynamin 1 like Homo sapiens 33-37 31387295-5 2019 In addition, CDDO-Me facilitated DRP1-mediated mitochondrial fissions, which selectively attenuated SE-induced CA1 neuronal death. bardoxolone methyl 13-20 carbonic anhydrase 1 Homo sapiens 111-114 31387295-8 2019 Co-treatment of CDDO-Me with LONP1 siRNA ameliorated only CA1 neuronal death, concomitant with abrogation of mitochondrial elongation induced by SE. bardoxolone methyl 16-23 lon peptidase 1, mitochondrial Homo sapiens 29-34 31387295-8 2019 Co-treatment of CDDO-Me with LONP1 siRNA ameliorated only CA1 neuronal death, concomitant with abrogation of mitochondrial elongation induced by SE. bardoxolone methyl 16-23 carbonic anhydrase 1 Homo sapiens 58-61 31387295-9 2019 Thus, our findings suggest that CDDO-Me may selectively attenuate SE-induced CA1 neuronal death by rescuing the abnormal mitochondrial machinery, independent of LONP1 activity. bardoxolone methyl 32-39 carbonic anhydrase 1 Homo sapiens 77-80 31170712-3 2019 METHODS: BEACON randomized patients (n = 2,185) with type 2 diabetes (T2DM) and stage 4 chronic kidney disease (CKD) 1:1 to receive Bard (20 mg) or placebo once daily. bardoxolone methyl 132-136 ubiquitin like 5 Homo sapiens 9-15 30456486-11 2019 Several other, clinically relevant, compounds (i.e., sulphorophane, nitrofurantoin and CDDO-Me) also enhanced the induction of Srxn1-GFP upon two consecutive repeated exposure. bardoxolone methyl 87-94 sulfiredoxin 1 Homo sapiens 127-132 30465784-8 2019 Interestingly, anti-rotaviral effects of RA-839 were also mimicked by 2-Cyano-3, 12-dioxo-oleana-1, 9(11)-dien-28-oic acid methyl ester (CDDO-Me) and Hemin, two classical pharmacological activators of Nrf2/ARE pathway. bardoxolone methyl 137-144 NFE2 like bZIP transcription factor 2 Homo sapiens 201-205 30279437-9 2018 The pre-clinically tested inhibitors of PI3K (PX-866) and NFkappaB (CDDO-Me) suppressed MSC-mediated HIV-1 reactivation. bardoxolone methyl 68-75 nuclear factor kappa B subunit 1 Homo sapiens 58-66 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 0-18 AKT serine/threonine kinase 1 Homo sapiens 99-102 28978980-8 2018 The increase in reactive oxygen species (ROS) and upregulation of angiotensinogen expression in the kidneys of the Ald group was significantly prevented in the Ald-BM group. bardoxolone methyl 164-166 angiotensinogen Homo sapiens 66-81 28978980-9 2018 The upregulation of human L-FABP expression induced in the kidneys and increase in urinary L-FABP in the Ald group were significantly suppressed by BM administration. bardoxolone methyl 148-150 fatty acid binding protein 1 Homo sapiens 26-32 28978980-9 2018 The upregulation of human L-FABP expression induced in the kidneys and increase in urinary L-FABP in the Ald group were significantly suppressed by BM administration. bardoxolone methyl 148-150 fatty acid binding protein 1 Homo sapiens 91-97 29953997-5 2018 CDDO-me exerted cytotoxic activity against W4P-LHB-expressing NIH3T3 cells, HepG2 cells, and Huh7 cells, and induced apoptotic cell death in a dose-dependent manner, demonstrating its anti-cancer activity against HCC. bardoxolone methyl 0-7 luteinizing hormone beta Mus musculus 47-50 29953997-5 2018 CDDO-me exerted cytotoxic activity against W4P-LHB-expressing NIH3T3 cells, HepG2 cells, and Huh7 cells, and induced apoptotic cell death in a dose-dependent manner, demonstrating its anti-cancer activity against HCC. bardoxolone methyl 0-7 MIR7-3 host gene Homo sapiens 93-97 29953997-6 2018 Sublethal concentrations of CDDO-me suppressed STAT3 activation by W4P-LHB ectopic expression and interleukin-6 treatment in W4P-LHB-NIH3T3 and Huh7 cells respectively. bardoxolone methyl 28-35 signal transducer and activator of transcription 3 Homo sapiens 47-52 29953997-6 2018 Sublethal concentrations of CDDO-me suppressed STAT3 activation by W4P-LHB ectopic expression and interleukin-6 treatment in W4P-LHB-NIH3T3 and Huh7 cells respectively. bardoxolone methyl 28-35 luteinizing hormone beta Mus musculus 71-74 29953997-6 2018 Sublethal concentrations of CDDO-me suppressed STAT3 activation by W4P-LHB ectopic expression and interleukin-6 treatment in W4P-LHB-NIH3T3 and Huh7 cells respectively. bardoxolone methyl 28-35 interleukin 6 Homo sapiens 98-111 29953997-6 2018 Sublethal concentrations of CDDO-me suppressed STAT3 activation by W4P-LHB ectopic expression and interleukin-6 treatment in W4P-LHB-NIH3T3 and Huh7 cells respectively. bardoxolone methyl 28-35 luteinizing hormone subunit beta Homo sapiens 129-132 29953997-6 2018 Sublethal concentrations of CDDO-me suppressed STAT3 activation by W4P-LHB ectopic expression and interleukin-6 treatment in W4P-LHB-NIH3T3 and Huh7 cells respectively. bardoxolone methyl 28-35 MIR7-3 host gene Homo sapiens 144-148 29953997-7 2018 The suppression of STAT3 activation by CDDO-me in W4P-LHB-NIH3T3 cells was further confirmed by decreased cyclin D1 protein levels and increased p21 and p53 mRNA synthesis. bardoxolone methyl 39-46 signal transducer and activator of transcription 3 Homo sapiens 19-24 29953997-7 2018 The suppression of STAT3 activation by CDDO-me in W4P-LHB-NIH3T3 cells was further confirmed by decreased cyclin D1 protein levels and increased p21 and p53 mRNA synthesis. bardoxolone methyl 39-46 luteinizing hormone subunit beta Homo sapiens 54-57 29953997-7 2018 The suppression of STAT3 activation by CDDO-me in W4P-LHB-NIH3T3 cells was further confirmed by decreased cyclin D1 protein levels and increased p21 and p53 mRNA synthesis. bardoxolone methyl 39-46 cyclin D1 Homo sapiens 106-115 29953997-7 2018 The suppression of STAT3 activation by CDDO-me in W4P-LHB-NIH3T3 cells was further confirmed by decreased cyclin D1 protein levels and increased p21 and p53 mRNA synthesis. bardoxolone methyl 39-46 H3 histone pseudogene 16 Homo sapiens 145-148 29953997-7 2018 The suppression of STAT3 activation by CDDO-me in W4P-LHB-NIH3T3 cells was further confirmed by decreased cyclin D1 protein levels and increased p21 and p53 mRNA synthesis. bardoxolone methyl 39-46 tumor protein p53 Homo sapiens 153-156 29953997-8 2018 In addition, CDDO-me treatment resulted in decreased cell migration and colony formation in in vitro assays using W4P-LHB-NIH3T3, HepG2, or Huh7 cell lines, supporting its anti-cancer activity through STAT3 inhibition. bardoxolone methyl 13-20 luteinizing hormone subunit beta Homo sapiens 118-121 29953997-8 2018 In addition, CDDO-me treatment resulted in decreased cell migration and colony formation in in vitro assays using W4P-LHB-NIH3T3, HepG2, or Huh7 cell lines, supporting its anti-cancer activity through STAT3 inhibition. bardoxolone methyl 13-20 MIR7-3 host gene Homo sapiens 140-144 29953997-8 2018 In addition, CDDO-me treatment resulted in decreased cell migration and colony formation in in vitro assays using W4P-LHB-NIH3T3, HepG2, or Huh7 cell lines, supporting its anti-cancer activity through STAT3 inhibition. bardoxolone methyl 13-20 signal transducer and activator of transcription 3 Homo sapiens 201-206 29953997-9 2018 Furthermore, -CDDO-me administration significantly suppressed tumor growth induced by W4P-LHB-expressing NIH3T3 cells in nude mice, confirming its anti-cancer activity. bardoxolone methyl 14-21 luteinizing hormone beta Mus musculus 90-93 29953997-10 2018 Collectively, our findings demonstrated that CDDO-me is capable of suppressing STAT3 activation in HCC cells and cells transformed by the natural variant of HBV protein. bardoxolone methyl 45-52 signal transducer and activator of transcription 3 Homo sapiens 79-84 28978980-1 2018 The aim of this study was to investigate the renoprotective effect of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator with an antioxidant effect, in a salt-sensitive hypertension model induced by aldosterone (Ald) and salt. bardoxolone methyl 70-88 NFE2 like bZIP transcription factor 2 Homo sapiens 97-140 28978980-1 2018 The aim of this study was to investigate the renoprotective effect of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator with an antioxidant effect, in a salt-sensitive hypertension model induced by aldosterone (Ald) and salt. bardoxolone methyl 70-88 NFE2 like bZIP transcription factor 2 Homo sapiens 142-146 28978980-1 2018 The aim of this study was to investigate the renoprotective effect of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator with an antioxidant effect, in a salt-sensitive hypertension model induced by aldosterone (Ald) and salt. bardoxolone methyl 90-92 NFE2 like bZIP transcription factor 2 Homo sapiens 97-140 28978980-1 2018 The aim of this study was to investigate the renoprotective effect of bardoxolone methyl (BM), a nuclear factor erythroid 2-related factor 2 (Nrf2) activator with an antioxidant effect, in a salt-sensitive hypertension model induced by aldosterone (Ald) and salt. bardoxolone methyl 90-92 NFE2 like bZIP transcription factor 2 Homo sapiens 142-146 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 0-18 mechanistic target of rapamycin kinase Homo sapiens 103-107 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 0-18 mitogen-activated protein kinase 1 Homo sapiens 112-115 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 0-18 mitogen-activated protein kinase 3 Homo sapiens 121-127 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 20-27 AKT serine/threonine kinase 1 Homo sapiens 99-102 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 20-27 mechanistic target of rapamycin kinase Homo sapiens 103-107 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 20-27 mitogen-activated protein kinase 1 Homo sapiens 112-115 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 20-27 mitogen-activated protein kinase 3 Homo sapiens 121-127 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 31-37 AKT serine/threonine kinase 1 Homo sapiens 99-102 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 31-37 mechanistic target of rapamycin kinase Homo sapiens 103-107 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 31-37 mitogen-activated protein kinase 1 Homo sapiens 112-115 29118925-0 2017 Bardoxolone methyl (CDDO-Me or RTA402) induces cell cycle arrest, apoptosis and autophagy via PI3K/Akt/mTOR and p38 MAPK/Erk1/2 signaling pathways in K562 cells. bardoxolone methyl 31-37 mitogen-activated protein kinase 3 Homo sapiens 121-127 29118925-2 2017 CDDO-Me (Bardoxolone methyl), a potent Nrf2 activator and NF-kappaB inhibitor, is a promising candidate for cancer treatment including leukemia. bardoxolone methyl 0-7 NFE2 like bZIP transcription factor 2 Homo sapiens 39-43 29118925-5 2017 A total of 1,555 proteins responded to CDDO-Me exposure, including FANCI, SRPK2, XPO5, HP1BP3, NELFCD, Na+,K+-ATPase 1, etc. bardoxolone methyl 39-46 FA complementation group I Felis catus 67-72 29118925-5 2017 A total of 1,555 proteins responded to CDDO-Me exposure, including FANCI, SRPK2, XPO5, HP1BP3, NELFCD, Na+,K+-ATPase 1, etc. bardoxolone methyl 39-46 SRSF protein kinase 2 Felis catus 74-79 29118925-5 2017 A total of 1,555 proteins responded to CDDO-Me exposure, including FANCI, SRPK2, XPO5, HP1BP3, NELFCD, Na+,K+-ATPase 1, etc. bardoxolone methyl 39-46 exportin 5 Felis catus 81-85 29118925-5 2017 A total of 1,555 proteins responded to CDDO-Me exposure, including FANCI, SRPK2, XPO5, HP1BP3, NELFCD, Na+,K+-ATPase 1, etc. bardoxolone methyl 39-46 heterochromatin protein 1 binding protein 3 Felis catus 87-93 29118925-5 2017 A total of 1,555 proteins responded to CDDO-Me exposure, including FANCI, SRPK2, XPO5, HP1BP3, NELFCD, Na+,K+-ATPase 1, etc. bardoxolone methyl 39-46 negative elongation factor complex member C/D Felis catus 95-101 29118925-9 2017 CDDO-Me caused mitochondria-, death receptor-dependent and ER stress-mediated apoptosis in K562 cells, also induced autophagy with the suppression of PI3K/Akt/mTOR signaling pathway. bardoxolone methyl 0-7 AKT serine/threonine kinase 1 Homo sapiens 155-158 29118925-9 2017 CDDO-Me caused mitochondria-, death receptor-dependent and ER stress-mediated apoptosis in K562 cells, also induced autophagy with the suppression of PI3K/Akt/mTOR signaling pathway. bardoxolone methyl 0-7 mechanistic target of rapamycin kinase Homo sapiens 159-163 29118925-10 2017 p38 MAPK/Erk1/2 signaling pathways contributed to both apoptosis- and autophagy-inducing effects of CDDO-Me in K562 cells. bardoxolone methyl 100-107 mitogen-activated protein kinase 1 Homo sapiens 0-3 29118925-10 2017 p38 MAPK/Erk1/2 signaling pathways contributed to both apoptosis- and autophagy-inducing effects of CDDO-Me in K562 cells. bardoxolone methyl 100-107 mitogen-activated protein kinase 3 Homo sapiens 9-15 28275049-3 2017 In contrast, both Bar-Me and CF3DODA-Me induce reactive oxygen species in HL-60 and Jurkat leukemia cells, inhibit cell growth, induce apoptosis and differentiation, and decrease expression of specificity proteins (Sp) 1, 3, and 4, and cMyc, and these effects are significantly attenuated after cotreatment with the antioxidant GSH. bardoxolone methyl 18-24 Sp1 transcription factor Homo sapiens 193-230 28273344-10 2017 Importantly, CDDO-Me treatment reduced allergen-induced ATP secretion and IL-33 release by airway epithelial cells in vitro and protected mice from IL-33 release and asthma-like pathological changes in the lungs. bardoxolone methyl 13-20 interleukin 33 Mus musculus 74-79 28273344-10 2017 Importantly, CDDO-Me treatment reduced allergen-induced ATP secretion and IL-33 release by airway epithelial cells in vitro and protected mice from IL-33 release and asthma-like pathological changes in the lungs. bardoxolone methyl 13-20 interleukin 33 Mus musculus 148-153 28596283-3 2017 CDDO-Me (bardoxolone methyl) is a drug that inhibits the survival of leukemic cells by targeting different pro-survival molecules, including STAT3. bardoxolone methyl 0-7 signal transducer and activator of transcription 3 Homo sapiens 141-146 28596283-4 2017 We found that CDDO-Me inhibits proliferation and survival of tyrosine kinase inhibitor-resistant BCR-ABL1+ cell lines and primary leukemic cells, including cells harboring BCR-ABL1T315I or T315I+ compound mutations. bardoxolone methyl 14-21 BCR activator of RhoGEF and GTPase Homo sapiens 97-105 28596283-4 2017 We found that CDDO-Me inhibits proliferation and survival of tyrosine kinase inhibitor-resistant BCR-ABL1+ cell lines and primary leukemic cells, including cells harboring BCR-ABL1T315I or T315I+ compound mutations. bardoxolone methyl 14-21 BCR activator of RhoGEF and GTPase Homo sapiens 97-100 28596283-5 2017 Furthermore, CDDO-Me was found to block growth and survival of CD34+/CD38- leukemic stem cells (LSC). bardoxolone methyl 13-20 CD34 molecule Homo sapiens 63-67 28596283-5 2017 Furthermore, CDDO-Me was found to block growth and survival of CD34+/CD38- leukemic stem cells (LSC). bardoxolone methyl 13-20 CD38 molecule Homo sapiens 69-73 28596283-9 2017 However, CDDO-Me was also found to increase the expression of heme-oxygenase-1, a heat-shock-protein that triggers drug resistance and cell survival. bardoxolone methyl 9-16 heme oxygenase 1 Homo sapiens 62-78 28596283-11 2017 Moreover, SMA-ZnPP was found to sensitize BCR-ABL1+ cells against the combination "CDDO-Me+ tyrosine kinase inhibitor". bardoxolone methyl 83-90 BCR activator of RhoGEF and GTPase Homo sapiens 42-50 28886135-4 2017 Interestingly, treatment of patient PBMCs with the Nuclear Factor-Erythroid-2-Related Factor 2 (NRF2) agonist, CDDO-Me(bardoxolone methyl), reduced MCP-1 production but not IL-6 or Interleukin-10 (IL-10) secretion. bardoxolone methyl 111-118 NFE2 like bZIP transcription factor 2 Homo sapiens 51-94 28886135-4 2017 Interestingly, treatment of patient PBMCs with the Nuclear Factor-Erythroid-2-Related Factor 2 (NRF2) agonist, CDDO-Me(bardoxolone methyl), reduced MCP-1 production but not IL-6 or Interleukin-10 (IL-10) secretion. bardoxolone methyl 111-118 NFE2 like bZIP transcription factor 2 Homo sapiens 96-100 28886135-4 2017 Interestingly, treatment of patient PBMCs with the Nuclear Factor-Erythroid-2-Related Factor 2 (NRF2) agonist, CDDO-Me(bardoxolone methyl), reduced MCP-1 production but not IL-6 or Interleukin-10 (IL-10) secretion. bardoxolone methyl 111-118 C-C motif chemokine ligand 2 Homo sapiens 148-153 28886135-4 2017 Interestingly, treatment of patient PBMCs with the Nuclear Factor-Erythroid-2-Related Factor 2 (NRF2) agonist, CDDO-Me(bardoxolone methyl), reduced MCP-1 production but not IL-6 or Interleukin-10 (IL-10) secretion. bardoxolone methyl 111-118 interleukin 10 Homo sapiens 181-195 28886135-4 2017 Interestingly, treatment of patient PBMCs with the Nuclear Factor-Erythroid-2-Related Factor 2 (NRF2) agonist, CDDO-Me(bardoxolone methyl), reduced MCP-1 production but not IL-6 or Interleukin-10 (IL-10) secretion. bardoxolone methyl 111-118 interleukin 10 Homo sapiens 197-202 28886135-5 2017 In enriched monocytes from healthy donors, CDDO-Me(bardoxolone methyl) also reduced LPS-induced MCP1/CCL2 production but did not alter IL-6 or IL-10 secretion. bardoxolone methyl 43-50 C-C motif chemokine ligand 2 Homo sapiens 96-100 28886135-5 2017 In enriched monocytes from healthy donors, CDDO-Me(bardoxolone methyl) also reduced LPS-induced MCP1/CCL2 production but did not alter IL-6 or IL-10 secretion. bardoxolone methyl 43-50 C-C motif chemokine ligand 2 Homo sapiens 101-105 28886135-7 2017 Hence, our findings with CDDO-Me(bardoxolone methyl) and Compound 7 are likely to reflect a generalizable aspect of NRF2 activation. bardoxolone methyl 25-32 NFE2 like bZIP transcription factor 2 Homo sapiens 116-120 28275049-3 2017 In contrast, both Bar-Me and CF3DODA-Me induce reactive oxygen species in HL-60 and Jurkat leukemia cells, inhibit cell growth, induce apoptosis and differentiation, and decrease expression of specificity proteins (Sp) 1, 3, and 4, and cMyc, and these effects are significantly attenuated after cotreatment with the antioxidant GSH. bardoxolone methyl 18-24 MYC proto-oncogene, bHLH transcription factor Homo sapiens 236-240 27780924-0 2016 CDDO-Me reveals USP7 as a novel target in ovarian cancer cells. bardoxolone methyl 0-7 ubiquitin specific peptidase 7 Homo sapiens 16-20 27979484-4 2017 The effect of bardoxolone methyl (BM) on miR-21, miR-223-5p, and miR-125b in renal IR injury was evaluated in this study. bardoxolone methyl 34-36 microRNA 21 Rattus norvegicus 41-47 27979484-4 2017 The effect of bardoxolone methyl (BM) on miR-21, miR-223-5p, and miR-125b in renal IR injury was evaluated in this study. bardoxolone methyl 34-36 microRNA 223 Rattus norvegicus 49-56 27780924-3 2016 In this study, we identified synthetic triterpenoid C-28 methyl ester of 2-cyano-3, 12-dioxoolen-1, 9-dien-28-oic acid (CDDO-Me) as a novel inhibitor of USP7 but not of other cysteine proteases such as cathepsin B and cathepsin D. bardoxolone methyl 120-127 cathepsin D Homo sapiens 218-229 27780924-4 2016 CDDO-Me inhibits USP7 activity via a mechanism that is independent of the presence of alpha, beta-unsaturated ketones. bardoxolone methyl 0-7 ubiquitin specific peptidase 7 Homo sapiens 17-21 27780924-3 2016 In this study, we identified synthetic triterpenoid C-28 methyl ester of 2-cyano-3, 12-dioxoolen-1, 9-dien-28-oic acid (CDDO-Me) as a novel inhibitor of USP7 but not of other cysteine proteases such as cathepsin B and cathepsin D. bardoxolone methyl 120-127 ubiquitin specific peptidase 7 Homo sapiens 153-157 27780924-5 2016 Molecular docking studies showed that CDDO-Me fits well in the ubiquitin carboxyl terminus-binding pocket on USP7. bardoxolone methyl 38-45 ubiquitin specific peptidase 7 Homo sapiens 109-113 27780924-6 2016 Given that CDDO-Me is known to be effective against ovarian cancer cells, we speculated that CDDO-Me may target USP7 in ovarian cancer cells. bardoxolone methyl 11-18 ubiquitin specific peptidase 7 Homo sapiens 112-116 27780924-3 2016 In this study, we identified synthetic triterpenoid C-28 methyl ester of 2-cyano-3, 12-dioxoolen-1, 9-dien-28-oic acid (CDDO-Me) as a novel inhibitor of USP7 but not of other cysteine proteases such as cathepsin B and cathepsin D. bardoxolone methyl 120-127 cathepsin B Homo sapiens 202-213 27780924-6 2016 Given that CDDO-Me is known to be effective against ovarian cancer cells, we speculated that CDDO-Me may target USP7 in ovarian cancer cells. bardoxolone methyl 93-100 ubiquitin specific peptidase 7 Homo sapiens 112-116 27780924-9 2016 Using the cellular thermal shift assay and the drug affinity responsive target stability assay, we further demonstrated that CDDO-Me directly binds to USP7 in cells, which leads to the decrease of its substrates such as MDM2, MDMX and UHRF1. bardoxolone methyl 125-132 ubiquitin specific peptidase 7 Homo sapiens 151-155 27780924-9 2016 Using the cellular thermal shift assay and the drug affinity responsive target stability assay, we further demonstrated that CDDO-Me directly binds to USP7 in cells, which leads to the decrease of its substrates such as MDM2, MDMX and UHRF1. bardoxolone methyl 125-132 MDM2 proto-oncogene Homo sapiens 220-224 27780924-9 2016 Using the cellular thermal shift assay and the drug affinity responsive target stability assay, we further demonstrated that CDDO-Me directly binds to USP7 in cells, which leads to the decrease of its substrates such as MDM2, MDMX and UHRF1. bardoxolone methyl 125-132 MDM4 regulator of p53 Homo sapiens 226-230 27780924-9 2016 Using the cellular thermal shift assay and the drug affinity responsive target stability assay, we further demonstrated that CDDO-Me directly binds to USP7 in cells, which leads to the decrease of its substrates such as MDM2, MDMX and UHRF1. bardoxolone methyl 125-132 ubiquitin like with PHD and ring finger domains 1 Homo sapiens 235-240 27780924-11 2016 In conclusion, we demonstrate that USP7 is a novel target of ovarian cancer cells; targeting USP7 may contribute to the anti-cancer effect of CDDO-Me. bardoxolone methyl 142-149 ubiquitin specific peptidase 7 Homo sapiens 35-39 27780924-11 2016 In conclusion, we demonstrate that USP7 is a novel target of ovarian cancer cells; targeting USP7 may contribute to the anti-cancer effect of CDDO-Me. bardoxolone methyl 142-149 ubiquitin specific peptidase 7 Homo sapiens 93-97 27780924-12 2016 The development of novel USP7 selective compounds based on the CDDO-Me-scaffold warrants further investigation. bardoxolone methyl 63-70 ubiquitin specific peptidase 7 Homo sapiens 25-29 27633142-7 2016 More importantly, activation of Nrf2 signaling by CDDO-Me significantly alleviated cyclophosphamide-induced myelosuppression, while this alleviation was diminished in Nrf2-/- mice. bardoxolone methyl 50-57 nuclear factor, erythroid derived 2, like 2 Mus musculus 32-36 27693327-5 2016 At the same time, the activation of Nrf2 with D, l-sulforaphane (SFN) and CDDO-Me could repress the replication of SVCV, and knockdown of Nrf2 by siRNA could promote the replication of SVCV. bardoxolone methyl 74-81 NFE2 like bZIP transcription factor 2 Homo sapiens 36-40 27480280-3 2016 We aimed to evaluate the ability of short-term treatment with the Nrf2 activator bardoxolone methyl (CDDO-Me) to protect against cisplatin-induced kidney cell toxicity. bardoxolone methyl 81-99 NFE2 like bZIP transcription factor 2 Homo sapiens 66-70 27480280-3 2016 We aimed to evaluate the ability of short-term treatment with the Nrf2 activator bardoxolone methyl (CDDO-Me) to protect against cisplatin-induced kidney cell toxicity. bardoxolone methyl 101-108 NFE2 like bZIP transcription factor 2 Homo sapiens 66-70 27480280-6 2016 Gene regulation (mRNA expression) studies revealed the ability of CDDO-Me to modify protective pathways including Nrf2 induced detoxifying genes [GCLC (increased 1.9-fold), NQO1 (increased 9.3-fold)], and an efflux transporter [SLC47A1 (increased 4.5-fold)] at 12h. bardoxolone methyl 66-73 NFE2 like bZIP transcription factor 2 Homo sapiens 114-118 27480280-6 2016 Gene regulation (mRNA expression) studies revealed the ability of CDDO-Me to modify protective pathways including Nrf2 induced detoxifying genes [GCLC (increased 1.9-fold), NQO1 (increased 9.3-fold)], and an efflux transporter [SLC47A1 (increased 4.5-fold)] at 12h. bardoxolone methyl 66-73 glutamate-cysteine ligase catalytic subunit Homo sapiens 146-150 27480280-6 2016 Gene regulation (mRNA expression) studies revealed the ability of CDDO-Me to modify protective pathways including Nrf2 induced detoxifying genes [GCLC (increased 1.9-fold), NQO1 (increased 9.3-fold)], and an efflux transporter [SLC47A1 (increased 4.5-fold)] at 12h. bardoxolone methyl 66-73 NAD(P)H quinone dehydrogenase 1 Homo sapiens 173-177 27480280-6 2016 Gene regulation (mRNA expression) studies revealed the ability of CDDO-Me to modify protective pathways including Nrf2 induced detoxifying genes [GCLC (increased 1.9-fold), NQO1 (increased 9.3-fold)], and an efflux transporter [SLC47A1 (increased 4.5-fold)] at 12h. bardoxolone methyl 66-73 solute carrier family 47 member 1 Homo sapiens 228-235 27480280-8 2016 Immunofluorescence revealed localization of GCLC and NQO1 to the nucleus and cytosol, respectively, with CDDO-Me administered prior to or after cisplatin exposure. bardoxolone methyl 105-112 glutamate-cysteine ligase catalytic subunit Homo sapiens 44-48 27480280-8 2016 Immunofluorescence revealed localization of GCLC and NQO1 to the nucleus and cytosol, respectively, with CDDO-Me administered prior to or after cisplatin exposure. bardoxolone methyl 105-112 NAD(P)H quinone dehydrogenase 1 Homo sapiens 53-57 27480280-9 2016 The findings of enhanced cell viability and increased expression of detoxifying enzymes (GCLC and NQO1) and the multidrug and toxin extrusion protein 1 (MATE1) efflux transporter (SLC47A1) in hPTCs exposed to CDDO-Me, suggest that intermittent treatment with CDDO-Me prior to or after cisplatin exposure may be a promising approach to mitigate acute kidney injury. bardoxolone methyl 209-216 solute carrier family 47 member 1 Homo sapiens 112-151 27480280-9 2016 The findings of enhanced cell viability and increased expression of detoxifying enzymes (GCLC and NQO1) and the multidrug and toxin extrusion protein 1 (MATE1) efflux transporter (SLC47A1) in hPTCs exposed to CDDO-Me, suggest that intermittent treatment with CDDO-Me prior to or after cisplatin exposure may be a promising approach to mitigate acute kidney injury. bardoxolone methyl 209-216 solute carrier family 47 member 1 Homo sapiens 153-158 27480280-9 2016 The findings of enhanced cell viability and increased expression of detoxifying enzymes (GCLC and NQO1) and the multidrug and toxin extrusion protein 1 (MATE1) efflux transporter (SLC47A1) in hPTCs exposed to CDDO-Me, suggest that intermittent treatment with CDDO-Me prior to or after cisplatin exposure may be a promising approach to mitigate acute kidney injury. bardoxolone methyl 209-216 solute carrier family 47 member 1 Homo sapiens 180-187 27480280-9 2016 The findings of enhanced cell viability and increased expression of detoxifying enzymes (GCLC and NQO1) and the multidrug and toxin extrusion protein 1 (MATE1) efflux transporter (SLC47A1) in hPTCs exposed to CDDO-Me, suggest that intermittent treatment with CDDO-Me prior to or after cisplatin exposure may be a promising approach to mitigate acute kidney injury. bardoxolone methyl 259-266 solute carrier family 47 member 1 Homo sapiens 153-158 27480280-9 2016 The findings of enhanced cell viability and increased expression of detoxifying enzymes (GCLC and NQO1) and the multidrug and toxin extrusion protein 1 (MATE1) efflux transporter (SLC47A1) in hPTCs exposed to CDDO-Me, suggest that intermittent treatment with CDDO-Me prior to or after cisplatin exposure may be a promising approach to mitigate acute kidney injury. bardoxolone methyl 259-266 solute carrier family 47 member 1 Homo sapiens 180-187 27192495-3 2016 The Nrf2 activator DMF (Dimethylfumarates) has been approved by the FDA as a new first-line oral drug to treat patients with relapsing forms of multiple sclerosis, while a phase 3 study of another promising candidate, CDDO-Me, was terminated for safety reasons. bardoxolone methyl 218-225 NFE2 like bZIP transcription factor 2 Homo sapiens 4-8 26908173-1 2016 Semisynthetic triterpenoids such as bardoxolone methyl (methyl-2-cyano 3,12-dioxooleano-1,9-dien-28-oate; CDDO-Me) (4) are potent inducers of antioxidant and anti-inflammatory signaling pathways, including those regulated by the transcription factor Nrf2. bardoxolone methyl 106-113 NFE2 like bZIP transcription factor 2 Rattus norvegicus 250-254 26918785-4 2016 We show that CDDO-Me treatment inhibits expression of IL-10 and VEGF in stimulated human M2 macrophages and TAMs but increases expression of TNF-alpha and IL-6. bardoxolone methyl 13-20 interleukin 10 Homo sapiens 54-59 26918785-4 2016 We show that CDDO-Me treatment inhibits expression of IL-10 and VEGF in stimulated human M2 macrophages and TAMs but increases expression of TNF-alpha and IL-6. bardoxolone methyl 13-20 vascular endothelial growth factor A Homo sapiens 64-68 26918785-4 2016 We show that CDDO-Me treatment inhibits expression of IL-10 and VEGF in stimulated human M2 macrophages and TAMs but increases expression of TNF-alpha and IL-6. bardoxolone methyl 13-20 tumor necrosis factor Homo sapiens 141-150 26918785-4 2016 We show that CDDO-Me treatment inhibits expression of IL-10 and VEGF in stimulated human M2 macrophages and TAMs but increases expression of TNF-alpha and IL-6. bardoxolone methyl 13-20 interleukin 6 Homo sapiens 155-159 26918785-5 2016 Surface expression of CD206 and CD163, which are characteristic of M2 activation, is significantly attenuated by CDDO-Me. bardoxolone methyl 113-120 mannose receptor C-type 1 Homo sapiens 22-27 26918785-5 2016 Surface expression of CD206 and CD163, which are characteristic of M2 activation, is significantly attenuated by CDDO-Me. bardoxolone methyl 113-120 CD163 molecule Homo sapiens 32-37 26918785-6 2016 In contrast, CDDO-Me up-regulates surface expression of HLA-DR and CD80, which are markers of M1 activation, and importantly potentiates macrophage activation of autologous T cells but inhibits endothelial cell vascularization. bardoxolone methyl 13-20 CD80 molecule Homo sapiens 67-71