PMID-sentid Pub_year Sent_text compound_name comp_offset prot_official_name organism prot_offset 34405716-0 2021 miR-128 participates in the pathogenesis of chronic constipation by regulating the p38alpha/M-CSF inflammatory signaling pathway. mir-128 0-7 mitogen-activated protein kinase 14 Mus musculus 83-91 34780888-5 2022 HAND2-AS1 endogenously sponged miR-128 and suppressed its activity via sequence complementation. mir-128 31-38 heart and neural crest derivatives expressed 2 Homo sapiens 0-5 34780888-5 2022 HAND2-AS1 endogenously sponged miR-128 and suppressed its activity via sequence complementation. mir-128 31-38 prostaglandin D2 receptor Homo sapiens 6-9 34780888-6 2022 Furthermore, HAND2-AS1 enhanced the expression of SIRT1 via binding to miR-128, thereby promoting ABCA1/G1 expression. mir-128 71-78 heart and neural crest derivatives expressed 2 Homo sapiens 13-18 34780888-6 2022 Furthermore, HAND2-AS1 enhanced the expression of SIRT1 via binding to miR-128, thereby promoting ABCA1/G1 expression. mir-128 71-78 prostaglandin D2 receptor Homo sapiens 19-22 34780888-6 2022 Furthermore, HAND2-AS1 enhanced the expression of SIRT1 via binding to miR-128, thereby promoting ABCA1/G1 expression. mir-128 71-78 sirtuin 1 Homo sapiens 50-55 34780888-6 2022 Furthermore, HAND2-AS1 enhanced the expression of SIRT1 via binding to miR-128, thereby promoting ABCA1/G1 expression. mir-128 71-78 ATP binding cassette subfamily A member 1 Homo sapiens 98-103 34151409-6 2021 Here, we focused on elucidating the possible role of miR-128 in AD pathology and found that miR-128 regulates the expression of two proteins essential for synaptic transmission, SNAP-25, and synaptotagmin1 (Syt1). mir-128 53-60 synaptosomal-associated protein 25 Mus musculus 178-185 34151409-6 2021 Here, we focused on elucidating the possible role of miR-128 in AD pathology and found that miR-128 regulates the expression of two proteins essential for synaptic transmission, SNAP-25, and synaptotagmin1 (Syt1). mir-128 53-60 synaptotagmin I Mus musculus 207-211 34965835-0 2021 Upregulation of miR-128 Mediates Heart Injury by Activating Wnt/beta-catenin Signaling Pathway in Heart Failure Mice. mir-128 16-23 Wnt family member 1 Homo sapiens 60-63 34965835-0 2021 Upregulation of miR-128 Mediates Heart Injury by Activating Wnt/beta-catenin Signaling Pathway in Heart Failure Mice. mir-128 16-23 catenin (cadherin associated protein), beta 1 Mus musculus 64-76 34965835-8 2021 Interference with miR-128 reduces Wnt1/beta-catenin expression in mouse failing hearts and ameliorates heart dysfunctional properties. mir-128 18-25 wingless-type MMTV integration site family, member 1 Mus musculus 34-38 34965835-8 2021 Interference with miR-128 reduces Wnt1/beta-catenin expression in mouse failing hearts and ameliorates heart dysfunctional properties. mir-128 18-25 catenin (cadherin associated protein), beta 1 Mus musculus 39-51 34965835-9 2021 We identified miR-128 directly targets to Axin1, an inhibitor of Wnt/beta-catenin signaling, and suppresses its inhibition on Wnt1/beta-catenin. mir-128 14-21 axin 1 Mus musculus 42-47 34965835-9 2021 We identified miR-128 directly targets to Axin1, an inhibitor of Wnt/beta-catenin signaling, and suppresses its inhibition on Wnt1/beta-catenin. mir-128 14-21 Wnt family member 1 Homo sapiens 65-68 34965835-9 2021 We identified miR-128 directly targets to Axin1, an inhibitor of Wnt/beta-catenin signaling, and suppresses its inhibition on Wnt1/beta-catenin. mir-128 14-21 catenin (cadherin associated protein), beta 1 Mus musculus 69-81 34965835-9 2021 We identified miR-128 directly targets to Axin1, an inhibitor of Wnt/beta-catenin signaling, and suppresses its inhibition on Wnt1/beta-catenin. mir-128 14-21 wingless-type MMTV integration site family, member 1 Mus musculus 126-130 34965835-9 2021 We identified miR-128 directly targets to Axin1, an inhibitor of Wnt/beta-catenin signaling, and suppresses its inhibition on Wnt1/beta-catenin. mir-128 14-21 catenin (cadherin associated protein), beta 1 Mus musculus 131-143 34965835-10 2021 Our study provides evidence indicating miR-128 as an inducer of HF and cardiac hypertrophy by enhancing Wnt1/beta-catenin in an Axin1-dependent nature. mir-128 39-46 wingless-type MMTV integration site family, member 1 Mus musculus 104-108 34965835-10 2021 Our study provides evidence indicating miR-128 as an inducer of HF and cardiac hypertrophy by enhancing Wnt1/beta-catenin in an Axin1-dependent nature. mir-128 39-46 catenin (cadherin associated protein), beta 1 Mus musculus 109-121 34965835-10 2021 Our study provides evidence indicating miR-128 as an inducer of HF and cardiac hypertrophy by enhancing Wnt1/beta-catenin in an Axin1-dependent nature. mir-128 39-46 axin 1 Mus musculus 128-133 34812491-14 2021 This study elaborated the mechanism by which Gs-Rb1 alleviates hepatotoxicity induced by foodborne 3-MCPD by stimulating autophagic flux via miR-128-targeted TFEB, which provides a reliable theoretical basis and target for the use of natural substances to reduce the harm of food processing pollutants on the human body. mir-128 141-148 RB transcriptional corepressor 1 Homo sapiens 48-51 34812491-14 2021 This study elaborated the mechanism by which Gs-Rb1 alleviates hepatotoxicity induced by foodborne 3-MCPD by stimulating autophagic flux via miR-128-targeted TFEB, which provides a reliable theoretical basis and target for the use of natural substances to reduce the harm of food processing pollutants on the human body. mir-128 141-148 transcription factor EB Homo sapiens 158-162 34405716-9 2021 Furthermore, co-culture assays indicated that regulating expression of miR-128 in colonic epithelial cells induced the secretion of IL-6 and TNF-alpha by macrophages. mir-128 71-78 interleukin 6 Mus musculus 132-136 34405716-9 2021 Furthermore, co-culture assays indicated that regulating expression of miR-128 in colonic epithelial cells induced the secretion of IL-6 and TNF-alpha by macrophages. mir-128 71-78 tumor necrosis factor Mus musculus 141-150 34405716-10 2021 In conclusion, our study demonstrated that miR-128 regulated the p38alpha/M-CSF signaling pathway to promote chronic inflammatory responses and changes in the immune microenvironment of the colon, thereby offering potential insights into the pathogenesis of CC and therapeutic targets for its treatment. mir-128 43-50 mitogen-activated protein kinase 14 Mus musculus 65-73 34092219-12 2021 CONCLUSIONS: Overall, down-regulating SNHG16 alleviated the sepsis-mediated ALI by regulating miR-128-3p/HMGB3. mir-128 94-101 high mobility group box 3 Rattus norvegicus 105-110 34384932-5 2021 We, herein using in silico analysis followed by in vitro experiments in C2C12 myoblasts, showed that miR-128 reduces mitochondrial biogenesis by directly targeting PGC1alpha. mir-128 101-108 PPARG coactivator 1 alpha Homo sapiens 164-173 34384932-6 2021 The expression of downstream genes, nuclear respiratory factors 1 and 2 (NRF1 and NRF2, respectively), and mitochondrial transcription factor A (TFAM) were decreased in C2C12 myoblasts upon overexpression of miR-128. mir-128 208-215 nuclear respiratory factor 1 Homo sapiens 36-71 34384932-6 2021 The expression of downstream genes, nuclear respiratory factors 1 and 2 (NRF1 and NRF2, respectively), and mitochondrial transcription factor A (TFAM) were decreased in C2C12 myoblasts upon overexpression of miR-128. mir-128 208-215 nuclear respiratory factor 1 Homo sapiens 73-77 34384932-6 2021 The expression of downstream genes, nuclear respiratory factors 1 and 2 (NRF1 and NRF2, respectively), and mitochondrial transcription factor A (TFAM) were decreased in C2C12 myoblasts upon overexpression of miR-128. mir-128 208-215 NFE2 like bZIP transcription factor 2 Homo sapiens 82-86 34384932-6 2021 The expression of downstream genes, nuclear respiratory factors 1 and 2 (NRF1 and NRF2, respectively), and mitochondrial transcription factor A (TFAM) were decreased in C2C12 myoblasts upon overexpression of miR-128. mir-128 208-215 transcription factor A, mitochondrial Homo sapiens 107-143 34384932-6 2021 The expression of downstream genes, nuclear respiratory factors 1 and 2 (NRF1 and NRF2, respectively), and mitochondrial transcription factor A (TFAM) were decreased in C2C12 myoblasts upon overexpression of miR-128. mir-128 208-215 transcription factor A, mitochondrial Homo sapiens 145-149 34384932-7 2021 Also, miR-128 is shown to promote mitochondrial dysfunction by directly targeting NADH Dehydrogenase (Ubiquinone) Fe-S Protein 4 (NDUFS4). mir-128 6-13 NADH:ubiquinone oxidoreductase subunit S4 Homo sapiens 130-136 34384932-8 2021 The mitochondrial dynamics and morphology were impaired post miR-128 overexpression, as revealed by downregulation of fusion proteins (mitofusin1 and 2, i.e., MFN1 and MFN2, respectively) and upregulation of fission protein (dynamin-related protein 1, i.e., DRP1). mir-128 61-68 mitofusin 1 Homo sapiens 135-151 34384932-8 2021 The mitochondrial dynamics and morphology were impaired post miR-128 overexpression, as revealed by downregulation of fusion proteins (mitofusin1 and 2, i.e., MFN1 and MFN2, respectively) and upregulation of fission protein (dynamin-related protein 1, i.e., DRP1). mir-128 61-68 mitofusin 1 Homo sapiens 159-163 34384932-8 2021 The mitochondrial dynamics and morphology were impaired post miR-128 overexpression, as revealed by downregulation of fusion proteins (mitofusin1 and 2, i.e., MFN1 and MFN2, respectively) and upregulation of fission protein (dynamin-related protein 1, i.e., DRP1). mir-128 61-68 mitofusin 2 Homo sapiens 168-172 34384932-8 2021 The mitochondrial dynamics and morphology were impaired post miR-128 overexpression, as revealed by downregulation of fusion proteins (mitofusin1 and 2, i.e., MFN1 and MFN2, respectively) and upregulation of fission protein (dynamin-related protein 1, i.e., DRP1). mir-128 61-68 dynamin 1 like Homo sapiens 225-250 34384932-8 2021 The mitochondrial dynamics and morphology were impaired post miR-128 overexpression, as revealed by downregulation of fusion proteins (mitofusin1 and 2, i.e., MFN1 and MFN2, respectively) and upregulation of fission protein (dynamin-related protein 1, i.e., DRP1). mir-128 61-68 dynamin 1 like Homo sapiens 258-262 34108447-9 2021 Furthermore, PHF6 overexpression affected IEC-6 cells by activating PI3K/AKT signaling which was mediated by the miR-128-3p/PHF6 axis. mir-128 113-120 PHD finger protein 6 Rattus norvegicus 13-17 34108447-9 2021 Furthermore, PHF6 overexpression affected IEC-6 cells by activating PI3K/AKT signaling which was mediated by the miR-128-3p/PHF6 axis. mir-128 113-120 AKT serine/threonine kinase 1 Rattus norvegicus 73-76 34108447-9 2021 Furthermore, PHF6 overexpression affected IEC-6 cells by activating PI3K/AKT signaling which was mediated by the miR-128-3p/PHF6 axis. mir-128 113-120 PHD finger protein 6 Rattus norvegicus 124-128 34646427-0 2021 Downregulation of miR-128 Ameliorates Ang II-Induced Cardiac Remodeling via SIRT1/PIK3R1 Multiple Targets. mir-128 18-25 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 38-44 34646427-0 2021 Downregulation of miR-128 Ameliorates Ang II-Induced Cardiac Remodeling via SIRT1/PIK3R1 Multiple Targets. mir-128 18-25 sirtuin 1 Mus musculus 76-81 34646427-0 2021 Downregulation of miR-128 Ameliorates Ang II-Induced Cardiac Remodeling via SIRT1/PIK3R1 Multiple Targets. mir-128 18-25 phosphoinositide-3-kinase regulatory subunit 1 Mus musculus 82-88 34646427-3 2021 The aim of the present study was to investigate the role of miR-128 in chronic angiotensin II (Ang II) infusion-induced cardiac remodeling and its underlying mechanism. mir-128 60-67 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 79-93 34646427-3 2021 The aim of the present study was to investigate the role of miR-128 in chronic angiotensin II (Ang II) infusion-induced cardiac remodeling and its underlying mechanism. mir-128 60-67 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 95-101 34646427-7 2021 Our results showed that chronic Ang II delivery for 28 days induced cardiac dysfunction, hypertrophy, fibrosis, apoptosis, and oxidative stress in the mice, while the miR-128 expression was notably enhanced in the left ventricle. mir-128 167-174 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 32-38 34646427-8 2021 Silencing miR-128 in the hearts of mice ameliorated Ang II-induced cardiac dysfunction, hypertrophy, fibrosis apoptosis, and oxidative stress injury. mir-128 10-17 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 52-58 34646427-9 2021 Moreover, Ang II induced excessive autophagy in the mouse hearts, which was suppressed by miR-128 knockdown. mir-128 90-97 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 10-16 34646427-10 2021 In cultured cells, Ang II treatment induced a marked elevation in the miR-128 expression. mir-128 70-77 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 19-25 34646427-11 2021 Downregulation of miR-128 in the cells by transfection with miR-128 antagomir attenuated Ang II-induced apoptosis and oxidative injury probably via directly targeting on the SIRT1/p53 pathway. mir-128 18-25 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 89-95 34646427-11 2021 Downregulation of miR-128 in the cells by transfection with miR-128 antagomir attenuated Ang II-induced apoptosis and oxidative injury probably via directly targeting on the SIRT1/p53 pathway. mir-128 18-25 sirtuin 1 Mus musculus 174-179 34646427-11 2021 Downregulation of miR-128 in the cells by transfection with miR-128 antagomir attenuated Ang II-induced apoptosis and oxidative injury probably via directly targeting on the SIRT1/p53 pathway. mir-128 18-25 transformation related protein 53, pseudogene Mus musculus 180-183 34646427-11 2021 Downregulation of miR-128 in the cells by transfection with miR-128 antagomir attenuated Ang II-induced apoptosis and oxidative injury probably via directly targeting on the SIRT1/p53 pathway. mir-128 60-67 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 89-95 34646427-11 2021 Downregulation of miR-128 in the cells by transfection with miR-128 antagomir attenuated Ang II-induced apoptosis and oxidative injury probably via directly targeting on the SIRT1/p53 pathway. mir-128 60-67 sirtuin 1 Mus musculus 174-179 34646427-11 2021 Downregulation of miR-128 in the cells by transfection with miR-128 antagomir attenuated Ang II-induced apoptosis and oxidative injury probably via directly targeting on the SIRT1/p53 pathway. mir-128 60-67 transformation related protein 53, pseudogene Mus musculus 180-183 34646427-12 2021 Intriguingly, we found that miR-128 inhibition activated PIK3R1/Akt/mTOR pathway and thereby significantly damped Ang II-stimulated pathological autophagy in cardiomyocytes, which consequently mitigated cell oxidative stress and apoptosis. mir-128 28-35 phosphoinositide-3-kinase regulatory subunit 1 Mus musculus 57-63 34646427-12 2021 Intriguingly, we found that miR-128 inhibition activated PIK3R1/Akt/mTOR pathway and thereby significantly damped Ang II-stimulated pathological autophagy in cardiomyocytes, which consequently mitigated cell oxidative stress and apoptosis. mir-128 28-35 thymoma viral proto-oncogene 1 Mus musculus 64-67 34646427-12 2021 Intriguingly, we found that miR-128 inhibition activated PIK3R1/Akt/mTOR pathway and thereby significantly damped Ang II-stimulated pathological autophagy in cardiomyocytes, which consequently mitigated cell oxidative stress and apoptosis. mir-128 28-35 mechanistic target of rapamycin kinase Mus musculus 68-72 34646427-12 2021 Intriguingly, we found that miR-128 inhibition activated PIK3R1/Akt/mTOR pathway and thereby significantly damped Ang II-stimulated pathological autophagy in cardiomyocytes, which consequently mitigated cell oxidative stress and apoptosis. mir-128 28-35 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 114-120 34646427-13 2021 In conclusion, downregulation of miR-128 ameliorates Ang II-provoked cardiac oxidative stress, hypertrophy, fibrosis, apoptosis, and dysfunction in mice, likely through targeting on PIK3R1/Akt/mTORC1 and/or SIRT1/p53 pathways. mir-128 33-40 angiotensinogen (serpin peptidase inhibitor, clade A, member 8) Mus musculus 53-59 34646427-13 2021 In conclusion, downregulation of miR-128 ameliorates Ang II-provoked cardiac oxidative stress, hypertrophy, fibrosis, apoptosis, and dysfunction in mice, likely through targeting on PIK3R1/Akt/mTORC1 and/or SIRT1/p53 pathways. mir-128 33-40 phosphoinositide-3-kinase regulatory subunit 1 Mus musculus 182-188 34646427-13 2021 In conclusion, downregulation of miR-128 ameliorates Ang II-provoked cardiac oxidative stress, hypertrophy, fibrosis, apoptosis, and dysfunction in mice, likely through targeting on PIK3R1/Akt/mTORC1 and/or SIRT1/p53 pathways. mir-128 33-40 thymoma viral proto-oncogene 1 Mus musculus 189-192 34646427-13 2021 In conclusion, downregulation of miR-128 ameliorates Ang II-provoked cardiac oxidative stress, hypertrophy, fibrosis, apoptosis, and dysfunction in mice, likely through targeting on PIK3R1/Akt/mTORC1 and/or SIRT1/p53 pathways. mir-128 33-40 CREB regulated transcription coactivator 1 Mus musculus 193-199 34646427-13 2021 In conclusion, downregulation of miR-128 ameliorates Ang II-provoked cardiac oxidative stress, hypertrophy, fibrosis, apoptosis, and dysfunction in mice, likely through targeting on PIK3R1/Akt/mTORC1 and/or SIRT1/p53 pathways. mir-128 33-40 sirtuin 1 Mus musculus 207-212 34646427-13 2021 In conclusion, downregulation of miR-128 ameliorates Ang II-provoked cardiac oxidative stress, hypertrophy, fibrosis, apoptosis, and dysfunction in mice, likely through targeting on PIK3R1/Akt/mTORC1 and/or SIRT1/p53 pathways. mir-128 33-40 transformation related protein 53, pseudogene Mus musculus 213-216 34296305-0 2021 Upregulation of miR-128 inhibits neuronal cell apoptosis following spinal cord injury via FasL downregulation by repressing ULK1. mir-128 16-23 unc-51 like autophagy activating kinase 1 Rattus norvegicus 124-128 34296305-10 2021 Overexpression of miR-128 or downregulation of ULK1 inhibited neuronal cell apoptosis and inflammation as evidenced by an increased BBB score and more neurons and nestin-positive cells, but reduced expression of inflammatory and apoptosis-related factors. mir-128 18-25 nestin Rattus norvegicus 163-169 34296305-11 2021 ULK1 was negatively regulated by miR-128, whereas FasL was positively regulated by ULK1. mir-128 33-40 unc-51 like autophagy activating kinase 1 Rattus norvegicus 0-4 34296305-13 2021 In conclusion, the upregulation of miR-128 depresses neuronal cell apoptosis by downregulating ULK1, thereby attenuating SCI via the downregulation of FasL. mir-128 35-42 unc-51 like autophagy activating kinase 1 Rattus norvegicus 95-99 34296305-13 2021 In conclusion, the upregulation of miR-128 depresses neuronal cell apoptosis by downregulating ULK1, thereby attenuating SCI via the downregulation of FasL. mir-128 35-42 Fas ligand Rattus norvegicus 151-155 34553041-6 2021 In addition, BMI-1, ABCC-5, E2F3, and c-MET were identified as candidate targets of miR-128, and the overexpression of miR-128 significantly reduced mRNA/protein levels of BMI-1, ABCC-5, E2F3, and c-MET in A549 and H460 cells. mir-128 84-91 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 13-18 34553041-6 2021 In addition, BMI-1, ABCC-5, E2F3, and c-MET were identified as candidate targets of miR-128, and the overexpression of miR-128 significantly reduced mRNA/protein levels of BMI-1, ABCC-5, E2F3, and c-MET in A549 and H460 cells. mir-128 84-91 ATP binding cassette subfamily C member 5 Homo sapiens 20-26 34553041-6 2021 In addition, BMI-1, ABCC-5, E2F3, and c-MET were identified as candidate targets of miR-128, and the overexpression of miR-128 significantly reduced mRNA/protein levels of BMI-1, ABCC-5, E2F3, and c-MET in A549 and H460 cells. mir-128 84-91 E2F transcription factor 3 Homo sapiens 28-32 34553041-6 2021 In addition, BMI-1, ABCC-5, E2F3, and c-MET were identified as candidate targets of miR-128, and the overexpression of miR-128 significantly reduced mRNA/protein levels of BMI-1, ABCC-5, E2F3, and c-MET in A549 and H460 cells. mir-128 84-91 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 38-43 34553041-6 2021 In addition, BMI-1, ABCC-5, E2F3, and c-MET were identified as candidate targets of miR-128, and the overexpression of miR-128 significantly reduced mRNA/protein levels of BMI-1, ABCC-5, E2F3, and c-MET in A549 and H460 cells. mir-128 84-91 E2F transcription factor 3 Homo sapiens 187-191 34553041-6 2021 In addition, BMI-1, ABCC-5, E2F3, and c-MET were identified as candidate targets of miR-128, and the overexpression of miR-128 significantly reduced mRNA/protein levels of BMI-1, ABCC-5, E2F3, and c-MET in A549 and H460 cells. mir-128 84-91 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 197-202 34124052-4 2021 Furthermore, we identified Ppp1cc as a direct target gene of miR-128 regulated by the PERK signaling pathway and showed that its repression is critical for a feedback loop that regulates the activity of UPR-associated signaling pathways, leading to cell migration, cell fusion, endoplasmic reticulum expansion, and myotube formation during myoblast differentiation. mir-128 61-68 protein phosphatase 1 catalytic subunit gamma Mus musculus 27-33 34124052-4 2021 Furthermore, we identified Ppp1cc as a direct target gene of miR-128 regulated by the PERK signaling pathway and showed that its repression is critical for a feedback loop that regulates the activity of UPR-associated signaling pathways, leading to cell migration, cell fusion, endoplasmic reticulum expansion, and myotube formation during myoblast differentiation. mir-128 61-68 eukaryotic translation initiation factor 2 alpha kinase 3 Mus musculus 86-90 35358540-6 2022 Finally, functional assays carried out in vitro proved SNHG16 could modulate GIST cell proliferation, migration, invasion and apoptosis via miR-128-3p/CASC3 axis. mir-128 140-147 small nucleolar RNA host gene 16 Homo sapiens 55-61 35587369-10 2022 These results suggested that KCNQ1OT1 regulates the osteoclast differentiation via the miR-128-3p/NFAT5 axis. mir-128 87-94 KCNQ1 overlapping transcript 1 Mus musculus 29-37 35587369-10 2022 These results suggested that KCNQ1OT1 regulates the osteoclast differentiation via the miR-128-3p/NFAT5 axis. mir-128 87-94 nuclear factor of activated T cells 5 Mus musculus 98-103 35252056-13 2022 Conclusion: The present results indicate that the miR-128-3p/c-Met axis may be potential therapeutic targets for circumventing lenvatinib resistance in HCC and warrant further investigation. mir-128 50-57 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 61-66 33994812-14 2021 MiR-128 was predicted to be a target of HCP5. mir-128 0-7 HLA complex P5 Homo sapiens 40-44 35142579-6 2022 NG-R1 treatment reduced miR-128-2-5p expression in the lung tissue, increased Tollip expression, inhibited HMGB1, TLR4, TRAF6, IRAK1, MyD88, NF-kappaB65, and p-IkappaB-alpha expression levels, suppressed NF-kappaB65 and the TLR4 expression levels, reduced MPO activity, reduced TNF-alpha, IL-1beta, IL-6, and ICAM-1 levels in BALF, and alleviated SAP-induced ALI. mir-128 24-31 reticulon 4 receptor Rattus norvegicus 0-5 35082915-0 2022 lncRNA-NEAT1 Sponges miR-128 to Promote Inflammatory Reaction and Phenotypic Transformation of Airway Smooth Muscle Cells. mir-128 21-28 nuclear paraspeckle assembly transcript 1 Homo sapiens 7-12 35082915-12 2022 Conclusion: lncRNA-NEAT1 sponges miR-128 to boost PDGF-BB-induced inflammatory reaction and phenotypic transformation of ASMCs to aggravate the occurrence and development of childhood bronchial asthma. mir-128 33-40 nuclear paraspeckle assembly transcript 1 Homo sapiens 19-24 35059433-0 2021 Exosome-Transmitted miR-128 Targets CCL18 to Inhibit the Proliferation and Metastasis of Urothelial Carcinoma. mir-128 20-27 C-C motif chemokine ligand 18 Homo sapiens 36-41 35059433-10 2021 The exosome-transmitted miR-128 can inhibit cell proliferation (p < 0.05), invasion (p < 0.05), and migration (p < 0.05) in UCs, and these effects can be reversed by CCL18. mir-128 24-31 C-C motif chemokine ligand 18 Homo sapiens 166-171 35059433-11 2021 In terms of apoptosis, miR-128 was able to promote the occurrence of BUC T24 apoptosis (p < 0.05), which can also be reversed by CCL18. mir-128 23-30 C-C motif chemokine ligand 18 Homo sapiens 129-134 35059433-13 2021 Conclusion: The miR-128 inhibits the proliferation, invasion, migration of UCs, and promotes its apoptosis by regulating CCL18 secretion. mir-128 16-23 C-C motif chemokine ligand 18 Homo sapiens 121-126 35000531-7 2022 Meanwhile, CCL2 down-regulating significantly repressed the proliferation, migration, and invasion by regulating miR-128. mir-128 113-120 C-C motif chemokine ligand 2 Homo sapiens 11-15 35000531-8 2022 In addition, we proved miR-128 was a direct target of CCL2 through double luciferase assay and bioinformatics analysis. mir-128 23-30 C-C motif chemokine ligand 2 Homo sapiens 54-58 35000531-10 2022 More importantly, miR-128 could reverse the effects of lncRNA CCL2 knocked down. mir-128 18-25 C-C motif chemokine ligand 2 Homo sapiens 62-66 35000531-12 2022 Meanwhile, miR-128 mimic and the knockout of CCL2 distinctly decreased PARP2 protein level. mir-128 11-18 poly(ADP-ribose) polymerase 2 Homo sapiens 71-76 35000531-13 2022 Additionally, luciferase report experiments certificated that PARP2 targeted miR-128, implying PARP2 directly interacted with miR-128 in gastric cancer. mir-128 77-84 poly(ADP-ribose) polymerase 2 Homo sapiens 62-67 35000531-13 2022 Additionally, luciferase report experiments certificated that PARP2 targeted miR-128, implying PARP2 directly interacted with miR-128 in gastric cancer. mir-128 77-84 poly(ADP-ribose) polymerase 2 Homo sapiens 95-100 35000531-13 2022 Additionally, luciferase report experiments certificated that PARP2 targeted miR-128, implying PARP2 directly interacted with miR-128 in gastric cancer. mir-128 126-133 poly(ADP-ribose) polymerase 2 Homo sapiens 62-67 35000531-13 2022 Additionally, luciferase report experiments certificated that PARP2 targeted miR-128, implying PARP2 directly interacted with miR-128 in gastric cancer. mir-128 126-133 poly(ADP-ribose) polymerase 2 Homo sapiens 95-100 35000531-14 2022 More interestingly, the downregulation of PARP could reverse the trend triggered by miR-128 inhibitor in gastric tumor. mir-128 84-91 collagen type XI alpha 2 chain Homo sapiens 42-46 33750005-9 2021 miR-128 has been reported to inhibit dendritic growth and branching in mouse brain neurons, which directly opposes the novel functions of R3HDM1. mir-128 0-7 R3H domain containing 1 Mus musculus 138-144 33743142-0 2021 B7-H3, Negatively Regulated by miR-128, Promotes Colorectal Cancer Cell Proliferation and Migration. mir-128 31-38 CD276 antigen Mus musculus 0-5 33743142-8 2021 Besides, the regulatory relationship between miR-128 and B7-H3 was validated through dual-luciferase reporter gene assay, qRT-PCR, and western blotting. mir-128 45-52 CD276 antigen Mus musculus 57-62 33743142-11 2021 Moreover, B7-H3 was identified as a target of miR-128, and miR-128 negatively regulated B7-H3 expression in CRC cells. mir-128 46-53 CD276 antigen Mus musculus 10-15 33743142-11 2021 Moreover, B7-H3 was identified as a target of miR-128, and miR-128 negatively regulated B7-H3 expression in CRC cells. mir-128 59-66 CD276 antigen Mus musculus 88-93 33743142-13 2021 Besides, B7-H3 expression is negatively regulated by miR-128 in CRC. mir-128 53-60 CD276 antigen Mus musculus 9-14 34047500-0 2021 Mechanism of ARPP21 antagonistic intron miR-128 on neurological function repair after stroke. mir-128 40-47 cAMP regulated phosphoprotein 21 Rattus norvegicus 13-19 34047500-2 2021 ARPP21 is a conserved host gene of miR-128 controlling neurodevelopmental functions. mir-128 35-42 cAMP regulated phosphoprotein 21 Rattus norvegicus 0-6 34047500-3 2021 This study investigated the mechanism of ARPP21 antagonistic intron miR-128 on neurological function repair after stroke. mir-128 68-75 cAMP regulated phosphoprotein 21 Rattus norvegicus 41-47 34047500-8 2021 The binding relationship between miR-128 and CREB1 was verified. mir-128 33-40 cAMP responsive element binding protein 1 Mus musculus 45-50 34047500-14 2021 miR-128 mimic could reverse these effects of pc-ARPP21 on neurons and astrocytes. mir-128 0-7 cAMP regulated phosphoprotein 21 Homo sapiens 48-54 34047500-15 2021 miR-128 targeted CREB1 and reduced BDNF secretion. mir-128 0-7 cAMP responsive element binding protein 1 Homo sapiens 17-22 34047500-15 2021 miR-128 targeted CREB1 and reduced BDNF secretion. mir-128 0-7 brain derived neurotrophic factor Homo sapiens 35-39 34047500-17 2021 CONCLUSION: ARPP21 upregulated CREB1 and BDNF expressions by antagonizing miR-128, thus inhibiting neuronal apoptosis and promoting neurological function repair after stroke. mir-128 74-81 cAMP regulated phosphoprotein 21 Rattus norvegicus 12-18 34047500-17 2021 CONCLUSION: ARPP21 upregulated CREB1 and BDNF expressions by antagonizing miR-128, thus inhibiting neuronal apoptosis and promoting neurological function repair after stroke. mir-128 74-81 cAMP responsive element binding protein 1 Rattus norvegicus 31-36 34047500-17 2021 CONCLUSION: ARPP21 upregulated CREB1 and BDNF expressions by antagonizing miR-128, thus inhibiting neuronal apoptosis and promoting neurological function repair after stroke. mir-128 74-81 brain-derived neurotrophic factor Rattus norvegicus 41-45 33993846-5 2021 miR-128 was decreased in GC patients and sponged by HCP5. mir-128 0-7 HLA complex P5 Homo sapiens 52-56 33993846-6 2021 HMGA2 was targeted by miR-128 and was increased in GC patients. mir-128 22-29 high mobility group AT-hook 2 Homo sapiens 0-5 33993846-7 2021 HCP5 aggravated the resistance of GC cells to DDP in vitro by elevating HMGA2 expression via sponging miR-128. mir-128 102-109 HLA complex P5 Homo sapiens 0-4 33993846-7 2021 HCP5 aggravated the resistance of GC cells to DDP in vitro by elevating HMGA2 expression via sponging miR-128. mir-128 102-109 high mobility group AT-hook 2 Homo sapiens 72-77 33994812-15 2021 It was demonstrated that HCP5 directly bound to miR-128 and regulated its expression in glioma cells. mir-128 48-55 HLA complex P5 Homo sapiens 25-29 33994812-16 2021 Furthermore, the effects of HCP5 knockdown on radiosensitivity of glioma cells were attenuated by the inhibitor of miR-128. mir-128 115-122 HLA complex P5 Homo sapiens 28-32 33986789-13 2021 Conclusions: miR-128-3p inhibition attenuated DOX-related acute cardiac injury via the regulation of PPAR-gamma in mice. mir-128 13-20 peroxisome proliferator activated receptor gamma Mus musculus 101-111 33826315-4 2021 miR-128 can directly target PPARGC1A and inhibit its expression. mir-128 0-7 PPARG coactivator 1 alpha Bos taurus 28-36 33622342-9 2021 Moreover, miR-128 mimics partially abrogated the effect of LINC00477 on granulosa cells. mir-128 10-17 long intergenic non-protein coding RNA 477 Homo sapiens 59-68 33848670-13 2021 Knockdown of miR-128 restored the altered proliferation, migration and invasion and the expression of ZEB1 and E-cadherin caused by knockdown of PVT1. mir-128 13-20 zinc finger E-box binding homeobox 1 Homo sapiens 102-106 33848670-13 2021 Knockdown of miR-128 restored the altered proliferation, migration and invasion and the expression of ZEB1 and E-cadherin caused by knockdown of PVT1. mir-128 13-20 cadherin 1 Homo sapiens 111-121 33848670-13 2021 Knockdown of miR-128 restored the altered proliferation, migration and invasion and the expression of ZEB1 and E-cadherin caused by knockdown of PVT1. mir-128 13-20 Pvt1 oncogene Homo sapiens 145-149 33746773-9 2021 Either overexpression of miR-128 or downregulation of beta-catenin by CWP232228, a beta-catenin-specific antagonist, blocked the promoting roles of ETS1 in cells. mir-128 25-32 E26 avian leukemia oncogene 1, 5' domain Mus musculus 148-152 33746773-10 2021 To conclude, this study provided evidence that ETS1 suppresses miR-128 transcription to activate the following HOXA13/beta-catenin axis, therefore promoting osteogenic differentiation ability of MC3T3-E1 cells. mir-128 63-70 E26 avian leukemia oncogene 1, 5' domain Mus musculus 47-51 33746773-10 2021 To conclude, this study provided evidence that ETS1 suppresses miR-128 transcription to activate the following HOXA13/beta-catenin axis, therefore promoting osteogenic differentiation ability of MC3T3-E1 cells. mir-128 63-70 homeobox A13 Mus musculus 111-117 33746773-10 2021 To conclude, this study provided evidence that ETS1 suppresses miR-128 transcription to activate the following HOXA13/beta-catenin axis, therefore promoting osteogenic differentiation ability of MC3T3-E1 cells. mir-128 63-70 catenin (cadherin associated protein), beta 1 Mus musculus 118-130 33347907-5 2021 In this study, miR-128 was identified as a negative regulator to participate in the NF-kappaB signaling pathway by targeting TAB2 in miiuy croaker. mir-128 15-22 nuclear factor kappa B subunit 1 Homo sapiens 84-93 33347907-9 2021 Particularly, we found that miR-128 can regulate TAB2-mediated NF-kappaB signaling pathways. mir-128 28-35 nuclear factor kappa B subunit 1 Homo sapiens 63-72 33347907-10 2021 In summary, our results indicate that miR-128 plays a critical role in suppressing inflammatory responses by regulating the TAB2-mediated NF-kappaB signaling pathway in miiuy croaker. mir-128 38-45 nuclear factor kappa B subunit 1 Homo sapiens 138-147 32772776-0 2021 miR-128 regulated the proliferation and autophagy in porcine adipose-derived stem cells through targeting the JNK signaling pathway. mir-128 0-7 mitogen-activated protein kinase 8 Homo sapiens 110-113 32772776-7 2021 However, followed by the block of the JNK signaling pathway using SP600125 inhibitor, the effects of miR-128 on the proliferation, apoptosis and autophagy of porcine ASCs were significantly suppressed. mir-128 101-108 mitogen-activated protein kinase 8 Homo sapiens 38-41 32772776-8 2021 CONCLUSION: It is demonstrated that the miR-128-JNK signaling pathway is a potential therapeutic target for the treatment of obesity. mir-128 40-47 mitogen-activated protein kinase 8 Homo sapiens 48-51 33571908-0 2021 miR-128 regulates epilepsy sensitivity in mice by suppressing SNAP-25 and SYT1 expression in the hippocampus. mir-128 0-7 synaptosomal-associated protein 25 Mus musculus 62-69 33571908-0 2021 miR-128 regulates epilepsy sensitivity in mice by suppressing SNAP-25 and SYT1 expression in the hippocampus. mir-128 0-7 synaptotagmin I Mus musculus 74-78 33571908-3 2021 Both SNAP-25 and SYT1 are regulated by miR-128 in vitro and in vivo. mir-128 39-46 synaptosomal-associated protein 25 Mus musculus 5-12 33571908-3 2021 Both SNAP-25 and SYT1 are regulated by miR-128 in vitro and in vivo. mir-128 39-46 synaptotagmin I Mus musculus 17-21 33571908-4 2021 Overexpressing miR-128 in cultured neurons decreased neurotransmitter released by suppressing SNAP-25 and SYT1 expression. mir-128 15-22 synaptosomal-associated protein 25 Mus musculus 94-101 33571908-4 2021 Overexpressing miR-128 in cultured neurons decreased neurotransmitter released by suppressing SNAP-25 and SYT1 expression. mir-128 15-22 synaptotagmin I Mus musculus 106-110 33622342-10 2021 CONCLUSION: LINC00477 may function as a ceRNA to inhibit proliferation and apoptosis of granulosa cells by modulating miR-128 expression. mir-128 118-125 long intergenic non-protein coding RNA 477 Homo sapiens 12-21 33379338-10 2020 Notably, miR-128 mediated SSR-enhanced HeLa and CaSki cells" adhesion and metastasis through suppressed ITGA5, ITGB5, sLex, CEACAM-6, MMP9, and MMP23 transcript levels. mir-128 9-16 integrin subunit alpha 5 Homo sapiens 104-109 33542630-10 2021 The serum miR-128 levels in AD patients were positively correlated with the serum IL-1beta (r=0.798, P<0.01) and serum TNF-alpha levels (r=0.733, P<0.01). mir-128 10-17 interleukin 1 alpha Homo sapiens 82-90 33542630-10 2021 The serum miR-128 levels in AD patients were positively correlated with the serum IL-1beta (r=0.798, P<0.01) and serum TNF-alpha levels (r=0.733, P<0.01). mir-128 10-17 tumor necrosis factor Homo sapiens 119-128 33472642-6 2021 Genetic knockdown of miR-128 in bone neoplasms cells suppressed the activation of the Wnt/beta-catenin and epithelial-mesenchymal transition (EMT) signaling pathways. mir-128 21-28 catenin beta 1 Homo sapiens 90-102 33472642-9 2021 CONCLUSIONS: These results define a tumor-regulated function for miR-128 in bone neoplasms by down-regulation of the Wnt/beta-catenin and EMT signal pathways, which provided a potential target for bone neoplasms gene therapy. mir-128 65-72 catenin beta 1 Homo sapiens 121-133 33080252-5 2021 Computational analysis identified the transcription factor EYA4 as a candidate target consistent with the observation that miR-128 and EYA4 display similar expression profiles. mir-128 123-130 EYA transcriptional coactivator and phosphatase 4 Gallus gallus 59-63 33080252-7 2021 In vivo experiments also suggest that EYA4 is regulated by miR-128. mir-128 59-66 EYA transcriptional coactivator and phosphatase 4 Gallus gallus 38-42 33080252-11 2021 Our results reveal that miR-128 is involved in regulating skeletal myogenesis by directly targeting EYA4 with indirect effects on other PSED members, including SIX4 and PAX3. mir-128 24-31 EYA transcriptional coactivator and phosphatase 4 Gallus gallus 100-104 33080252-11 2021 Our results reveal that miR-128 is involved in regulating skeletal myogenesis by directly targeting EYA4 with indirect effects on other PSED members, including SIX4 and PAX3. mir-128 24-31 SIX homeobox 5 Gallus gallus 160-164 33080252-11 2021 Our results reveal that miR-128 is involved in regulating skeletal myogenesis by directly targeting EYA4 with indirect effects on other PSED members, including SIX4 and PAX3. mir-128 24-31 paired box 3 Gallus gallus 169-173 33080252-12 2021 Hence, the inhibitory effect on myogenesis observed after miR-128 knockdown was rescued by concomitant knockdown of PAX3. mir-128 58-65 paired box 3 Gallus gallus 116-120 33379338-10 2020 Notably, miR-128 mediated SSR-enhanced HeLa and CaSki cells" adhesion and metastasis through suppressed ITGA5, ITGB5, sLex, CEACAM-6, MMP9, and MMP23 transcript levels. mir-128 9-16 integrin subunit beta 5 Homo sapiens 111-116 33379338-10 2020 Notably, miR-128 mediated SSR-enhanced HeLa and CaSki cells" adhesion and metastasis through suppressed ITGA5, ITGB5, sLex, CEACAM-6, MMP9, and MMP23 transcript levels. mir-128 9-16 CEA cell adhesion molecule 6 Homo sapiens 124-132 33379338-10 2020 Notably, miR-128 mediated SSR-enhanced HeLa and CaSki cells" adhesion and metastasis through suppressed ITGA5, ITGB5, sLex, CEACAM-6, MMP9, and MMP23 transcript levels. mir-128 9-16 matrix metallopeptidase 9 Homo sapiens 134-138 33379338-10 2020 Notably, miR-128 mediated SSR-enhanced HeLa and CaSki cells" adhesion and metastasis through suppressed ITGA5, ITGB5, sLex, CEACAM-6, MMP9, and MMP23 transcript levels. mir-128 9-16 matrix metallopeptidase 23B Homo sapiens 144-149 33167678-6 2020 Direct binding of miR-128 to PVT1 was investigated using a probe pulldown assay. mir-128 18-25 Pvt1 oncogene Mus musculus 29-33 33167678-10 2020 Mechanistically, we found that PVT1 competes endogenously with miR-128 in the regulation of vascular endothelial growth factor C (VEGFC) expression, which is significantly associated with an unfavorable prognosis in lung cancer. mir-128 63-70 vascular endothelial growth factor C Homo sapiens 92-128 33167678-10 2020 Mechanistically, we found that PVT1 competes endogenously with miR-128 in the regulation of vascular endothelial growth factor C (VEGFC) expression, which is significantly associated with an unfavorable prognosis in lung cancer. mir-128 63-70 vascular endothelial growth factor C Homo sapiens 130-135 33167678-11 2020 We identified that copy number amplification significantly contributes to the high level of PVT1 transcripts in lung cancer, which promotes cell proliferation and metastatic behavior via modulating VEGFC expression by endogenous competition with miR-128. mir-128 246-253 Pvt1 oncogene Homo sapiens 92-96 33167678-11 2020 We identified that copy number amplification significantly contributes to the high level of PVT1 transcripts in lung cancer, which promotes cell proliferation and metastatic behavior via modulating VEGFC expression by endogenous competition with miR-128. mir-128 246-253 vascular endothelial growth factor C Homo sapiens 198-203 33116855-10 2020 HOXA10 as the target of miR-128/miR-142 was verified in OSCC cells. mir-128 24-31 homeobox A10 Homo sapiens 0-6 33116617-13 2020 GOLM1 was a target of miR-128 and was negatively regulated by miR-128. mir-128 22-29 golgi membrane protein 1 Mus musculus 0-5 33116617-13 2020 GOLM1 was a target of miR-128 and was negatively regulated by miR-128. mir-128 62-69 golgi membrane protein 1 Mus musculus 0-5 33116855-13 2020 Conclusion: MiR-128/miR-142 suppressed OSCC tumorigenesis and metastasis by targeting HOXA10, providing a new promising therapeutic approach for OSCC patient diagnosis and treatment. mir-128 12-19 homeobox A10 Homo sapiens 86-92 32993809-0 2020 MiR-128 suppresses metastatic capacity by targeting metadherin in breast cancer cells. mir-128 0-7 metadherin Homo sapiens 52-62 32993809-9 2020 Moreover, knockdown of MTDH in MDA-MB-231 cells obviously impaired the migration and invasion capabilities, whereas re-expression of MTDH abrogated the suppressive effect caused by miR-128. mir-128 181-188 metadherin Homo sapiens 133-137 32486927-8 2020 Moreover, overexpression of Rnd3 could reverse the activation of NF-kappaB caused by miR-128-3p agomir during liver I/R injury. mir-128 85-92 Rho family GTPase 3 Mus musculus 28-32 32958647-0 2020 Astroglial FMRP deficiency cell-autonomously up-regulates miR-128 and disrupts developmental astroglial mGluR5 signaling. mir-128 58-65 fragile X messenger ribonucleoprotein 1 Homo sapiens 11-15 32739259-0 2020 Obesity-related hypoxia via miR-128 decreases insulin-receptor expression in human and mouse adipose tissue promoting systemic insulin resistance. mir-128 28-35 insulin receptor Homo sapiens 46-62 32739259-0 2020 Obesity-related hypoxia via miR-128 decreases insulin-receptor expression in human and mouse adipose tissue promoting systemic insulin resistance. mir-128 28-35 insulin Homo sapiens 46-53 33277837-0 2020 IRF-1 mediated long non-coding RNA PVT1-214 promotes oxaliplatin resistance of colorectal cancer via miR-128 inhibition. mir-128 101-108 interferon regulatory factor 1 Homo sapiens 0-5 32486927-8 2020 Moreover, overexpression of Rnd3 could reverse the activation of NF-kappaB caused by miR-128-3p agomir during liver I/R injury. mir-128 85-92 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 65-74 32218208-4 2020 The expression of PAICS was regulated by miR-128 and transcriptionally activated by Myc in CRC cells. mir-128 41-48 phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccinocarboxamide synthase Homo sapiens 18-23 32678046-12 2020 CONCLUSION: The high expression of miR-128 can down-regulate the expression of CB1, promote the activation of NF-KB and p-JNK, increase the level of apoptosis and promote the occurrence of diabetic bladder disease. mir-128 35-42 cannabinoid receptor 1 Rattus norvegicus 79-82 32678046-12 2020 CONCLUSION: The high expression of miR-128 can down-regulate the expression of CB1, promote the activation of NF-KB and p-JNK, increase the level of apoptosis and promote the occurrence of diabetic bladder disease. mir-128 35-42 RELA proto-oncogene, NF-kB subunit Rattus norvegicus 110-115 32678046-12 2020 CONCLUSION: The high expression of miR-128 can down-regulate the expression of CB1, promote the activation of NF-KB and p-JNK, increase the level of apoptosis and promote the occurrence of diabetic bladder disease. mir-128 35-42 mitogen-activated protein kinase 8 Rattus norvegicus 122-125 32536914-6 2020 miR-128 was inversely correlated with cluster of differentiation 47 (CD47), which was positively related to zinc finger E-box-binding homeobox 1 (ZEB1) in PDAC. mir-128 0-7 zinc finger E-box binding homeobox 1 Mus musculus 108-144 32536914-6 2020 miR-128 was inversely correlated with cluster of differentiation 47 (CD47), which was positively related to zinc finger E-box-binding homeobox 1 (ZEB1) in PDAC. mir-128 0-7 zinc finger E-box binding homeobox 1 Mus musculus 146-150 32222058-0 2020 Mossy cell synaptic dysfunction causes memory imprecision via miR-128 inhibition of STIM2 in Alzheimer"s disease mouse model. mir-128 62-69 stromal interaction molecule 2 Mus musculus 84-89 32222058-4 2020 Silencing miR-128 or disrupting miR-128 binding to STIM2 evokes STIM2 expression, restores synaptic function, and rescues memory imprecision in AD mice. mir-128 10-17 stromal interaction molecule 2 Mus musculus 64-69 32222058-4 2020 Silencing miR-128 or disrupting miR-128 binding to STIM2 evokes STIM2 expression, restores synaptic function, and rescues memory imprecision in AD mice. mir-128 32-39 stromal interaction molecule 2 Mus musculus 51-56 32222058-4 2020 Silencing miR-128 or disrupting miR-128 binding to STIM2 evokes STIM2 expression, restores synaptic function, and rescues memory imprecision in AD mice. mir-128 32-39 stromal interaction molecule 2 Mus musculus 64-69 32377170-12 2020 MIR4435-2HG knockdown inhibited proliferation, invasion and migration but induced apoptosis of OC cells via miR-128-3p/CDK14 axis. mir-128 108-115 MIR4435-2 host gene Homo sapiens 0-11 32678046-2 2020 In this study we investigate the mechanism of miR-128 targeting CB1 expression to mediate the occurrence of diabetic bladder disease. mir-128 46-53 cannabinoid receptor 1 Rattus norvegicus 64-67 32678046-9 2020 miR-128 could target the inhibition of CB1 expression, and high expression of CB1 could antagonize miR-128 against diabetic bladder. mir-128 0-7 cannabinoid receptor 1 Rattus norvegicus 39-42 32678046-9 2020 miR-128 could target the inhibition of CB1 expression, and high expression of CB1 could antagonize miR-128 against diabetic bladder. mir-128 99-106 cannabinoid receptor 1 Rattus norvegicus 78-81 32678046-10 2020 In the diabetic bladder, miR-128 can regulate the expression of NF-KB and p-JNK through CB1 and affect the level of apoptosis. mir-128 25-32 RELA proto-oncogene, NF-kB subunit Rattus norvegicus 64-69 32678046-10 2020 In the diabetic bladder, miR-128 can regulate the expression of NF-KB and p-JNK through CB1 and affect the level of apoptosis. mir-128 25-32 mitogen-activated protein kinase 8 Rattus norvegicus 76-79 32678046-10 2020 In the diabetic bladder, miR-128 can regulate the expression of NF-KB and p-JNK through CB1 and affect the level of apoptosis. mir-128 25-32 cannabinoid receptor 1 Rattus norvegicus 88-91 32678046-11 2020 miR-128 regulates NF-KB/p-JNK through CB1, thus affecting the occurrence of diabetic bladder disease. mir-128 0-7 RELA proto-oncogene, NF-kB subunit Rattus norvegicus 18-23 32678046-11 2020 miR-128 regulates NF-KB/p-JNK through CB1, thus affecting the occurrence of diabetic bladder disease. mir-128 0-7 mitogen-activated protein kinase 8 Rattus norvegicus 26-29 32678046-11 2020 miR-128 regulates NF-KB/p-JNK through CB1, thus affecting the occurrence of diabetic bladder disease. mir-128 0-7 cannabinoid receptor 1 Rattus norvegicus 38-41 32292498-11 2020 Results: The miR-128 level was found to be positively correlated with the increase in Nfatc1 level in mouse/human bone specimens and mouse primary BMMs. mir-128 13-20 nuclear factor of activated T cells, cytoplasmic, calcineurin dependent 1 Mus musculus 86-92 32292498-17 2020 Conclusions: Our findings reveal a critical mechanism for osteoclastogenesis that is mediated by the miR-128/SIRT1/NF-kappaB signaling axis, highlighting a possible avenue for the further exploration of diagnostic and therapeutic target molecules in PMOP. mir-128 101-108 sirtuin 1 Mus musculus 109-114 32292498-17 2020 Conclusions: Our findings reveal a critical mechanism for osteoclastogenesis that is mediated by the miR-128/SIRT1/NF-kappaB signaling axis, highlighting a possible avenue for the further exploration of diagnostic and therapeutic target molecules in PMOP. mir-128 101-108 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 115-124 32127947-8 2020 The next study indicated that lnc-SNHG16 knockdown markedly increased miR-128 level which is down-regulated in CC. mir-128 70-77 small nucleolar RNA host gene 16 Homo sapiens 34-40 32127947-9 2020 Moreover, miR-128 overexpression significantly inhibited proliferation, EMT process and tumor growth by directly targeting GSPT1 and WNT3A. mir-128 10-17 G1 to S phase transition 1 Homo sapiens 123-128 32127947-9 2020 Moreover, miR-128 overexpression significantly inhibited proliferation, EMT process and tumor growth by directly targeting GSPT1 and WNT3A. mir-128 10-17 Wnt family member 3A Homo sapiens 133-138 32127947-10 2020 Finally, lnc-SNHG16 activates but miR-128 inactivates the WNT/beta-catenin pathways in CC cells. mir-128 34-41 catenin beta 1 Homo sapiens 62-74 31037985-1 2019 MiR-128, one of the most enriched miRNAs in the human brain, has been reported to protect MCAO mice via inhibiting P38alpha MAPK. mir-128 0-7 mitogen-activated protein kinase 14 Mus musculus 115-123 31341054-3 2019 We have further determined that miR-128 represses the expression of all three TNPO proteins (transportins TNPO1, TNPO2, and TNPO3). mir-128 32-39 transportin 1 Homo sapiens 106-111 31341054-3 2019 We have further determined that miR-128 represses the expression of all three TNPO proteins (transportins TNPO1, TNPO2, and TNPO3). mir-128 32-39 transportin 2 Homo sapiens 113-118 31341054-3 2019 We have further determined that miR-128 represses the expression of all three TNPO proteins (transportins TNPO1, TNPO2, and TNPO3). mir-128 32-39 transportin 3 Homo sapiens 124-129 31341054-5 2019 Here, we report that type I interferon (IFN)-inducible miR-128 directly targets two sites in the TNPO3 mRNA, significantly downregulating TNPO3 mRNA and protein expression levels. mir-128 55-62 transportin 3 Homo sapiens 97-102 31341054-5 2019 Here, we report that type I interferon (IFN)-inducible miR-128 directly targets two sites in the TNPO3 mRNA, significantly downregulating TNPO3 mRNA and protein expression levels. mir-128 55-62 transportin 3 Homo sapiens 138-143 31341054-7 2019 Manipulation of miR-128 levels in HIV-1 target cell lines and in primary CD4+ T-cells by overexpression or knockdown showed that reduction of TNPO3 levels by miR-128 significantly affects HIV-1 replication but not murine leukemia virus (MLV) infection and that miR-128 modulation of HIV-1 replication is reduced with TNPO3-independent HIV-1 virus, suggesting that miR-128-indued TNPO3 repression contributes to the inhibition of HIV-1 replication. mir-128 16-23 transportin 3 Homo sapiens 142-147 31341054-7 2019 Manipulation of miR-128 levels in HIV-1 target cell lines and in primary CD4+ T-cells by overexpression or knockdown showed that reduction of TNPO3 levels by miR-128 significantly affects HIV-1 replication but not murine leukemia virus (MLV) infection and that miR-128 modulation of HIV-1 replication is reduced with TNPO3-independent HIV-1 virus, suggesting that miR-128-indued TNPO3 repression contributes to the inhibition of HIV-1 replication. mir-128 158-165 transportin 3 Homo sapiens 142-147 31341054-7 2019 Manipulation of miR-128 levels in HIV-1 target cell lines and in primary CD4+ T-cells by overexpression or knockdown showed that reduction of TNPO3 levels by miR-128 significantly affects HIV-1 replication but not murine leukemia virus (MLV) infection and that miR-128 modulation of HIV-1 replication is reduced with TNPO3-independent HIV-1 virus, suggesting that miR-128-indued TNPO3 repression contributes to the inhibition of HIV-1 replication. mir-128 158-165 transportin 3 Homo sapiens 142-147 31341054-7 2019 Manipulation of miR-128 levels in HIV-1 target cell lines and in primary CD4+ T-cells by overexpression or knockdown showed that reduction of TNPO3 levels by miR-128 significantly affects HIV-1 replication but not murine leukemia virus (MLV) infection and that miR-128 modulation of HIV-1 replication is reduced with TNPO3-independent HIV-1 virus, suggesting that miR-128-indued TNPO3 repression contributes to the inhibition of HIV-1 replication. mir-128 158-165 transportin 3 Homo sapiens 142-147 31341054-9 2019 Thus, we have established a novel role of miR-128 in antiviral defense in human cells, namely inhibiting HIV-1 replication by altering the cellular milieu through targeting factors that include TNPO3.IMPORTANCE HIV-1 is the causative agent of AIDS. mir-128 42-49 transportin 3 Homo sapiens 194-199 31477582-0 2019 MiR-128 inhibits the osteogenic differentiation in osteoporosis by down-regulating SIRT6 expression. mir-128 0-7 sirtuin 6 Mus musculus 83-88 31477582-4 2019 In addition, the results showed that overexpression of miR-128 significantly inhibited the mRNA and protein expression levels of osteocalcin (OC), alkaline phosphatase (ALP) and collagen I type-alpha1 (COL1A1) in C2C12 cells, while miR-128 inhibitor could reverse this effect. mir-128 55-62 bone gamma-carboxyglutamate protein 2 Mus musculus 129-140 31477582-4 2019 In addition, the results showed that overexpression of miR-128 significantly inhibited the mRNA and protein expression levels of osteocalcin (OC), alkaline phosphatase (ALP) and collagen I type-alpha1 (COL1A1) in C2C12 cells, while miR-128 inhibitor could reverse this effect. mir-128 55-62 collagen, type I, alpha 1 Mus musculus 202-208 31477582-4 2019 In addition, the results showed that overexpression of miR-128 significantly inhibited the mRNA and protein expression levels of osteocalcin (OC), alkaline phosphatase (ALP) and collagen I type-alpha1 (COL1A1) in C2C12 cells, while miR-128 inhibitor could reverse this effect. mir-128 232-239 bone gamma-carboxyglutamate protein 2 Mus musculus 129-140 31477582-6 2019 The qRT-PCR and Western Blot results found that miR-128 significantly down-regulated the mRNA and protein expressions of SIRT6. mir-128 48-55 sirtuin 6 Mus musculus 121-126 31477582-7 2019 Furthermore, silencing SIRT6 significantly inhibited the promoting effect of the miR-128 inhibitor on the expression of osteoblast markers.Conclusion: The above results confirmed that miR-128 inhibited osteoblast differentiation in OP by down-regulating SIRT6 expression, thus accelerating the development of OP. mir-128 81-88 sirtuin 6 Mus musculus 23-28 31477582-7 2019 Furthermore, silencing SIRT6 significantly inhibited the promoting effect of the miR-128 inhibitor on the expression of osteoblast markers.Conclusion: The above results confirmed that miR-128 inhibited osteoblast differentiation in OP by down-regulating SIRT6 expression, thus accelerating the development of OP. mir-128 81-88 sirtuin 6 Mus musculus 254-259 31477582-7 2019 Furthermore, silencing SIRT6 significantly inhibited the promoting effect of the miR-128 inhibitor on the expression of osteoblast markers.Conclusion: The above results confirmed that miR-128 inhibited osteoblast differentiation in OP by down-regulating SIRT6 expression, thus accelerating the development of OP. mir-128 184-191 sirtuin 6 Mus musculus 23-28 31477582-7 2019 Furthermore, silencing SIRT6 significantly inhibited the promoting effect of the miR-128 inhibitor on the expression of osteoblast markers.Conclusion: The above results confirmed that miR-128 inhibited osteoblast differentiation in OP by down-regulating SIRT6 expression, thus accelerating the development of OP. mir-128 184-191 sirtuin 6 Mus musculus 254-259 30701536-2 2019 This study was performed to clarify the effect of microRNA-128 (miR-128) on the CMEC injury in coronary heart disease (CHD) by binding to insulin receptor substrate 1 (IRS1). mir-128 64-71 insulin receptor substrate 1 Rattus norvegicus 138-166 30701536-2 2019 This study was performed to clarify the effect of microRNA-128 (miR-128) on the CMEC injury in coronary heart disease (CHD) by binding to insulin receptor substrate 1 (IRS1). mir-128 64-71 insulin receptor substrate 1 Rattus norvegicus 168-172 30701536-9 2019 Notably, miR-128 targeted and negatively regulated IRS1. mir-128 9-16 insulin receptor substrate 1 Rattus norvegicus 51-55 30701536-11 2019 The experimental results demonstrated that miR-128 mimics and IRS1 siRNA in rat CMECs promoted viability, migration ability, tube formation, VEGF expression, SOD activity, while repressing cell apoptosis, MDA and ROS levels. mir-128 43-50 vascular endothelial growth factor A Rattus norvegicus 141-145 31271703-6 2019 The association between miR-128 and HOXB8 was confirmed using dual-luciferase reporter assay. mir-128 24-31 homeobox B8 Homo sapiens 36-41 31271703-9 2019 miR-128 directly targeted HOXB8 in ovarian cancer cell lines. mir-128 0-7 homeobox B8 Homo sapiens 26-31 31271703-10 2019 Knockdown of HOXB8 abolished the effects of miR-128 inhibitor on ovarian cancer cell proliferation and paclitaxel sensitivity. mir-128 44-51 homeobox B8 Homo sapiens 13-18 31271703-11 2019 Summarily, miR-128 displayed a tumour suppressor role in ovarian cancer via targeting HOXB8. mir-128 11-18 homeobox B8 Homo sapiens 86-91 31352238-7 2019 Additionally, PCAT-1 functioned as a sponge of miR-128 in GC cells. mir-128 47-54 prostate cancer associated transcript 1 Homo sapiens 14-20 31352238-8 2019 Moreover, inhibition of miR-128 reversed the inductive effect of PCAT-1 knockdown on DDP sensitivity of GC cells. mir-128 24-31 prostate cancer associated transcript 1 Homo sapiens 65-71 31173166-0 2019 miR-128 is upregulated in epilepsy and promotes apoptosis through the SIRT1 cascade. mir-128 0-7 sirtuin 1 Rattus norvegicus 70-75 31173166-3 2019 Subsequently, results from an in vitro epilepsy model demonstrated that overexpression of miR-128 promoted nerve cell apoptosis, increased the protein expression of tumor protein p53, BCL2 associated X (Bax) and Cytochrome c, and enhanced caspase-3/9 activity, whereas it suppressed the protein expression of sirtuin 1 (SIRT1). mir-128 90-97 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 179-182 31173166-3 2019 Subsequently, results from an in vitro epilepsy model demonstrated that overexpression of miR-128 promoted nerve cell apoptosis, increased the protein expression of tumor protein p53, BCL2 associated X (Bax) and Cytochrome c, and enhanced caspase-3/9 activity, whereas it suppressed the protein expression of sirtuin 1 (SIRT1). mir-128 90-97 BCL2 associated X, apoptosis regulator Rattus norvegicus 184-201 31173166-3 2019 Subsequently, results from an in vitro epilepsy model demonstrated that overexpression of miR-128 promoted nerve cell apoptosis, increased the protein expression of tumor protein p53, BCL2 associated X (Bax) and Cytochrome c, and enhanced caspase-3/9 activity, whereas it suppressed the protein expression of sirtuin 1 (SIRT1). mir-128 90-97 BCL2 associated X, apoptosis regulator Rattus norvegicus 203-206 31173166-3 2019 Subsequently, results from an in vitro epilepsy model demonstrated that overexpression of miR-128 promoted nerve cell apoptosis, increased the protein expression of tumor protein p53, BCL2 associated X (Bax) and Cytochrome c, and enhanced caspase-3/9 activity, whereas it suppressed the protein expression of sirtuin 1 (SIRT1). mir-128 90-97 caspase 3 Rattus norvegicus 239-250 31173166-3 2019 Subsequently, results from an in vitro epilepsy model demonstrated that overexpression of miR-128 promoted nerve cell apoptosis, increased the protein expression of tumor protein p53, BCL2 associated X (Bax) and Cytochrome c, and enhanced caspase-3/9 activity, whereas it suppressed the protein expression of sirtuin 1 (SIRT1). mir-128 90-97 sirtuin 1 Rattus norvegicus 309-318 31173166-3 2019 Subsequently, results from an in vitro epilepsy model demonstrated that overexpression of miR-128 promoted nerve cell apoptosis, increased the protein expression of tumor protein p53, BCL2 associated X (Bax) and Cytochrome c, and enhanced caspase-3/9 activity, whereas it suppressed the protein expression of sirtuin 1 (SIRT1). mir-128 90-97 sirtuin 1 Rattus norvegicus 320-325 31173166-5 2019 Furthermore, treatment with CAY10602, a SIRT1 agonist, reduced the effects of miR-128 on nerve cells in vitro. mir-128 78-85 sirtuin 1 Rattus norvegicus 40-45 31173166-6 2019 Treatment with pifithrin-beta hydrobromide, a p53 inhibitor, was additionally able to mitigate the effects of miR-128 in vitro. mir-128 110-117 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 46-49 31173166-7 2019 In conclusion, the present findings indicated that anti-miR-128 may exert neuroprotective effects in epilepsy, through the SIRT1/p53/Bax/Cytochrome c/caspase signaling pathway. mir-128 56-63 sirtuin 1 Rattus norvegicus 123-128 31173166-7 2019 In conclusion, the present findings indicated that anti-miR-128 may exert neuroprotective effects in epilepsy, through the SIRT1/p53/Bax/Cytochrome c/caspase signaling pathway. mir-128 56-63 Wistar clone pR53P1 p53 pseudogene Rattus norvegicus 129-132 31173166-7 2019 In conclusion, the present findings indicated that anti-miR-128 may exert neuroprotective effects in epilepsy, through the SIRT1/p53/Bax/Cytochrome c/caspase signaling pathway. mir-128 56-63 BCL2 associated X, apoptosis regulator Rattus norvegicus 133-136 31173166-7 2019 In conclusion, the present findings indicated that anti-miR-128 may exert neuroprotective effects in epilepsy, through the SIRT1/p53/Bax/Cytochrome c/caspase signaling pathway. mir-128 56-63 caspase 9 Rattus norvegicus 150-157 31037985-9 2019 Moreover, an in vitro study demonstrated that miR-128 antagomir aggravated primary neuronal damage and exacerbated cell cycle reactivation induced by OGD/R stimulation; the underlying mechanism involved increasing cyclin A2, PTEN, and ERK expression and promoting phosphorylation of PTEN and ERK. mir-128 46-53 cyclin A2 Homo sapiens 214-223 31037985-9 2019 Moreover, an in vitro study demonstrated that miR-128 antagomir aggravated primary neuronal damage and exacerbated cell cycle reactivation induced by OGD/R stimulation; the underlying mechanism involved increasing cyclin A2, PTEN, and ERK expression and promoting phosphorylation of PTEN and ERK. mir-128 46-53 phosphatase and tensin homolog Homo sapiens 225-229 31037985-9 2019 Moreover, an in vitro study demonstrated that miR-128 antagomir aggravated primary neuronal damage and exacerbated cell cycle reactivation induced by OGD/R stimulation; the underlying mechanism involved increasing cyclin A2, PTEN, and ERK expression and promoting phosphorylation of PTEN and ERK. mir-128 46-53 mitogen-activated protein kinase 1 Homo sapiens 235-238 31037985-9 2019 Moreover, an in vitro study demonstrated that miR-128 antagomir aggravated primary neuronal damage and exacerbated cell cycle reactivation induced by OGD/R stimulation; the underlying mechanism involved increasing cyclin A2, PTEN, and ERK expression and promoting phosphorylation of PTEN and ERK. mir-128 46-53 phosphatase and tensin homolog Homo sapiens 283-287 31037985-9 2019 Moreover, an in vitro study demonstrated that miR-128 antagomir aggravated primary neuronal damage and exacerbated cell cycle reactivation induced by OGD/R stimulation; the underlying mechanism involved increasing cyclin A2, PTEN, and ERK expression and promoting phosphorylation of PTEN and ERK. mir-128 46-53 mitogen-activated protein kinase 1 Homo sapiens 292-295 31173288-9 2019 Spearman"s correlation coefficient was adopted to evaluate the correlation between miR-128 and PVT1-214 levels. mir-128 83-90 Pvt1 oncogene Homo sapiens 95-99 31080058-5 2019 Moreover, miR-128, miR-9, and let-7 are functionally distinct; capable of specifying neurons for layer VI and layer V and layers IV, III, and II, respectively; and transiently altering their relative levels of expression can modulate stem-cell competence in a neurogenic-stage-specific manner to shift neuron production between earlier-born and later-born fates, partly by temporally regulating a neurogenesis program involving Hmga2. mir-128 10-17 high mobility group AT-hook 2 Homo sapiens 428-433 30576236-5 2019 The relationship between MEG3/Girdin and miR-128 was determined and verified. mir-128 41-48 maternally expressed 3 Mus musculus 25-29 30576236-5 2019 The relationship between MEG3/Girdin and miR-128 was determined and verified. mir-128 41-48 coiled-coil domain containing 88A Homo sapiens 30-36 30576236-8 2019 To further analyze the MEG3-mediated regulatory mechanism, miR-128 upregulation and inhibition were introduced into the HUVECs. mir-128 59-66 maternally expressed 3 Mus musculus 23-27 30576236-10 2019 MEG3 downregulation was found to be capable of inhibiting Girdin but enhancing miR-128 expression. mir-128 79-86 maternally expressed 3 Mus musculus 0-4 30576236-12 2019 These findings provide evidence supporting that MEG3 leads to miR-128 downregulation and Girdin upregulation, which promotes platelet phagocytosis, thus protecting VECs from senescence. mir-128 62-69 maternally expressed 3 Mus musculus 48-52 30875657-9 2019 The potential relationship between miR-128 and WNT1 was analyzed. mir-128 35-42 wingless-type MMTV integration site family, member 1 Mus musculus 47-51 30875657-11 2019 WNT1 was targeted and negatively regulated by miR-128. mir-128 46-53 wingless-type MMTV integration site family, member 1 Mus musculus 0-4 30875657-13 2019 CONCLUSION: Collectively, miR-128 enhanced neuroprotective effect of DEX on HIBD neonatal mice by inhibiting WNT1. mir-128 26-33 wingless-type MMTV integration site family, member 1 Mus musculus 109-113 31173288-13 2019 Additionally, PVT1-214 functions as a competing endogenous RNA (ceRNA) by binding to miR-128. mir-128 85-92 Pvt1 oncogene Homo sapiens 14-18 31173288-14 2019 Inhibition of miR-128 releases Tropomyosin receptor kinase C (TrkC) from the complementary binding complex, subsequently increasing the protein level of TrkC in GC cells. mir-128 14-21 neurotrophic receptor tyrosine kinase 3 Homo sapiens 31-60 31173288-14 2019 Inhibition of miR-128 releases Tropomyosin receptor kinase C (TrkC) from the complementary binding complex, subsequently increasing the protein level of TrkC in GC cells. mir-128 14-21 neurotrophic receptor tyrosine kinase 3 Homo sapiens 62-66 31173288-14 2019 Inhibition of miR-128 releases Tropomyosin receptor kinase C (TrkC) from the complementary binding complex, subsequently increasing the protein level of TrkC in GC cells. mir-128 14-21 neurotrophic receptor tyrosine kinase 3 Homo sapiens 153-157 31173288-15 2019 CONCLUSIONS: PVT1-214-induced miR-128 repression regulates TrkC to further the progression of GC, indicating that this process will provide a promising therapeutic target in GC. mir-128 30-37 Pvt1 oncogene Homo sapiens 13-17 31173288-15 2019 CONCLUSIONS: PVT1-214-induced miR-128 repression regulates TrkC to further the progression of GC, indicating that this process will provide a promising therapeutic target in GC. mir-128 30-37 neurotrophic receptor tyrosine kinase 3 Homo sapiens 59-63 30556928-9 2019 MAPK1 was highly expressed in ovarian cancer-resistant tissues and cells and could be regulated by linc00161 and miR-128. mir-128 113-120 mitogen-activated protein kinase 1 Homo sapiens 0-5 30054858-9 2019 In contrast, reduction of miR-128, -15b, -195, -26b, -34b, -376b, and -381 by CSPGs was accompanied by increased EZR, KIF5A, DCX, GSK3B, and ROCK2 proteins. mir-128 26-33 ezrin Homo sapiens 113-116 30054858-9 2019 In contrast, reduction of miR-128, -15b, -195, -26b, -34b, -376b, and -381 by CSPGs was accompanied by increased EZR, KIF5A, DCX, GSK3B, and ROCK2 proteins. mir-128 26-33 kinesin family member 5A Homo sapiens 118-123 30867751-5 2019 In addition, it was identified that miR-128 could directly bind to the 3"-untranslated region of CIP2A. mir-128 36-43 cellular inhibitor of PP2A Homo sapiens 97-102 30265647-0 2019 MiR-128/SOX7 alleviates myocardial ischemia injury by regulating IL-33/sST2 in acute myocardial infarction. mir-128 0-7 interleukin 33 Rattus norvegicus 65-70 30265647-6 2019 The relationship between SRY-related HMG-box (SOX7) and miR-128 was verified using luciferase reporter assay. mir-128 56-63 SRY-box transcription factor 7 Rattus norvegicus 46-50 30265647-10 2019 Down-regulated miR-128 reversed the effects of IH on expression of SOX7, sST2 and cell apoptosis, while down-regulated sST2 abolished the effects of miR-128 inhibitor. mir-128 15-22 SRY-box transcription factor 7 Rattus norvegicus 67-71 30265647-11 2019 In addition, overexpressed IL-33 abolished the effects of miR-128 inhibitor that induced by IH on the expression of SOX7 and cell apoptosis. mir-128 58-65 interleukin 33 Rattus norvegicus 27-32 30265647-11 2019 In addition, overexpressed IL-33 abolished the effects of miR-128 inhibitor that induced by IH on the expression of SOX7 and cell apoptosis. mir-128 58-65 SRY-box transcription factor 7 Rattus norvegicus 116-120 30265647-13 2019 The present study indicated that miR-128 modulated cell apoptosis by targeting SOX7, which was mediated by IL-33/sST2 signaling pathway. mir-128 33-40 SRY-box transcription factor 7 Rattus norvegicus 79-83 30265647-13 2019 The present study indicated that miR-128 modulated cell apoptosis by targeting SOX7, which was mediated by IL-33/sST2 signaling pathway. mir-128 33-40 interleukin 33 Rattus norvegicus 107-112 30132868-0 2019 LncRNA SNHG3 induces EMT and sorafenib resistance by modulating the miR-128/CD151 pathway in hepatocellular carcinoma. mir-128 68-75 small nucleolar RNA host gene 3 Homo sapiens 7-12 30132868-0 2019 LncRNA SNHG3 induces EMT and sorafenib resistance by modulating the miR-128/CD151 pathway in hepatocellular carcinoma. mir-128 68-75 CD151 molecule (Raph blood group) Homo sapiens 76-81 30132868-5 2019 Moreover, SNHG3 overexpression induced HCC cells EMT via miR-128/CD151 cascade activation. mir-128 57-64 small nucleolar RNA host gene 3 Homo sapiens 10-15 30551451-0 2019 MiR-128 suppresses the growth of thyroid carcinoma by negatively regulating SPHK1. mir-128 0-7 sphingosine kinase 1 Homo sapiens 76-81 30535495-0 2019 Methylation-associated silencing of miR-128 promotes the development of esophageal cancer by targeting COX-2 in areas with a high incidence of esophageal cancer. mir-128 36-43 prostaglandin-endoperoxide synthase 2 Mus musculus 103-108 30535495-7 2019 Furthermore, the expression of miR-128 was downregulated by methylation, and COX-2, a direct target of miR-128, was upregulated with the reduction in miR-128. mir-128 103-110 prostaglandin-endoperoxide synthase 2 Mus musculus 77-82 30535495-7 2019 Furthermore, the expression of miR-128 was downregulated by methylation, and COX-2, a direct target of miR-128, was upregulated with the reduction in miR-128. mir-128 103-110 prostaglandin-endoperoxide synthase 2 Mus musculus 77-82 30535495-8 2019 Upregulation of miR-128 inhibited the cell cycle, proliferation and metastasis, and the expression of COX-2, cyclin D1 and retinoblastoma protein (Rb). mir-128 16-23 prostaglandin-endoperoxide synthase 2 Mus musculus 102-107 30535495-8 2019 Upregulation of miR-128 inhibited the cell cycle, proliferation and metastasis, and the expression of COX-2, cyclin D1 and retinoblastoma protein (Rb). mir-128 16-23 cyclin D1 Mus musculus 109-118 30535495-9 2019 Furthermore, the relative expression level of miR-128 was negatively associated with COX-2 in ESCC tissues. mir-128 46-53 prostaglandin-endoperoxide synthase 2 Mus musculus 85-90 30535495-10 2019 Collectively, these findings indicate that methylation-associated silencing of miR-128 promotes the development of esophageal cancer through upregulation of the expression of cyclin D1 and Rb by targeting COX-2 in ZD regions with a high incidence of esophageal cancer. mir-128 79-86 cyclin D1 Mus musculus 175-184 30535495-10 2019 Collectively, these findings indicate that methylation-associated silencing of miR-128 promotes the development of esophageal cancer through upregulation of the expression of cyclin D1 and Rb by targeting COX-2 in ZD regions with a high incidence of esophageal cancer. mir-128 79-86 prostaglandin-endoperoxide synthase 2 Mus musculus 205-210 30412727-10 2019 The interaction between miR-128 and PPARgamma were validated by bioinformatics analysis and luciferase reporter assay. mir-128 24-31 peroxisome proliferator activated receptor gamma Homo sapiens 36-45 30412727-13 2019 Moreover, PPARgamma was a target of miR-128. mir-128 36-43 peroxisome proliferator activated receptor gamma Mus musculus 10-19 30412727-14 2019 Additionally, miR-128 knockout or PPARgamma upregulation inhibited AD-like performances, amyloid plaque formation, Abeta generation, APP amyloidogenic processing and inflammatory responses in AD mice, while these effects of miR-128 knockout were abrogated by PPARgamma inhibitor. mir-128 14-21 amyloid beta (A4) precursor protein Mus musculus 115-120 30412727-14 2019 Additionally, miR-128 knockout or PPARgamma upregulation inhibited AD-like performances, amyloid plaque formation, Abeta generation, APP amyloidogenic processing and inflammatory responses in AD mice, while these effects of miR-128 knockout were abrogated by PPARgamma inhibitor. mir-128 14-21 peroxisome proliferator activated receptor gamma Mus musculus 259-268 30412727-15 2019 The results indicated MiR-128 knockout weakened AD-like performances, and reduced Abeta production and inflammatory responses by targeting PPARgamma in AD mice. mir-128 22-29 amyloid beta (A4) precursor protein Mus musculus 82-87 30412727-15 2019 The results indicated MiR-128 knockout weakened AD-like performances, and reduced Abeta production and inflammatory responses by targeting PPARgamma in AD mice. mir-128 22-29 peroxisome proliferator activated receptor gamma Mus musculus 139-148 30076714-0 2019 LncRNA PVT1 regulates VEGFC through inhibiting miR-128 in bladder cancer cells. mir-128 47-54 Pvt1 oncogene Homo sapiens 7-11 30076714-0 2019 LncRNA PVT1 regulates VEGFC through inhibiting miR-128 in bladder cancer cells. mir-128 47-54 vascular endothelial growth factor C Homo sapiens 22-27 30076714-7 2019 The results showed that the increased number of PVT1 transcripts interacted directly with miR-128 to decrease miR-128 binding to the VEGFC 3"-untranslated region. mir-128 90-97 Pvt1 oncogene Mus musculus 48-52 30076714-7 2019 The results showed that the increased number of PVT1 transcripts interacted directly with miR-128 to decrease miR-128 binding to the VEGFC 3"-untranslated region. mir-128 90-97 vascular endothelial growth factor C Homo sapiens 133-138 30076714-7 2019 The results showed that the increased number of PVT1 transcripts interacted directly with miR-128 to decrease miR-128 binding to the VEGFC 3"-untranslated region. mir-128 110-117 Pvt1 oncogene Mus musculus 48-52 30076714-7 2019 The results showed that the increased number of PVT1 transcripts interacted directly with miR-128 to decrease miR-128 binding to the VEGFC 3"-untranslated region. mir-128 110-117 vascular endothelial growth factor C Homo sapiens 133-138 30076714-8 2019 This effect suppressed VEGFC mRNA degradation by miR-128. mir-128 49-56 vascular endothelial growth factor C Homo sapiens 23-28 30076414-0 2019 Long noncoding RNA PVT1-214 promotes proliferation and invasion of colorectal cancer by stabilizing Lin28 and interacting with miR-128. mir-128 127-134 Pvt1 oncogene Homo sapiens 19-23 30551451-8 2019 Following, we characterized sphingosine kinase-1 (SPHK1) as a direct target of miR-128 that interacted with the 3"-untranslated region (UTR) of SPHK1, and the results were confirmed by using luciferase-reporter assay. mir-128 79-86 sphingosine kinase 1 Homo sapiens 28-48 30551451-8 2019 Following, we characterized sphingosine kinase-1 (SPHK1) as a direct target of miR-128 that interacted with the 3"-untranslated region (UTR) of SPHK1, and the results were confirmed by using luciferase-reporter assay. mir-128 79-86 sphingosine kinase 1 Homo sapiens 50-55 30551451-8 2019 Following, we characterized sphingosine kinase-1 (SPHK1) as a direct target of miR-128 that interacted with the 3"-untranslated region (UTR) of SPHK1, and the results were confirmed by using luciferase-reporter assay. mir-128 79-86 sphingosine kinase 1 Homo sapiens 144-149 30551451-9 2019 We also observed that SPHK1 expression was decreased and negatively correlated with miR-128 expression in PTC and FTC tissues clinically. mir-128 84-91 sphingosine kinase 1 Homo sapiens 22-27 30551451-10 2019 Importantly, ectopic expression of SPHK1 significantly abrogated the tumor-suppressive effect induced by miR-128, as supported by the reduced apoptosis, while the enhanced proliferation and metastasis. mir-128 105-112 sphingosine kinase 1 Homo sapiens 35-40 30551451-11 2019 Finally, over-expressing miR-128 apparently reduced the tumor growth rate and tumor weight in vivo using xenograft tumor model, accompanied with a remarkable decrease of SPHK1. mir-128 25-32 sphingosine kinase 1 Homo sapiens 170-175 30546426-2 2018 The mechanism of miR-128 on the regulation of Ribophorin-II (RPN2) in CRC cells was explored in the present study. mir-128 17-24 ribophorin II Homo sapiens 61-65 30407643-0 2018 NAIF1 suppresses osteosarcoma progression and is regulated by miR-128. mir-128 62-69 nuclear apoptosis inducing factor 1 Homo sapiens 0-5 30407643-7 2018 The bioinformatic prediction and luciferase reporter assay revealed that miR-128 is a direct upstream regulator of NAIF1 and regulates NAIF1 expression by binding the 3"-UTR of NAIF1. mir-128 73-80 nuclear apoptosis inducing factor 1 Homo sapiens 115-120 30407643-7 2018 The bioinformatic prediction and luciferase reporter assay revealed that miR-128 is a direct upstream regulator of NAIF1 and regulates NAIF1 expression by binding the 3"-UTR of NAIF1. mir-128 73-80 nuclear apoptosis inducing factor 1 Homo sapiens 135-140 30407643-7 2018 The bioinformatic prediction and luciferase reporter assay revealed that miR-128 is a direct upstream regulator of NAIF1 and regulates NAIF1 expression by binding the 3"-UTR of NAIF1. mir-128 73-80 nuclear apoptosis inducing factor 1 Homo sapiens 135-140 30407643-9 2018 Moreover, miR-128 expression levels were inversely correlated with that of NAIF1 in osteosarcoma tissues. mir-128 10-17 nuclear apoptosis inducing factor 1 Homo sapiens 75-80 30407643-10 2018 Finally, functional assay showed that miR-128 significantly suppressed osteosarcoma progression partially mediated by inhibiting NAIF1 expression. mir-128 38-45 nuclear apoptosis inducing factor 1 Homo sapiens 129-134 30407643-11 2018 These data indicate that the miR-128 and its target gene NAIF1 played important roles by regulating OS cell proliferation and migration phenotype. mir-128 29-36 nuclear apoptosis inducing factor 1 Homo sapiens 57-62 30407643-15 2018 The data of this study indicated that the miR-128 and its target gene NAIF1 played important roles by regulating OS cell proliferation and migration phenotype. mir-128 42-49 nuclear apoptosis inducing factor 1 Homo sapiens 70-75 30575919-0 2018 MiR-128 promotes the apoptosis of glioma cells via binding to NEK2. mir-128 0-7 NIMA related kinase 2 Homo sapiens 62-66 30575919-1 2018 OBJECTIVE: The aim of this study was to explore whether miR-128 could promote the apoptosis of glioma cells by targeting NIMA related kinase-2 (NEK2), thus participating in the occurrence and progression of glioma. mir-128 56-63 NIMA related kinase 2 Homo sapiens 121-142 30575919-1 2018 OBJECTIVE: The aim of this study was to explore whether miR-128 could promote the apoptosis of glioma cells by targeting NIMA related kinase-2 (NEK2), thus participating in the occurrence and progression of glioma. mir-128 56-63 NIMA related kinase 2 Homo sapiens 144-148 30575919-6 2018 Meanwhile, rescue experiments were conducted to determine whether miR-128 could promote the apoptosis of glioma cells by targeting NEK2. mir-128 66-73 NIMA related kinase 2 Homo sapiens 131-135 30575919-10 2018 Meanwhile, overexpression of miR-128 promoted the apoptosis of U87 cells, upregulated protein levels of cleaved Caspase-3 and BCL2-associated X (Bax), and downregulated B-cell lymphoma-2 (Bcl-2). mir-128 29-36 caspase 3 Homo sapiens 112-121 30575919-10 2018 Meanwhile, overexpression of miR-128 promoted the apoptosis of U87 cells, upregulated protein levels of cleaved Caspase-3 and BCL2-associated X (Bax), and downregulated B-cell lymphoma-2 (Bcl-2). mir-128 29-36 BCL2 associated X, apoptosis regulator Homo sapiens 126-143 30575919-10 2018 Meanwhile, overexpression of miR-128 promoted the apoptosis of U87 cells, upregulated protein levels of cleaved Caspase-3 and BCL2-associated X (Bax), and downregulated B-cell lymphoma-2 (Bcl-2). mir-128 29-36 BCL2 associated X, apoptosis regulator Homo sapiens 145-148 30575919-10 2018 Meanwhile, overexpression of miR-128 promoted the apoptosis of U87 cells, upregulated protein levels of cleaved Caspase-3 and BCL2-associated X (Bax), and downregulated B-cell lymphoma-2 (Bcl-2). mir-128 29-36 BCL2 apoptosis regulator Homo sapiens 169-186 30575919-10 2018 Meanwhile, overexpression of miR-128 promoted the apoptosis of U87 cells, upregulated protein levels of cleaved Caspase-3 and BCL2-associated X (Bax), and downregulated B-cell lymphoma-2 (Bcl-2). mir-128 29-36 BCL2 apoptosis regulator Homo sapiens 188-193 30575919-11 2018 Dual-luciferase reporter gene assay indicated that miR-128 directly bound to NEK2. mir-128 51-58 NIMA related kinase 2 Homo sapiens 77-81 30575919-12 2018 Further rescue experiments suggested that NEK2 overexpression partially reversed the effect of miR-128 on the apoptosis of glioma cells. mir-128 95-102 NIMA related kinase 2 Homo sapiens 42-46 30575919-13 2018 CONCLUSIONS: Downregulated miR-128 inhibited the apoptosis of glioma cells via targeting NEK2. mir-128 27-34 NIMA related kinase 2 Homo sapiens 89-93 30546426-7 2018 In the present study, it was identified that aberrant decreased miR-128 was negatively correlated with RPN2 in CRC tissues. mir-128 64-71 ribophorin II Homo sapiens 103-107 30546426-11 2018 Markedly decreased Akt phosphorylation and cyclin D1 levels and increased p53 levels were detected when cells were transfected with miR-128 mimics. mir-128 132-139 AKT serine/threonine kinase 1 Homo sapiens 19-22 30546426-11 2018 Markedly decreased Akt phosphorylation and cyclin D1 levels and increased p53 levels were detected when cells were transfected with miR-128 mimics. mir-128 132-139 cyclin D1 Homo sapiens 43-52 30546426-11 2018 Markedly decreased Akt phosphorylation and cyclin D1 levels and increased p53 levels were detected when cells were transfected with miR-128 mimics. mir-128 132-139 tumor protein p53 Homo sapiens 74-77 30546426-13 2018 Subsequent to screening with miRNA target prediction databases, the specificity of miR-128-targeted RPN2 was validated by a luciferase reporter assay. mir-128 83-90 ribophorin II Homo sapiens 100-104 30546426-14 2018 In conclusion, the results suggested that miR-128 was a specific negative regulator of RPN2, which regulated colorectal cancer cell proliferation and migration by affecting the Akt-p53-cyclin pathway. mir-128 42-49 ribophorin II Homo sapiens 87-91 30546426-14 2018 In conclusion, the results suggested that miR-128 was a specific negative regulator of RPN2, which regulated colorectal cancer cell proliferation and migration by affecting the Akt-p53-cyclin pathway. mir-128 42-49 AKT serine/threonine kinase 1 Homo sapiens 177-180 30546426-14 2018 In conclusion, the results suggested that miR-128 was a specific negative regulator of RPN2, which regulated colorectal cancer cell proliferation and migration by affecting the Akt-p53-cyclin pathway. mir-128 42-49 tumor protein p53 Homo sapiens 181-184 30546426-14 2018 In conclusion, the results suggested that miR-128 was a specific negative regulator of RPN2, which regulated colorectal cancer cell proliferation and migration by affecting the Akt-p53-cyclin pathway. mir-128 42-49 proliferating cell nuclear antigen Homo sapiens 185-191 30177387-3 2018 In this study, we found that miR-128 is a critical tumor suppressor that is downregulated in GC patients and GC cells and that GAREM is a direct downstream target of miR-128. mir-128 166-173 GRB2 associated regulator of MAPK1 subtype 1 Homo sapiens 127-132 30328325-0 2018 Inhibition of miR-128 Abates Abeta-Mediated Cytotoxicity by Targeting PPAR-gamma via NF-kappaB Inactivation in Primary Mouse Cortical Neurons and Neuro2a Cells. mir-128 14-21 amyloid beta (A4) precursor protein Mus musculus 29-34 30328325-0 2018 Inhibition of miR-128 Abates Abeta-Mediated Cytotoxicity by Targeting PPAR-gamma via NF-kappaB Inactivation in Primary Mouse Cortical Neurons and Neuro2a Cells. mir-128 14-21 peroxisome proliferator activated receptor gamma Mus musculus 70-80 30328325-0 2018 Inhibition of miR-128 Abates Abeta-Mediated Cytotoxicity by Targeting PPAR-gamma via NF-kappaB Inactivation in Primary Mouse Cortical Neurons and Neuro2a Cells. mir-128 14-21 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 85-94 30328325-11 2018 RESULTS: MiR-128 expression was upregulated and PPAR-gamma expression was downregulated in plasma from AD patients and amyloid-beta (Abeta)-treated primary mouse cortical neurons (MCN) and Neuro2a (N2a) cells. mir-128 9-16 amyloid beta precursor protein Homo sapiens 119-131 30328325-11 2018 RESULTS: MiR-128 expression was upregulated and PPAR-gamma expression was downregulated in plasma from AD patients and amyloid-beta (Abeta)-treated primary mouse cortical neurons (MCN) and Neuro2a (N2a) cells. mir-128 9-16 amyloid beta precursor protein Homo sapiens 133-138 30328325-12 2018 Inhibition of miR-128 decreased Abeta-mediated cytotoxicity through inactivation of NF-kappaB in MCN and N2a cells. mir-128 14-21 amyloid beta (A4) precursor protein Mus musculus 32-37 30328325-12 2018 Inhibition of miR-128 decreased Abeta-mediated cytotoxicity through inactivation of NF-kappaB in MCN and N2a cells. mir-128 14-21 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 84-93 30328325-13 2018 Moreover, PPAR-gamma was a target of miR-128. mir-128 37-44 peroxisome proliferator activated receptor gamma Mus musculus 10-20 30328325-15 2018 Furthermore, PPAR-gamma downregulation was able to abolish the effect of anti-miR-128 on cytotoxicity and NF-kappaB activity in MCN and N2a cells. mir-128 78-85 peroxisome proliferator activated receptor gamma Mus musculus 13-23 30328325-15 2018 Furthermore, PPAR-gamma downregulation was able to abolish the effect of anti-miR-128 on cytotoxicity and NF-kappaB activity in MCN and N2a cells. mir-128 78-85 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 106-115 30328325-16 2018 CONCLUSION: MiR-128 inhibitor decreased Abeta-mediated cytotoxicity by upregulating PPAR-gamma via inactivation of NF-kappaB in MCN and N2a cells, providing a new potential target in AD treatment. mir-128 12-19 amyloid beta (A4) precursor protein Mus musculus 40-45 30328325-16 2018 CONCLUSION: MiR-128 inhibitor decreased Abeta-mediated cytotoxicity by upregulating PPAR-gamma via inactivation of NF-kappaB in MCN and N2a cells, providing a new potential target in AD treatment. mir-128 12-19 peroxisome proliferator activated receptor gamma Mus musculus 84-94 30328325-16 2018 CONCLUSION: MiR-128 inhibitor decreased Abeta-mediated cytotoxicity by upregulating PPAR-gamma via inactivation of NF-kappaB in MCN and N2a cells, providing a new potential target in AD treatment. mir-128 12-19 nuclear factor of kappa light polypeptide gene enhancer in B cells 1, p105 Mus musculus 115-124 30206206-7 2018 In vitro, miR-128 signaling hindered Atg12 expression, LC3-II conversion, and autophagic puncta formation through targeting the 3"-untranslated region of Atg12. mir-128 10-17 autophagy related 12 Homo sapiens 37-42 30206206-7 2018 In vitro, miR-128 signaling hindered Atg12 expression, LC3-II conversion, and autophagic puncta formation through targeting the 3"-untranslated region of Atg12. mir-128 10-17 autophagy related 12 Homo sapiens 154-159 30203508-0 2018 NEAT1 contributes to ox-LDL-induced inflammation and oxidative stress in macrophages through inhibiting miR-128. mir-128 104-111 nuclear paraspeckle assembly transcript 1 (non-protein coding) Mus musculus 0-5 30203508-12 2018 Interestingly, miR-128 mimics was able to reverse AS-correlated events induced by overexpression of NEAT1. mir-128 15-22 nuclear paraspeckle assembly transcript 1 (non-protein coding) Mus musculus 100-105 30203508-13 2018 By using bioinformatics analysis, miR-128 was predicted as a target of NEAT1 and the correlation between them was validated in our study. mir-128 34-41 nuclear paraspeckle assembly transcript 1 (non-protein coding) Mus musculus 71-76 30203508-14 2018 Taken these together, it was implied that NEAT1 participated in ox-LDL-induced inflammation and oxidative stress in AS development through sponging miR-128. mir-128 148-155 nuclear paraspeckle assembly transcript 1 (non-protein coding) Mus musculus 42-47 29777777-6 2018 As verified using Immunoblotting assays, miR-128 significantly regulated the protein levels of VEGFC/VEGFR3, whereas miR-3916 only slightly modulated VEGFC and VEGFR3 proteins. mir-128 41-48 vascular endothelial growth factor C Homo sapiens 95-100 29777777-6 2018 As verified using Immunoblotting assays, miR-128 significantly regulated the protein levels of VEGFC/VEGFR3, whereas miR-3916 only slightly modulated VEGFC and VEGFR3 proteins. mir-128 41-48 fms related receptor tyrosine kinase 4 Homo sapiens 101-107 29777777-7 2018 Contrary to VEGFC, miR-128 overexpression remarkably suppressed LEC proliferation, Ca2+ release and ERK1/2-Akt signaling; moreover, the effect of VEGFC could be partially attenuated by miR-128. mir-128 19-26 mitogen-activated protein kinase 3 Homo sapiens 100-106 29777777-7 2018 Contrary to VEGFC, miR-128 overexpression remarkably suppressed LEC proliferation, Ca2+ release and ERK1/2-Akt signaling; moreover, the effect of VEGFC could be partially attenuated by miR-128. mir-128 19-26 AKT serine/threonine kinase 1 Homo sapiens 107-110 29777777-7 2018 Contrary to VEGFC, miR-128 overexpression remarkably suppressed LEC proliferation, Ca2+ release and ERK1/2-Akt signaling; moreover, the effect of VEGFC could be partially attenuated by miR-128. mir-128 19-26 vascular endothelial growth factor C Homo sapiens 146-151 29777777-7 2018 Contrary to VEGFC, miR-128 overexpression remarkably suppressed LEC proliferation, Ca2+ release and ERK1/2-Akt signaling; moreover, the effect of VEGFC could be partially attenuated by miR-128. mir-128 185-192 vascular endothelial growth factor C Homo sapiens 146-151 29777777-8 2018 In summary, miR-128 interacts with the 3"-UTR of VEGFC and VEGFR3 to inhibit their expression, thus suppressing LEC proliferation through Ca2+ and ERK1/2-Akt signaling. mir-128 12-19 vascular endothelial growth factor C Homo sapiens 49-54 29777777-8 2018 In summary, miR-128 interacts with the 3"-UTR of VEGFC and VEGFR3 to inhibit their expression, thus suppressing LEC proliferation through Ca2+ and ERK1/2-Akt signaling. mir-128 12-19 fms related receptor tyrosine kinase 4 Homo sapiens 59-65 29777777-8 2018 In summary, miR-128 interacts with the 3"-UTR of VEGFC and VEGFR3 to inhibit their expression, thus suppressing LEC proliferation through Ca2+ and ERK1/2-Akt signaling. mir-128 12-19 mitogen-activated protein kinase 3 Homo sapiens 147-153 29777777-8 2018 In summary, miR-128 interacts with the 3"-UTR of VEGFC and VEGFR3 to inhibit their expression, thus suppressing LEC proliferation through Ca2+ and ERK1/2-Akt signaling. mir-128 12-19 AKT serine/threonine kinase 1 Homo sapiens 154-157 30025750-0 2018 miR-128 targets the CC chemokine ligand 18 gene (CCL18) in cutaneous malignant melanoma progression. mir-128 0-7 C-C motif chemokine ligand 18 Homo sapiens 20-42 30025750-0 2018 miR-128 targets the CC chemokine ligand 18 gene (CCL18) in cutaneous malignant melanoma progression. mir-128 0-7 C-C motif chemokine ligand 18 Homo sapiens 49-54 30025750-6 2018 The changes in expression of CCL18 after miR-128 mimic transfection of A375 melanoma cells were determined by both qRT-PCR and Western-bloting. mir-128 41-48 C-C motif chemokine ligand 18 Homo sapiens 29-34 30025750-12 2018 RESULTS: The expression of miR-128 had negative relevance with CCL18 in CMM. mir-128 27-34 C-C motif chemokine ligand 18 Homo sapiens 63-68 30025750-14 2018 Transfected miR-128 mimic significantly reduced CCL18 expression and this impairment of CCL18 gene promoted apoptosis, inhibited migration and colony formation of A375 melanoma cells. mir-128 12-19 C-C motif chemokine ligand 18 Homo sapiens 48-53 30025750-14 2018 Transfected miR-128 mimic significantly reduced CCL18 expression and this impairment of CCL18 gene promoted apoptosis, inhibited migration and colony formation of A375 melanoma cells. mir-128 12-19 C-C motif chemokine ligand 18 Homo sapiens 88-93 30025750-17 2018 Overexpression of miR-128 inhibits the oncogenic effect of CCL18. mir-128 18-25 C-C motif chemokine ligand 18 Homo sapiens 59-64 30177387-4 2018 Overexpression of miR-128 in HGC-27 and MKN-45 cells resulted in suppressed cell growth and promoted cell apoptosis through a GAREM-dependent mechanism. mir-128 18-25 GRB2 associated regulator of MAPK1 subtype 1 Homo sapiens 126-131 30177387-5 2018 Moreover, the precise mechanisms underlying the antitumor effect of miR-128 in GC are at least partially due to suppressing activation of the MAPK signaling pathway, induced by suppressing GAREM expression. mir-128 68-75 GRB2 associated regulator of MAPK1 subtype 1 Homo sapiens 189-194 30121007-9 2018 Furthermore, we found that tumor suppressor miR-128 negatively regulated PAICS expression by binding to its 3"-untranslated region. mir-128 44-51 phosphoribosylaminoimidazole carboxylase and phosphoribosylaminoimidazolesuccinocarboxamide synthase Gallus gallus 73-78 30223934-6 2018 Besides, the association between miR-128-3p and ERK and PI3K/AKT pathways was further explored. mir-128 33-40 mitogen-activated protein kinase 1 Homo sapiens 48-51 30223934-6 2018 Besides, the association between miR-128-3p and ERK and PI3K/AKT pathways was further explored. mir-128 33-40 AKT serine/threonine kinase 1 Homo sapiens 61-64 29524580-4 2018 However, the inhibitory effects of miR-128 mimics on the invasion and proliferation of glioma cells were reversed by overexpression of cyclooxygenase-2 (COX-2). mir-128 35-42 prostaglandin-endoperoxide synthase 2 Homo sapiens 135-151 29524580-4 2018 However, the inhibitory effects of miR-128 mimics on the invasion and proliferation of glioma cells were reversed by overexpression of cyclooxygenase-2 (COX-2). mir-128 35-42 prostaglandin-endoperoxide synthase 2 Homo sapiens 153-158 29524580-5 2018 Our data showed that COX-2 was a candidate target of miR-128. mir-128 53-60 prostaglandin-endoperoxide synthase 2 Homo sapiens 21-26 29524580-6 2018 Luciferase activity of 3"-UTR of COX-2 was reduced in the presence of miR-128. mir-128 70-77 prostaglandin-endoperoxide synthase 2 Homo sapiens 33-38 29524580-7 2018 Additionally, miR-128 obviously decreased COX-2 mRNA stability determined by real time PCR. mir-128 14-21 prostaglandin-endoperoxide synthase 2 Homo sapiens 42-47 29524580-8 2018 Contrarily, we found that miR-128 inhibitor significantly increased the COX-2 mRNA expression, and elevated the protein expression of MMP9 and ki67, and promoted the proliferation of glioma cells. mir-128 26-33 prostaglandin-endoperoxide synthase 2 Homo sapiens 72-77 29524580-8 2018 Contrarily, we found that miR-128 inhibitor significantly increased the COX-2 mRNA expression, and elevated the protein expression of MMP9 and ki67, and promoted the proliferation of glioma cells. mir-128 26-33 matrix metallopeptidase 9 Homo sapiens 134-138 29524580-10 2018 All of these results supported that miR-128 was a direct regulator of COX-2. mir-128 36-43 prostaglandin-endoperoxide synthase 2 Homo sapiens 70-75 29524580-11 2018 Further studies proved that COX-2 was elevated in glioma tissues and its expression was negatively correlated with the levels of miR-128. mir-128 129-136 prostaglandin-endoperoxide synthase 2 Homo sapiens 28-33 29497903-11 2018 The levels of both cleaved caspase-3 and cleaved caspase-9 were decreased in hippocampus exposed to the miR-128 mimic, whereas they were markedly increased in miR-128 inhibitor-treated hippocampus. mir-128 104-111 caspase 3 Rattus norvegicus 27-36 29805606-5 2018 First, the expressions levels of miR-128 and its target gene, SAM and SH3 domain-containing 1 (SASH1), were measured in tissues from patients with osteosarcoma, and their correlation with osteosarcoma in terms of the pathological level were examined. mir-128 33-40 SAM and SH3 domain containing 1 Homo sapiens 62-93 29805606-5 2018 First, the expressions levels of miR-128 and its target gene, SAM and SH3 domain-containing 1 (SASH1), were measured in tissues from patients with osteosarcoma, and their correlation with osteosarcoma in terms of the pathological level were examined. mir-128 33-40 SAM and SH3 domain containing 1 Homo sapiens 95-100 29805606-7 2018 Subsequently, the association between SASH1 and miR-128 was evaluated using a dual luciferase gene reporter assay. mir-128 48-55 SAM and SH3 domain containing 1 Homo sapiens 38-43 29805606-12 2018 Additionally, the expression of B-cell lymphoma 2 (Bcl-2) was downregulated in the miR-128-inhibited group, compared with that in the control group, whereas the expression levels of SASH1, Bcl-2-associated X protein and caspase-3 were upregulated in the group with miR-128 inhibition (P<0.05). mir-128 83-90 BCL2 apoptosis regulator Homo sapiens 32-49 29805606-12 2018 Additionally, the expression of B-cell lymphoma 2 (Bcl-2) was downregulated in the miR-128-inhibited group, compared with that in the control group, whereas the expression levels of SASH1, Bcl-2-associated X protein and caspase-3 were upregulated in the group with miR-128 inhibition (P<0.05). mir-128 83-90 BCL2 apoptosis regulator Homo sapiens 51-56 29805606-12 2018 Additionally, the expression of B-cell lymphoma 2 (Bcl-2) was downregulated in the miR-128-inhibited group, compared with that in the control group, whereas the expression levels of SASH1, Bcl-2-associated X protein and caspase-3 were upregulated in the group with miR-128 inhibition (P<0.05). mir-128 83-90 SAM and SH3 domain containing 1 Homo sapiens 182-187 29091297-5 2018 JAG1 is targeted and negatively regulated by miR-128. mir-128 45-52 jagged canonical Notch ligand 1 Homo sapiens 0-4 29091297-7 2018 Compared with the resistance group, the anti-miR-128 group showed decreasedBax expression along with a lowered cell inhibition rate and apoptosis rate, but increased JAG1 and Bcl-2 expression with reduced cells arrested in the S phase; while the miR-128 mimic group showed an opposite trend; the si-JAG1 group showed decreased Bcl-2 expression and reduced cells in the S phase. mir-128 45-52 jagged canonical Notch ligand 1 Homo sapiens 166-170 29091297-7 2018 Compared with the resistance group, the anti-miR-128 group showed decreasedBax expression along with a lowered cell inhibition rate and apoptosis rate, but increased JAG1 and Bcl-2 expression with reduced cells arrested in the S phase; while the miR-128 mimic group showed an opposite trend; the si-JAG1 group showed decreased Bcl-2 expression and reduced cells in the S phase. mir-128 45-52 BCL2 apoptosis regulator Homo sapiens 175-180 29091297-7 2018 Compared with the resistance group, the anti-miR-128 group showed decreasedBax expression along with a lowered cell inhibition rate and apoptosis rate, but increased JAG1 and Bcl-2 expression with reduced cells arrested in the S phase; while the miR-128 mimic group showed an opposite trend; the si-JAG1 group showed decreased Bcl-2 expression and reduced cells in the S phase. mir-128 45-52 jagged canonical Notch ligand 1 Homo sapiens 299-303 29091297-7 2018 Compared with the resistance group, the anti-miR-128 group showed decreasedBax expression along with a lowered cell inhibition rate and apoptosis rate, but increased JAG1 and Bcl-2 expression with reduced cells arrested in the S phase; while the miR-128 mimic group showed an opposite trend; the si-JAG1 group showed decreased Bcl-2 expression and reduced cells in the S phase. mir-128 45-52 BCL2 apoptosis regulator Homo sapiens 327-332 29600803-0 2018 Publisher Correction: Simultaneous overactivation of Wnt/beta-catenin and TGFbeta signalling by miR-128-3p confers chemoresistance-associated metastasis in NSCLC. mir-128 96-103 transforming growth factor beta 1 Homo sapiens 74-81 29805525-0 2018 miR-128 induces pancreas cancer cell apoptosis by targeting MDM4. mir-128 0-7 MDM4 regulator of p53 Homo sapiens 60-64 29805606-12 2018 Additionally, the expression of B-cell lymphoma 2 (Bcl-2) was downregulated in the miR-128-inhibited group, compared with that in the control group, whereas the expression levels of SASH1, Bcl-2-associated X protein and caspase-3 were upregulated in the group with miR-128 inhibition (P<0.05). mir-128 83-90 BCL2 apoptosis regulator Homo sapiens 189-194 29805606-12 2018 Additionally, the expression of B-cell lymphoma 2 (Bcl-2) was downregulated in the miR-128-inhibited group, compared with that in the control group, whereas the expression levels of SASH1, Bcl-2-associated X protein and caspase-3 were upregulated in the group with miR-128 inhibition (P<0.05). mir-128 83-90 caspase 3 Homo sapiens 220-229 29805606-12 2018 Additionally, the expression of B-cell lymphoma 2 (Bcl-2) was downregulated in the miR-128-inhibited group, compared with that in the control group, whereas the expression levels of SASH1, Bcl-2-associated X protein and caspase-3 were upregulated in the group with miR-128 inhibition (P<0.05). mir-128 265-272 BCL2 apoptosis regulator Homo sapiens 32-49 29805606-12 2018 Additionally, the expression of B-cell lymphoma 2 (Bcl-2) was downregulated in the miR-128-inhibited group, compared with that in the control group, whereas the expression levels of SASH1, Bcl-2-associated X protein and caspase-3 were upregulated in the group with miR-128 inhibition (P<0.05). mir-128 265-272 BCL2 apoptosis regulator Homo sapiens 51-56 29805606-13 2018 SASH1 was confirmed as a direct target of miR-128 using a dual luciferase gene reporter assay. mir-128 42-49 SAM and SH3 domain containing 1 Homo sapiens 0-5 29805606-15 2018 The results of the present study suggested that miR-128 may regulate the tumorigenesis and evolution of osteosarcoma through targeting SASH1. mir-128 48-55 SAM and SH3 domain containing 1 Homo sapiens 135-140 29600803-0 2018 Publisher Correction: Simultaneous overactivation of Wnt/beta-catenin and TGFbeta signalling by miR-128-3p confers chemoresistance-associated metastasis in NSCLC. mir-128 96-103 catenin beta 1 Homo sapiens 57-69 29581509-5 2018 ARPP21 antagonizes miR-128 activity by co-regulating a subset of miR-128 target mRNAs enriched for neurodevelopmental functions. mir-128 19-26 cAMP regulated phosphoprotein 21 Homo sapiens 0-6 29497903-11 2018 The levels of both cleaved caspase-3 and cleaved caspase-9 were decreased in hippocampus exposed to the miR-128 mimic, whereas they were markedly increased in miR-128 inhibitor-treated hippocampus. mir-128 104-111 caspase 9 Rattus norvegicus 49-58 30537735-12 2018 CONCLUSION: Evidence from experimental models revealed that miR-128 might reduce apoptosis of DA neurons while increasing the expression of EAAT4 which might be related to the downregulation of AXIN1. mir-128 60-67 solute carrier family 1 (high affinity aspartate/glutamate transporter), member 6 Mus musculus 140-145 30257253-0 2018 miR-128 Targets the SIRT1/ROS/DR5 Pathway to Sensitize Colorectal Cancer to TRAIL-Induced Apoptosis. mir-128 0-7 sirtuin 1 Homo sapiens 20-25 30257253-0 2018 miR-128 Targets the SIRT1/ROS/DR5 Pathway to Sensitize Colorectal Cancer to TRAIL-Induced Apoptosis. mir-128 0-7 TNF receptor superfamily member 10b Homo sapiens 30-33 30257253-0 2018 miR-128 Targets the SIRT1/ROS/DR5 Pathway to Sensitize Colorectal Cancer to TRAIL-Induced Apoptosis. mir-128 0-7 TNF superfamily member 10 Homo sapiens 76-81 30257253-5 2018 MTT assays were used to evaluate the effect of miR-128 on TRAIL-induced cytotoxicity against CRC cell lines. mir-128 47-54 TNF superfamily member 10 Homo sapiens 58-63 30257253-7 2018 Western blot, flow cytometry, and luciferase reporter assays were performed to evaluate the potential mechanism and pathway of miR-128-promoted apoptosis in TRAIL-treated CRC cells. mir-128 127-134 TNF superfamily member 10 Homo sapiens 157-162 30257253-9 2018 The enforced expression of miR-128 sensitized CRC cells to TRAIL-induced cytotoxicity by inducing apoptosis. mir-128 27-34 TNF superfamily member 10 Homo sapiens 59-64 30257253-10 2018 Mechanistically, bioinformatics, western blot analysis, and luciferase reporter assays showed that miR-128 directly targeted sirtuin 1 (SIRT1) in CRC cells. mir-128 99-106 sirtuin 1 Homo sapiens 125-134 30257253-10 2018 Mechanistically, bioinformatics, western blot analysis, and luciferase reporter assays showed that miR-128 directly targeted sirtuin 1 (SIRT1) in CRC cells. mir-128 99-106 sirtuin 1 Homo sapiens 136-141 30257253-11 2018 miR-128 overexpression suppressed SIRT1 expression, which promoted the production of ROS in TRAIL-treated CRC cells. mir-128 0-7 sirtuin 1 Homo sapiens 34-39 30257253-11 2018 miR-128 overexpression suppressed SIRT1 expression, which promoted the production of ROS in TRAIL-treated CRC cells. mir-128 0-7 TNF superfamily member 10 Homo sapiens 92-97 30537735-4 2018 The targeting relationship between miR-128 and AXIN1 was verified via a bioinformatics prediction and dual-luciferase reporter gene assay. mir-128 35-42 axin 1 Mus musculus 47-52 30537735-5 2018 After separation, DA neurons were subjected to a series of inhibitors, activators and shRNAs to validate the mechanisms of miR-128 in controlling of AXIN1 in PD. mir-128 123-130 axin 1 Mus musculus 149-154 30537735-10 2018 AXIN1 was targeted and negatively mediated by miR-128. mir-128 46-53 axin 1 Mus musculus 0-5 30537735-11 2018 In the DA neurons, upregulated miR-128 expression or sh-AXIN1 increased the positive expression rate of EAAT4 together with mRNA and protein levels, but decreased the mRNA and protein levels of AXIN1, apoptosis rate along with the positive expression rate of AXIN1; however, the opposite trend was found in response to transfection with miR-128 inhibitors. mir-128 31-38 solute carrier family 1 (high affinity aspartate/glutamate transporter), member 6 Mus musculus 104-109 30537735-11 2018 In the DA neurons, upregulated miR-128 expression or sh-AXIN1 increased the positive expression rate of EAAT4 together with mRNA and protein levels, but decreased the mRNA and protein levels of AXIN1, apoptosis rate along with the positive expression rate of AXIN1; however, the opposite trend was found in response to transfection with miR-128 inhibitors. mir-128 31-38 axin 1 Mus musculus 194-199 30537735-11 2018 In the DA neurons, upregulated miR-128 expression or sh-AXIN1 increased the positive expression rate of EAAT4 together with mRNA and protein levels, but decreased the mRNA and protein levels of AXIN1, apoptosis rate along with the positive expression rate of AXIN1; however, the opposite trend was found in response to transfection with miR-128 inhibitors. mir-128 31-38 axin 1 Mus musculus 194-199 29552332-0 2018 Correction: Posttranscriptional regulation of Galectin-3 by miR-128 contributes to colorectal cancer progression. mir-128 60-67 galectin 3 Homo sapiens 46-56 30537735-12 2018 CONCLUSION: Evidence from experimental models revealed that miR-128 might reduce apoptosis of DA neurons while increasing the expression of EAAT4 which might be related to the downregulation of AXIN1. mir-128 60-67 axin 1 Mus musculus 194-199 28928797-5 2017 A luciferase assay was applied to assess the binding of miR-128 to c-Met mRNA. mir-128 56-63 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 67-72 28974576-0 2017 MicroRNA miR-128 represses LINE-1 (L1) retrotransposition by down-regulating the nuclear import factor TNPO1. mir-128 9-16 L1 cell adhesion molecule Homo sapiens 27-37 28974576-0 2017 MicroRNA miR-128 represses LINE-1 (L1) retrotransposition by down-regulating the nuclear import factor TNPO1. mir-128 9-16 transportin 1 Homo sapiens 103-108 28974576-7 2017 We found that miR-128 targets the 3" UTR of nuclear import factor transportin 1 (TNPO1) mRNA. mir-128 14-21 transportin 1 Homo sapiens 66-79 28974576-7 2017 We found that miR-128 targets the 3" UTR of nuclear import factor transportin 1 (TNPO1) mRNA. mir-128 14-21 transportin 1 Homo sapiens 81-86 28974576-8 2017 Manipulation of miR-128 and TNPO1 levels demonstrated that induction or depletion of TNPO1 affects L1 retrotransposition and nuclear import of an L1-ribonucleoprotein complex (using L1-encoded ORF1p as a proxy for L1-ribonucleoprotein complexes). mir-128 16-23 transportin 1 Homo sapiens 85-90 28974576-9 2017 Moreover, TNPO1 overexpression partially reversed the repressive effect of miR-128 on L1 retrotransposition. mir-128 75-82 transportin 1 Homo sapiens 10-15 28928797-8 2017 Similarly, miR-128 expression in primary cardiomyocytes cultured under deprivation of oxygen and glucose increased with the culture time and reached a peak at 12 h. c-Met expression decreased significantly (P<0.05) and the ratio of apoptotic cells increased significantly (P<0.05), following transfection of miR-128 mimics. mir-128 314-321 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 165-170 28928797-10 2017 The dual luciferase assay indicated that fluorescence intensity decreased significantly in miR-128 mimics and wild type c-Met group (P<0.05), indicating that miR-128 can directly target c-Met. mir-128 91-98 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 189-194 28928797-10 2017 The dual luciferase assay indicated that fluorescence intensity decreased significantly in miR-128 mimics and wild type c-Met group (P<0.05), indicating that miR-128 can directly target c-Met. mir-128 161-168 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 120-125 28928797-10 2017 The dual luciferase assay indicated that fluorescence intensity decreased significantly in miR-128 mimics and wild type c-Met group (P<0.05), indicating that miR-128 can directly target c-Met. mir-128 161-168 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 189-194 28928797-11 2017 Therefore, the results of the current study suggest that miR-128 may promote myocardial cell injury by regulating c-Met expression. mir-128 57-64 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 114-119 28497421-10 2017 The decrease in miR-128 was sufficient to induce the loss of nephrin and the impairment of filtration barrier function, while the increase of miR-21 exacerbated the process. mir-128 16-23 NPHS1 adhesion molecule, nephrin Homo sapiens 61-68 28497421-11 2017 Snail and phosphatase and tensin homolog (PTEN) were identified as the targets of miR-128 and miR-21. mir-128 82-89 snail family transcriptional repressor 1 Homo sapiens 0-5 28497421-11 2017 Snail and phosphatase and tensin homolog (PTEN) were identified as the targets of miR-128 and miR-21. mir-128 82-89 phosphatase and tensin homolog Homo sapiens 42-46 28497421-12 2017 Decreased miR-128 induced Snail expression, and the increased miR-21 stabilized Snail by regulating the PTEN/Akt/GSK3beta pathway. mir-128 10-17 snail family transcriptional repressor 1 Homo sapiens 26-31 28497421-13 2017 Supplementation of miR-128 and inhibition of miR-21 suppressed Snail expression and prevented the podocyte injury induced by LPS. mir-128 19-26 snail family transcriptional repressor 1 Homo sapiens 63-68 29067466-0 2017 miR-128 enhances dendritic cell-mediated anti-tumor immunity via targeting of p38. mir-128 0-7 mitogen-activated protein kinase 14 Mus musculus 78-81 29067466-8 2017 Additionally, miR-128 inhibited the protein expression of p38 in DCs in a dose-dependent manner, however no significant effect on the p38 mRNA level was observed. mir-128 14-21 mitogen-activated protein kinase 14 Mus musculus 58-61 29067466-9 2017 Furthermore, miR-128 mimic or p38 inhibitor decreased the mRNA expression and secretion of interleukin (IL)-6 and IL-10 cytokines and increased the level of IL-12 in DCs, whereas an miR-128 inhibitor exhibited the opposite effects. mir-128 13-20 interleukin 6 Mus musculus 91-109 29067466-9 2017 Furthermore, miR-128 mimic or p38 inhibitor decreased the mRNA expression and secretion of interleukin (IL)-6 and IL-10 cytokines and increased the level of IL-12 in DCs, whereas an miR-128 inhibitor exhibited the opposite effects. mir-128 13-20 interleukin 10 Mus musculus 114-119 29067466-9 2017 Furthermore, miR-128 mimic or p38 inhibitor decreased the mRNA expression and secretion of interleukin (IL)-6 and IL-10 cytokines and increased the level of IL-12 in DCs, whereas an miR-128 inhibitor exhibited the opposite effects. mir-128 182-189 mitogen-activated protein kinase 14 Mus musculus 30-33 28514100-3 2017 The miR-128/NEK2 pathway has been reported to predict prognosis in colorectal cancer; however, the determination of a relationship between miR-128 and NEK2 in lung cancer has remained elusive. mir-128 139-146 NIMA related kinase 2 Homo sapiens 151-155 29067466-10 2017 These findings suggested that miR-128 regulated the immune response of DCs via p38-downstream cytokines. mir-128 30-37 mitogen-activated protein kinase 14 Mus musculus 79-82 29067466-12 2017 The results therefore suggest that miR-128 enhances the anti-tumor immunity response of DCs via targeting of the p38 mitogen activated protein kinase signaling pathway. mir-128 35-42 mitogen-activated protein kinase 14 Mus musculus 113-116 28514100-4 2017 We explored the association between miR-128 and NEK2 in lung cancer. mir-128 36-43 NIMA related kinase 2 Homo sapiens 48-52 28514100-11 2017 Dual luciferase assay further confirmed that NEK2 was a direct target of miR-128 in lung cancer, and transfection with miR-128 mimic could decrease the NEK2 protein level while the miR-128 inhibitor increased NEK2 expression. mir-128 73-80 NIMA related kinase 2 Homo sapiens 45-49 28514100-11 2017 Dual luciferase assay further confirmed that NEK2 was a direct target of miR-128 in lung cancer, and transfection with miR-128 mimic could decrease the NEK2 protein level while the miR-128 inhibitor increased NEK2 expression. mir-128 73-80 NIMA related kinase 2 Homo sapiens 152-156 28514100-11 2017 Dual luciferase assay further confirmed that NEK2 was a direct target of miR-128 in lung cancer, and transfection with miR-128 mimic could decrease the NEK2 protein level while the miR-128 inhibitor increased NEK2 expression. mir-128 73-80 NIMA related kinase 2 Homo sapiens 152-156 28514100-11 2017 Dual luciferase assay further confirmed that NEK2 was a direct target of miR-128 in lung cancer, and transfection with miR-128 mimic could decrease the NEK2 protein level while the miR-128 inhibitor increased NEK2 expression. mir-128 119-126 NIMA related kinase 2 Homo sapiens 45-49 28514100-11 2017 Dual luciferase assay further confirmed that NEK2 was a direct target of miR-128 in lung cancer, and transfection with miR-128 mimic could decrease the NEK2 protein level while the miR-128 inhibitor increased NEK2 expression. mir-128 119-126 NIMA related kinase 2 Homo sapiens 152-156 28514100-11 2017 Dual luciferase assay further confirmed that NEK2 was a direct target of miR-128 in lung cancer, and transfection with miR-128 mimic could decrease the NEK2 protein level while the miR-128 inhibitor increased NEK2 expression. mir-128 119-126 NIMA related kinase 2 Homo sapiens 152-156 28514100-12 2017 Finally, the apoptotic effect of lung cancer cells induced by miR-128 mimic could be reversed by NEK2 overexpression. mir-128 62-69 NIMA related kinase 2 Homo sapiens 97-101 28514100-13 2017 CONCLUSIONS: NEK2 was regulated by miR-128 in lung cancer and miR-128 induced lung cancer cell apoptosis by mediating NEK2 expression. mir-128 35-42 NIMA related kinase 2 Homo sapiens 13-17 28514100-13 2017 CONCLUSIONS: NEK2 was regulated by miR-128 in lung cancer and miR-128 induced lung cancer cell apoptosis by mediating NEK2 expression. mir-128 35-42 NIMA related kinase 2 Homo sapiens 118-122 28514100-13 2017 CONCLUSIONS: NEK2 was regulated by miR-128 in lung cancer and miR-128 induced lung cancer cell apoptosis by mediating NEK2 expression. mir-128 62-69 NIMA related kinase 2 Homo sapiens 13-17 28514100-13 2017 CONCLUSIONS: NEK2 was regulated by miR-128 in lung cancer and miR-128 induced lung cancer cell apoptosis by mediating NEK2 expression. mir-128 62-69 NIMA related kinase 2 Homo sapiens 118-122 28424413-4 2017 Using prediction tools, western blotting, and luciferase reporter assays, we found that Bmi-1 was the direct target of miR-128. mir-128 119-126 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 88-93 28424413-5 2017 Additionally, overexpression of miR-128 inhibited gastric cancer cell migration, invasion, and proliferation by targeting Bmi-1 in vitro and in vivo. mir-128 32-39 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 122-127 28424413-8 2017 In fact, we found that miR-128 could reverse epithelial-to-mesenchymal transition induced by Bmi-1 via the PI3K/AKT pathway. mir-128 23-30 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 93-98 28424413-8 2017 In fact, we found that miR-128 could reverse epithelial-to-mesenchymal transition induced by Bmi-1 via the PI3K/AKT pathway. mir-128 23-30 AKT serine/threonine kinase 1 Homo sapiens 112-115 27485180-0 2017 miR-128 promoted adipogenic differentiation and inhibited osteogenic differentiation of human mesenchymal stem cells by suppression of VEGF pathway. mir-128 0-7 vascular endothelial growth factor A Homo sapiens 135-139 27485180-8 2017 Furthermore, over-expression of miR-128 down-regulated VEGF expression in adipogenically and osteogenically differentiated cells. mir-128 32-39 vascular endothelial growth factor A Homo sapiens 55-59 27485180-9 2017 We further identified VEGF as a key regulator in miR-128-induced adipogenic and osteogenic differentiation. mir-128 49-56 vascular endothelial growth factor A Homo sapiens 22-26 27485180-11 2017 CONCLUSION: It was indicated that miR-128 could regulate adipogenic and osteogenic differentiation of hMSCs significantly through the suppression of VEGF pathway. mir-128 34-41 vascular endothelial growth factor A Homo sapiens 149-153 28146425-0 2017 Posttranscriptional regulation of Galectin-3 by miR-128 contributes to colorectal cancer progression. mir-128 48-55 galectin 3 Homo sapiens 34-44 28179359-8 2017 SRC knockdown was rescued by antagomir to miR-128. mir-128 42-49 SRC proto-oncogene, non-receptor tyrosine kinase Homo sapiens 0-3 28146425-4 2017 To explore the mechanism for Galectin-3 dysregulation, we found that miR-128 level was frequently down-regulated in CRC and negatively correlated with Galectin-3 level. mir-128 69-76 galectin 3 Homo sapiens 29-39 28146425-4 2017 To explore the mechanism for Galectin-3 dysregulation, we found that miR-128 level was frequently down-regulated in CRC and negatively correlated with Galectin-3 level. mir-128 69-76 galectin 3 Homo sapiens 151-161 28146425-5 2017 Using bioinformatics analysis and experimental validation, we showed that miR-128 could directly target Galectin-3 to repress its protein level. mir-128 74-81 galectin 3 Homo sapiens 104-114 28146425-8 2017 Galectin-3 expression impaired the cancer suppressive effects of miR-128. mir-128 65-72 galectin 3 Homo sapiens 0-10 28114268-10 2017 In addition, miR-128 overexpression significantly decreased the expression levels of microglial M1 phenotypic markers CD86 and CD32, and increased the expression levels of M2 phenotypic markers Arg1 and CD206. mir-128 13-20 CD86 antigen Mus musculus 118-122 27905005-6 2017 To reveal the mechanism for posttranscription of p38alpha protein, we tested the effect of miR-128-3p. mir-128 91-98 mitogen-activated protein kinase 14 Mus musculus 49-57 28114268-10 2017 In addition, miR-128 overexpression significantly decreased the expression levels of microglial M1 phenotypic markers CD86 and CD32, and increased the expression levels of M2 phenotypic markers Arg1 and CD206. mir-128 13-20 mannose receptor, C type 1 Mus musculus 203-208 28114268-11 2017 Furthermore, miR-128 overexpression obviously decreased the concentration of TNF-alpha, IL-1beta, and IL-6. mir-128 13-20 tumor necrosis factor Mus musculus 77-86 28114268-11 2017 Furthermore, miR-128 overexpression obviously decreased the concentration of TNF-alpha, IL-1beta, and IL-6. mir-128 13-20 interleukin 1 beta Mus musculus 88-96 28114268-11 2017 Furthermore, miR-128 overexpression obviously decreased the concentration of TNF-alpha, IL-1beta, and IL-6. mir-128 13-20 interleukin 6 Mus musculus 102-106 28114268-12 2017 We found that miR-128 overexpression significantly downregulated the expression levels of P38 andP-P38. mir-128 14-21 mitogen-activated protein kinase 14 Mus musculus 90-93 28114268-12 2017 We found that miR-128 overexpression significantly downregulated the expression levels of P38 andP-P38. mir-128 14-21 mitogen-activated protein kinase 14 Mus musculus 99-102 28114268-13 2017 CONCLUSIONS Our findings indicate that down-regulation of miR-128 in murine microglial cells may contribute to the development of NPP following SCI via activation of P38. mir-128 58-65 mitogen-activated protein kinase 14 Mus musculus 166-169 28114268-10 2017 In addition, miR-128 overexpression significantly decreased the expression levels of microglial M1 phenotypic markers CD86 and CD32, and increased the expression levels of M2 phenotypic markers Arg1 and CD206. mir-128 13-20 Fc receptor, IgG, low affinity IIb Mus musculus 127-131 28114268-10 2017 In addition, miR-128 overexpression significantly decreased the expression levels of microglial M1 phenotypic markers CD86 and CD32, and increased the expression levels of M2 phenotypic markers Arg1 and CD206. mir-128 13-20 arginase, liver Mus musculus 194-198 27695040-3 2016 In mouse and human ALS spinal cord, the reduction of miR-128 that destabilizes TrkC.T1 mRNA results in up-regulated TrkC.T1 and TNF-alpha in astrocytes. mir-128 53-60 neurotrophic receptor tyrosine kinase 3 Homo sapiens 79-83 27690301-0 2016 MiR-128 reverses the gefitinib resistance of the lung cancer stem cells by inhibiting the c-met/PI3K/AKT pathway. mir-128 0-7 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 90-95 27690301-0 2016 MiR-128 reverses the gefitinib resistance of the lung cancer stem cells by inhibiting the c-met/PI3K/AKT pathway. mir-128 0-7 AKT serine/threonine kinase 1 Homo sapiens 101-104 27690301-5 2016 Mechanically, we showed significant down-regulation of miR-128 in the PC9-CSCs compared with the non-CSCs. mir-128 55-62 proprotein convertase subtilisin/kexin type 9 Homo sapiens 70-73 27690301-6 2016 Overexpression of miR-128 significantly increased the sensitivity of PC9-CSCs to gefitinib-induced apoptosis. mir-128 18-25 proprotein convertase subtilisin/kexin type 9 Homo sapiens 69-72 27690301-7 2016 In addition, the gene of c-met was proved to be directly inhibited by miR-128. mir-128 70-77 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 25-30 27690301-8 2016 Enforced expression of c-met could "rescue" the miR-128 promoted apoptosis and cleavage of caspases in PC9-CSCs treated with gefitinib. mir-128 48-55 MET proto-oncogene, receptor tyrosine kinase Homo sapiens 23-28 27690301-8 2016 Enforced expression of c-met could "rescue" the miR-128 promoted apoptosis and cleavage of caspases in PC9-CSCs treated with gefitinib. mir-128 48-55 proprotein convertase subtilisin/kexin type 9 Homo sapiens 103-106 27695040-3 2016 In mouse and human ALS spinal cord, the reduction of miR-128 that destabilizes TrkC.T1 mRNA results in up-regulated TrkC.T1 and TNF-alpha in astrocytes. mir-128 53-60 neurotrophic receptor tyrosine kinase 3 Homo sapiens 116-120 27695040-3 2016 In mouse and human ALS spinal cord, the reduction of miR-128 that destabilizes TrkC.T1 mRNA results in up-regulated TrkC.T1 and TNF-alpha in astrocytes. mir-128 53-60 tumor necrosis factor Homo sapiens 128-137 26700675-8 2016 Importantly, we identified that the 3"-untranslated region (3"-UTR) of ITGA2 was a direct target of miR-128. mir-128 100-107 integrin subunit alpha 2 Homo sapiens 71-76 26307612-9 2016 We also identified that TROVE2 is a novel target of miR-128 by the luciferase reporter system. mir-128 52-59 Ro60, Y RNA binding protein Homo sapiens 24-30 26307612-11 2016 Finally, the levels of TROVE2 are negatively correlated with miR-128 in astrocytoma tissues. mir-128 61-68 Ro60, Y RNA binding protein Homo sapiens 23-29 27222923-1 2016 MicroRNA-128-3p (miR-128) is a brain-enriched microRNA reported to target Doublecortin (Dcx), a key transcriptional factor during adult neurogenesis. mir-128 17-24 doublecortin Mus musculus 74-86 27322220-6 2016 Knockdown of the most abundant miRNAs in exosomes and the MCF-7 proliferation assay showed that miR-128 in exosomes negatively regulates the level of Bax in MCF-7 recipient cells and inhibits cell proliferation. mir-128 96-103 BCL2 associated X, apoptosis regulator Homo sapiens 150-153 27893811-10 2016 Furthermore, the overexpression of miR-128 alone enhanced apoptotic death of glioma cells through caspase-3/9 activation, poly(ADP ribose) polymerase degradation, reactive oxygen species generation, mitochondrial membrane potential loss, and non-protective autophagy formation. mir-128 35-42 caspase 3 Homo sapiens 98-107 27893811-10 2016 Furthermore, the overexpression of miR-128 alone enhanced apoptotic death of glioma cells through caspase-3/9 activation, poly(ADP ribose) polymerase degradation, reactive oxygen species generation, mitochondrial membrane potential loss, and non-protective autophagy formation. mir-128 35-42 poly(ADP-ribose) polymerase 1 Homo sapiens 122-149 27893811-12 2016 TMZ inhibited mTOR signaling through miR-128 regulation. mir-128 37-44 mechanistic target of rapamycin kinase Homo sapiens 14-18 27893811-13 2016 These results indicate that miR-128-inhibited mTOR signaling is involved in TMZ-mediated cytotoxicity. mir-128 28-35 mechanistic target of rapamycin kinase Homo sapiens 46-50 27222923-1 2016 MicroRNA-128-3p (miR-128) is a brain-enriched microRNA reported to target Doublecortin (Dcx), a key transcriptional factor during adult neurogenesis. mir-128 17-24 doublecortin Mus musculus 88-91 27222923-2 2016 However, the downstream physiological effects of this miR-128-DCX axis remain unclear. mir-128 54-61 doublecortin Mus musculus 62-65 27222923-4 2016 During differentiation of neural stem cells, over-expressing miR-128 with a lentivirus system inhibited the up-regulation of Dcx on Day 5, subsequently decreasing the percentage of TuJ+ cells on Day 16. mir-128 61-68 doublecortin Mus musculus 125-128 27222923-5 2016 Administration of the lentivirus encoding miR-128 into mouse hippocampi significantly impaired water maze learning after 14days, which could be attenuated when the Dcx-encoding virus was delivered simultaneously. mir-128 42-49 doublecortin Mus musculus 164-167 27222923-6 2016 In addition, similar changes including miR-128 up-regulation, Dcx down-regulation and learning defects were observed after a 14-day infusion of Abeta-42, which were also partially reversed by over-expressing Dcx. mir-128 39-46 doublecortin Mus musculus 208-211 27150726-2 2016 In vitro, the expression of PPARG was detected by reverse transcription-quantitative polymerase chain reaction and western blotting in neonatal rat ventricular myocytes (NRVMs) and HEK293 cells transfected with the mimics or inhibitors of miR-128 or control RNA. mir-128 239-246 peroxisome proliferator-activated receptor gamma Rattus norvegicus 28-33 27150726-3 2016 Luciferase reporter assays were used to identify whether PPARG is a direct target of miR-128. mir-128 85-92 peroxisome proliferator activated receptor gamma Homo sapiens 57-62 27150726-7 2016 PPARG mRNA and protein were downregulated in NRVMs transfected with miR-128 mimics, but upregulated by antagomir-128 compared with control. mir-128 68-75 peroxisome proliferator activated receptor gamma Homo sapiens 0-5 27150726-8 2016 This indicates that PPARG is a direct miR-128 target. mir-128 38-45 peroxisome proliferator activated receptor gamma Homo sapiens 20-25 27150726-9 2016 Activation of Akt (p-Akt), Mcl-1 and PPARG expression in the myocardium were increased by miR-128 inhibition. mir-128 90-97 AKT serine/threonine kinase 1 Homo sapiens 14-17 27150726-9 2016 Activation of Akt (p-Akt), Mcl-1 and PPARG expression in the myocardium were increased by miR-128 inhibition. mir-128 90-97 AKT serine/threonine kinase 1 Homo sapiens 19-24 27150726-9 2016 Activation of Akt (p-Akt), Mcl-1 and PPARG expression in the myocardium were increased by miR-128 inhibition. mir-128 90-97 MCL1 apoptosis regulator, BCL2 family member Homo sapiens 27-32 27150726-9 2016 Activation of Akt (p-Akt), Mcl-1 and PPARG expression in the myocardium were increased by miR-128 inhibition. mir-128 90-97 peroxisome proliferator activated receptor gamma Homo sapiens 37-42 27150726-10 2016 Furthermore, miR-128 antagomirs significantly reduced apoptosis in hearts subjected to I/R injury, which was blocked by the PPARG inhibitor GW9662. mir-128 13-20 peroxisome proliferator activated receptor gamma Homo sapiens 124-129 27150726-11 2016 In conclusion, miR-128 inhibition attenuated I/R injury-induced cardiomyocyte apoptosis by the targeted activation of PPARG signaling. mir-128 15-22 peroxisome proliferator activated receptor gamma Homo sapiens 118-123 27341528-0 2016 AurkA controls self-renewal of breast cancer-initiating cells promoting wnt3a stabilization through suppression of miR-128. mir-128 115-122 aurora kinase A Homo sapiens 0-5 27341528-7 2016 Interestingly, we found that AurkA suppressed the expression of miR-128, inhibitor of wnt3a mRNA stabilization. mir-128 64-71 aurora kinase A Homo sapiens 29-34 27341528-7 2016 Interestingly, we found that AurkA suppressed the expression of miR-128, inhibitor of wnt3a mRNA stabilization. mir-128 64-71 Wnt family member 3A Homo sapiens 86-91 27341528-8 2016 Namely, miR-128 suppression realized after AurkA binding to Snail. mir-128 8-15 aurora kinase A Homo sapiens 43-48 27341528-8 2016 Namely, miR-128 suppression realized after AurkA binding to Snail. mir-128 8-15 snail family transcriptional repressor 1 Homo sapiens 60-65 27341528-11 2016 In addition, it suggests a new therapeutic strategy taking advantage of miR-128 to suppress AurkA-Wnt3a signaling. mir-128 72-79 aurora kinase A Homo sapiens 92-97 27341528-11 2016 In addition, it suggests a new therapeutic strategy taking advantage of miR-128 to suppress AurkA-Wnt3a signaling. mir-128 72-79 Wnt family member 3A Homo sapiens 98-103 27133073-6 2016 In addition, mini-hF9 transcripts were accumulated after transfection with miR-128 or miR-125 mimics in E7a and E7b. mir-128 75-82 coagulation factor IX Homo sapiens 18-21 27087048-0 2016 miR-128 regulates differentiation of hair follicle mesenchymal stem cells into smooth muscle cells by targeting SMAD2. mir-128 0-7 SMAD family member 2 Homo sapiens 112-117 27087048-3 2016 In present study, we found that miR-128 was remarkably decreased during the differentiation of hHFMSCs into SMCs induced by transforming growth factor-beta1 (TGF-beta1). mir-128 32-39 transforming growth factor beta 1 Homo sapiens 124-156 27087048-3 2016 In present study, we found that miR-128 was remarkably decreased during the differentiation of hHFMSCs into SMCs induced by transforming growth factor-beta1 (TGF-beta1). mir-128 32-39 transforming growth factor beta 1 Homo sapiens 158-167 27087048-4 2016 Moreover, overexpression of miR-128 led to decreased expression of SMC cellular marker proteins, such as smooth muscle actin (SMA) and calponin, in TGF-beta1-induced SMC differentiation. mir-128 28-35 transforming growth factor beta 1 Homo sapiens 148-157 27087048-5 2016 Further, we identified that miR-128 targeted the 3"-UTR of SMAD2 transcript for translational inhibition of SMAD2 protein, and knockdown of SMAD2 abrogated the promotional effect of antagomir-128 (miR-128 neutralizer) on SMC differentiation. mir-128 28-35 SMAD family member 2 Homo sapiens 59-64 27087048-5 2016 Further, we identified that miR-128 targeted the 3"-UTR of SMAD2 transcript for translational inhibition of SMAD2 protein, and knockdown of SMAD2 abrogated the promotional effect of antagomir-128 (miR-128 neutralizer) on SMC differentiation. mir-128 28-35 SMAD family member 2 Homo sapiens 108-113 27087048-5 2016 Further, we identified that miR-128 targeted the 3"-UTR of SMAD2 transcript for translational inhibition of SMAD2 protein, and knockdown of SMAD2 abrogated the promotional effect of antagomir-128 (miR-128 neutralizer) on SMC differentiation. mir-128 28-35 SMAD family member 2 Homo sapiens 108-113 27087048-6 2016 These results suggest that miR-128 regulates the differentiation of hHFMSCs into SMCs via targeting SMAD2, a main transcription regulator in TGF-beta signaling pathway involving SMC differentiation. mir-128 27-34 SMAD family member 2 Homo sapiens 100-105 27087048-6 2016 These results suggest that miR-128 regulates the differentiation of hHFMSCs into SMCs via targeting SMAD2, a main transcription regulator in TGF-beta signaling pathway involving SMC differentiation. mir-128 27-34 transforming growth factor beta 1 Homo sapiens 141-149 26700675-9 2016 Luciferase reporter assays confirmed that miR-128 binding to the 3"-UTR regions of ITGA2 inhibited the expression of ITGA2 in MG-63 cells. mir-128 42-49 integrin subunit alpha 2 Homo sapiens 83-88 26700675-9 2016 Luciferase reporter assays confirmed that miR-128 binding to the 3"-UTR regions of ITGA2 inhibited the expression of ITGA2 in MG-63 cells. mir-128 42-49 integrin subunit alpha 2 Homo sapiens 117-122 26700675-10 2016 At the same time, overexpressed ITGA2 also reversed EMT inhibited by miR-128. mir-128 69-76 integrin subunit alpha 2 Homo sapiens 32-37 26700675-11 2016 In conclusion, this study suggested that high miR-128 expression suppressed osteosarcoma cell migration, invasion, and EMT development through targeting ITGA2, which may be recommended as a therapeutic target for osteosarcoma. mir-128 46-53 integrin subunit alpha 2 Homo sapiens 153-158 26833195-9 2016 Changes in miR-128 and Sp1 expression levels also affected the protein levels of MyoD and CDKN1A. mir-128 11-18 myogenic differentiation 1 Bos taurus 81-85 27186316-0 2016 miR-128 modulates hepatocellular carcinoma by inhibition of ITGA2 and ITGA5 expression [Retraction]. mir-128 0-7 integrin subunit alpha 2 Homo sapiens 60-65 27186316-0 2016 miR-128 modulates hepatocellular carcinoma by inhibition of ITGA2 and ITGA5 expression [Retraction]. mir-128 0-7 integrin subunit alpha 5 Homo sapiens 70-75 26460960-3 2015 Suppression of VEGF-C by miR-27b, miR-101 and miR-128 was investigated by luciferase assays, Western blot and ELISA. mir-128 46-53 vascular endothelial growth factor C Homo sapiens 15-21 26883496-5 2016 Overexpression of miR-128 suppressed the translation of PCM1, and knockdown of endogenous PCM1 phenocopied the observed effects of miR-128 overexpression. mir-128 18-25 pericentriolar material 1 Homo sapiens 56-60 26883496-5 2016 Overexpression of miR-128 suppressed the translation of PCM1, and knockdown of endogenous PCM1 phenocopied the observed effects of miR-128 overexpression. mir-128 131-138 pericentriolar material 1 Homo sapiens 90-94 26883496-6 2016 Furthermore, concomitant overexpression of PCM1 and miR-128 in NPCs rescued the phenotype associated with miR-128 overexpression, enhancing neurogenesis but inhibiting proliferation, both in vitro and in utero. mir-128 106-113 pericentriolar material 1 Homo sapiens 43-47 27186417-3 2016 Here, we show that miR-128 directly targets PFK liver type (PFKL) in lung cancer cells and regulates endogenous expression of PFKL at both the mRNA and protein levels. mir-128 19-26 phosphofructokinase, liver type Homo sapiens 60-64 27186417-3 2016 Here, we show that miR-128 directly targets PFK liver type (PFKL) in lung cancer cells and regulates endogenous expression of PFKL at both the mRNA and protein levels. mir-128 19-26 phosphofructokinase, liver type Homo sapiens 126-130 27186417-6 2016 Moreover, we observed that miR-128 expression inversely correlated with PFKL mRNA levels in clinic lung cancer samples and that increased PFKL expression predicted poor overall survival in lung cancer patients. mir-128 27-34 phosphofructokinase, liver type Homo sapiens 72-76 27186417-7 2016 Mechanistically, we showed that miR-128 regulates PFKL via a feedback loop that involves inhibition of the AKT signaling pathway. mir-128 32-39 phosphofructokinase, liver type Homo sapiens 50-54 27186417-7 2016 Mechanistically, we showed that miR-128 regulates PFKL via a feedback loop that involves inhibition of the AKT signaling pathway. mir-128 32-39 AKT serine/threonine kinase 1 Homo sapiens 107-110 26460960-9 2015 Overexpression of miR-27b, miR-101, or miR-128 suppressed migration, proliferation activity, and tube formation in HUVECs by repressing VEGF-C secretion in gastric cancer cells. mir-128 39-46 vascular endothelial growth factor C Homo sapiens 136-142 26460960-10 2015 We conclude that miR-27b, miR-101 and miR-128 inhibit angiogenesis by down-regulating VEGF-C expression in gastric cancers. mir-128 38-45 vascular endothelial growth factor C Homo sapiens 86-92 25958325-9 2015 Taken together, these results reveal that there is a novel pathway in skeletal muscle development in which miR-128 regulates myostatin at CDS region to inhibit proliferation but promote differentiation of myoblast cells. mir-128 107-114 myostatin Mus musculus 125-134 26722309-0 2015 miR-128 downregulation promotes growth and metastasis of bladder cancer cells and involves VEGF-C upregulation. mir-128 0-7 vascular endothelial growth factor C Homo sapiens 91-97 26722309-3 2015 To investigate the regulation of miR-128 on VEGF-C expression and their effects on proliferation and metastasis of bladder cancer, T24 and 5637 BC cells were transfected with pre-miR-128, anti-miR-128 and their respective negative control. mir-128 33-40 vascular endothelial growth factor C Homo sapiens 44-50 26722309-5 2015 The present results indicated that miR-128 negatively regulated VEGF-C expression in BC T24 and 5637 BC cells. mir-128 35-42 vascular endothelial growth factor C Homo sapiens 64-70 26722309-6 2015 VEGF-C is a direct target of miR-128 in BC cells. mir-128 29-36 vascular endothelial growth factor C Homo sapiens 0-6 26352220-7 2015 In addition, insulin receptor substrate 1 (IRS1), a key mediator in oncogenic insulin-like growth factor (IGF) signaling, was confirmed as a direct target of miR-128 by a luciferase reporter assay. mir-128 158-165 insulin receptor substrate 1 Homo sapiens 13-41 26352220-7 2015 In addition, insulin receptor substrate 1 (IRS1), a key mediator in oncogenic insulin-like growth factor (IGF) signaling, was confirmed as a direct target of miR-128 by a luciferase reporter assay. mir-128 158-165 insulin receptor substrate 1 Homo sapiens 43-47 26352220-8 2015 Western blot analysis indicated that the overexpression of miR-128 significantly downregulated IRS1 expression and its downstream Akt signaling in CRC cells. mir-128 59-66 insulin receptor substrate 1 Homo sapiens 95-99 26352220-9 2015 Moreover, miR-128 was negatively associated with IRS1 in CRC tissues compared to adjacent non-tumor tissues. mir-128 10-17 insulin receptor substrate 1 Homo sapiens 49-53 26352220-10 2015 Taken together, these data suggested that miR-128 serves as a tumor suppressor and blocks CRC growth and metastasis by targeting IRS1. mir-128 42-49 insulin receptor substrate 1 Homo sapiens 129-133 26550456-0 2015 miR-128 modulates hepatocellular carcinoma by inhibition of ITGA2 and ITGA5 expression. mir-128 0-7 integrin subunit alpha 2 Homo sapiens 60-65 26550456-0 2015 miR-128 modulates hepatocellular carcinoma by inhibition of ITGA2 and ITGA5 expression. mir-128 0-7 integrin subunit alpha 5 Homo sapiens 70-75 26550456-6 2015 ITGA2 and ITGA5 were inversely correlated with the expression of miR-128 in HCC cells. mir-128 65-72 integrin subunit alpha 2 Homo sapiens 0-5 26550456-6 2015 ITGA2 and ITGA5 were inversely correlated with the expression of miR-128 in HCC cells. mir-128 65-72 integrin subunit alpha 5 Homo sapiens 10-15 26550456-7 2015 Importantly, we demonstrate that the overexpression of miR-128 significantly inhibits HCC cell metastasis and stem-cell like properties via ITGA2 and ITGA5. mir-128 55-62 integrin subunit alpha 2 Homo sapiens 140-145 26550456-7 2015 Importantly, we demonstrate that the overexpression of miR-128 significantly inhibits HCC cell metastasis and stem-cell like properties via ITGA2 and ITGA5. mir-128 55-62 integrin subunit alpha 5 Homo sapiens 150-155 26553132-5 2015 It promoted the association of p53 to the promoter region of miR-128, and enhanced the transcriptional activation of p53 on miR-128 expression. mir-128 61-68 tumor protein p53 Homo sapiens 31-34 26553132-6 2015 miR-128 can downregulate prohibitin expression, and subsequently promote apoptosis. mir-128 0-7 prohibitin 1 Homo sapiens 25-35 25781272-1 2015 Brain-enriched miR-128 is repressed in glioma cells, and could inhibit the proliferation of gliomas by targeting genes such as E2F3a and BMI1. mir-128 15-22 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 137-141 25781272-7 2015 Moreover, we observed that the expression of CORO1C, TROVE2, and HnRNPF were higher in glioma cell lines compared to normal brain tissues and presented a tendency toward downregulation after overexpression of miR-128 in T98G cells. mir-128 209-216 coronin 1C Homo sapiens 45-51 25781272-7 2015 Moreover, we observed that the expression of CORO1C, TROVE2, and HnRNPF were higher in glioma cell lines compared to normal brain tissues and presented a tendency toward downregulation after overexpression of miR-128 in T98G cells. mir-128 209-216 Ro60, Y RNA binding protein Homo sapiens 53-59 25781272-7 2015 Moreover, we observed that the expression of CORO1C, TROVE2, and HnRNPF were higher in glioma cell lines compared to normal brain tissues and presented a tendency toward downregulation after overexpression of miR-128 in T98G cells. mir-128 209-216 heterogeneous nuclear ribonucleoprotein F Homo sapiens 65-71 25742789-5 2015 Furthermore, we demonstrated that three murine microRNAs, namely miR-128, -134, and -330, can target the three Mmps Mmp3, Mmp10, and Mmp13, respectively. mir-128 65-72 matrix metallopeptidase 13 Mus musculus 111-115 25921099-0 2015 miR-128 modulates chemosensitivity and invasion of prostate cancer cells through targeting ZEB1. mir-128 0-7 zinc finger E-box binding homeobox 1 Homo sapiens 91-95 25742789-5 2015 Furthermore, we demonstrated that three murine microRNAs, namely miR-128, -134, and -330, can target the three Mmps Mmp3, Mmp10, and Mmp13, respectively. mir-128 65-72 matrix metallopeptidase 3 Mus musculus 116-120 25742789-5 2015 Furthermore, we demonstrated that three murine microRNAs, namely miR-128, -134, and -330, can target the three Mmps Mmp3, Mmp10, and Mmp13, respectively. mir-128 65-72 matrix metallopeptidase 10 Mus musculus 122-127 25742789-5 2015 Furthermore, we demonstrated that three murine microRNAs, namely miR-128, -134, and -330, can target the three Mmps Mmp3, Mmp10, and Mmp13, respectively. mir-128 65-72 matrix metallopeptidase 13 Mus musculus 133-138 25246803-2 2014 Especially, it has been demonstrated that miR-128 may play an important role in the proliferation of human osteosarcoma cells in vitro by directly inhibiting PTEN, which functions as a tumor suppressor in this malignancy. mir-128 42-49 phosphatase and tensin homolog Homo sapiens 158-162 25556700-0 2015 miR-128 regulates neuronal migration, outgrowth and intrinsic excitability via the intellectual disability gene Phf6. mir-128 0-7 PHD finger protein 6 Mus musculus 112-116 25556700-5 2015 We show that Phf6, a gene mutated in the cognitive disorder Borjeson-Forssman-Lehmann syndrome, is an important regulatory target for miR-128. mir-128 134-141 PHD finger protein 6 Mus musculus 13-17 25556700-6 2015 Restoring PHF6 expression counteracts the deleterious effect of miR-128 on neuronal migration, outgrowth and intrinsic physiological properties. mir-128 64-71 PHD finger protein 6 Mus musculus 10-14 25556700-7 2015 Our results place miR-128 upstream of PHF6 in a pathway vital for cortical lamination as well as for the development of neuronal morphology and intrinsic excitability. mir-128 18-25 PHD finger protein 6 Mus musculus 38-42 25248111-6 2014 Overexpression of miR-128 resensitized SKOV3/CP cells to cisplatin and reduced the expression of cisplatin-resistant-related proteins ABCC5 and Bmi-1, whereas miR-128 inhibitors increased cisplatin resistance in SKOV3 cells. mir-128 18-25 ATP binding cassette subfamily C member 5 Homo sapiens 134-139 25248111-6 2014 Overexpression of miR-128 resensitized SKOV3/CP cells to cisplatin and reduced the expression of cisplatin-resistant-related proteins ABCC5 and Bmi-1, whereas miR-128 inhibitors increased cisplatin resistance in SKOV3 cells. mir-128 18-25 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 144-149 25246803-9 2014 CONCLUSION: These findings indicate for the first time that the deregulation of miR-128 and its target gene PTEN may be involved in the aggressive progression of human osteosarcoma. mir-128 80-87 phosphatase and tensin homolog Homo sapiens 108-112 25246803-10 2014 Notably, the upregulation of miR-128 cooperating with the downregulation of PTEN may confer an unfavorable prognosis in patients with this malignancy. mir-128 29-36 phosphatase and tensin homolog Homo sapiens 76-80 24959930-11 2014 The CGGA data showed that the Pearson correlation index between SNAI1 and miR-128 was negatively correlated. mir-128 74-81 snail family transcriptional repressor 1 Homo sapiens 64-69 24859886-0 2014 Loss of SNAIL inhibits cellular growth and metabolism through the miR-128-mediated RPS6KB1/HIF-1alpha/PKM2 signaling pathway in prostate cancer cells. mir-128 66-73 snail family transcriptional repressor 1 Homo sapiens 8-13 24859886-0 2014 Loss of SNAIL inhibits cellular growth and metabolism through the miR-128-mediated RPS6KB1/HIF-1alpha/PKM2 signaling pathway in prostate cancer cells. mir-128 66-73 ribosomal protein S6 kinase B1 Homo sapiens 83-90 24859886-0 2014 Loss of SNAIL inhibits cellular growth and metabolism through the miR-128-mediated RPS6KB1/HIF-1alpha/PKM2 signaling pathway in prostate cancer cells. mir-128 66-73 hypoxia inducible factor 1 subunit alpha Homo sapiens 91-101 24859886-0 2014 Loss of SNAIL inhibits cellular growth and metabolism through the miR-128-mediated RPS6KB1/HIF-1alpha/PKM2 signaling pathway in prostate cancer cells. mir-128 66-73 pyruvate kinase M1/2 Homo sapiens 102-106 24859886-9 2014 Furthermore, down-expression of miR-128 partially restored the effect of si-SNAIL on the suppression of cellular growth, metabolism, and RPS6KB1/HIF-1alpha/PKM2 signaling pathway. mir-128 32-39 snail family transcriptional repressor 1 Homo sapiens 76-81 24859886-9 2014 Furthermore, down-expression of miR-128 partially restored the effect of si-SNAIL on the suppression of cellular growth, metabolism, and RPS6KB1/HIF-1alpha/PKM2 signaling pathway. mir-128 32-39 ribosomal protein S6 kinase B1 Homo sapiens 137-144 24859886-9 2014 Furthermore, down-expression of miR-128 partially restored the effect of si-SNAIL on the suppression of cellular growth, metabolism, and RPS6KB1/HIF-1alpha/PKM2 signaling pathway. mir-128 32-39 hypoxia inducible factor 1 subunit alpha Homo sapiens 145-155 24859886-9 2014 Furthermore, down-expression of miR-128 partially restored the effect of si-SNAIL on the suppression of cellular growth, metabolism, and RPS6KB1/HIF-1alpha/PKM2 signaling pathway. mir-128 32-39 pyruvate kinase M1/2 Homo sapiens 156-160 24859886-10 2014 To our knowledge, it is the first time to demonstrate that SNAIL/miR-128/RPS6KB1 pathway plays a critical role in the progression of PCa. mir-128 65-72 snail family transcriptional repressor 1 Homo sapiens 59-64 24859886-10 2014 To our knowledge, it is the first time to demonstrate that SNAIL/miR-128/RPS6KB1 pathway plays a critical role in the progression of PCa. mir-128 65-72 ribosomal protein S6 kinase B1 Homo sapiens 73-80 25001183-5 2014 Interestingly, ectopic miR-128 overexpression could significantly inhibit vascular endothelial growth factor (VEGF)-C expression and reduce the activity of a luciferase reporter containing the VEGF-C 3"-untranslated region. mir-128 23-30 vascular endothelial growth factor A Homo sapiens 74-108 25001183-5 2014 Interestingly, ectopic miR-128 overexpression could significantly inhibit vascular endothelial growth factor (VEGF)-C expression and reduce the activity of a luciferase reporter containing the VEGF-C 3"-untranslated region. mir-128 23-30 vascular endothelial growth factor A Homo sapiens 110-114 25001183-5 2014 Interestingly, ectopic miR-128 overexpression could significantly inhibit vascular endothelial growth factor (VEGF)-C expression and reduce the activity of a luciferase reporter containing the VEGF-C 3"-untranslated region. mir-128 23-30 vascular endothelial growth factor C Homo sapiens 193-199 25001183-6 2014 In addition, overexpression of miR-128 in NSCLC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGF-A, vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3, critical factors responsible for cancer angiogenesis and lymphangiogenesis, and subsequently decreased phosphorylation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (AKT) and p38 signalling pathways. mir-128 31-38 vascular endothelial growth factor A Homo sapiens 143-149 25001183-6 2014 In addition, overexpression of miR-128 in NSCLC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGF-A, vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3, critical factors responsible for cancer angiogenesis and lymphangiogenesis, and subsequently decreased phosphorylation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (AKT) and p38 signalling pathways. mir-128 31-38 vascular endothelial growth factor A Homo sapiens 151-185 25001183-6 2014 In addition, overexpression of miR-128 in NSCLC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGF-A, vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3, critical factors responsible for cancer angiogenesis and lymphangiogenesis, and subsequently decreased phosphorylation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (AKT) and p38 signalling pathways. mir-128 31-38 kinase insert domain receptor Homo sapiens 196-204 25001183-6 2014 In addition, overexpression of miR-128 in NSCLC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGF-A, vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3, critical factors responsible for cancer angiogenesis and lymphangiogenesis, and subsequently decreased phosphorylation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (AKT) and p38 signalling pathways. mir-128 31-38 fms related receptor tyrosine kinase 4 Homo sapiens 209-216 25001183-6 2014 In addition, overexpression of miR-128 in NSCLC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGF-A, vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3, critical factors responsible for cancer angiogenesis and lymphangiogenesis, and subsequently decreased phosphorylation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (AKT) and p38 signalling pathways. mir-128 31-38 mitogen-activated protein kinase 1 Homo sapiens 340-377 25001183-6 2014 In addition, overexpression of miR-128 in NSCLC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGF-A, vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3, critical factors responsible for cancer angiogenesis and lymphangiogenesis, and subsequently decreased phosphorylation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (AKT) and p38 signalling pathways. mir-128 31-38 mitogen-activated protein kinase 1 Homo sapiens 379-382 25001183-6 2014 In addition, overexpression of miR-128 in NSCLC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGF-A, vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3, critical factors responsible for cancer angiogenesis and lymphangiogenesis, and subsequently decreased phosphorylation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (AKT) and p38 signalling pathways. mir-128 31-38 AKT serine/threonine kinase 1 Homo sapiens 416-419 25001183-6 2014 In addition, overexpression of miR-128 in NSCLC cells and human umbilical vein endothelial cells (HUVECs) cells led to decreased expression of VEGF-A, vascular endothelial growth factor receptor (VEGFR)-2 and VEGFR-3, critical factors responsible for cancer angiogenesis and lymphangiogenesis, and subsequently decreased phosphorylation of extracellular signal-regulated kinase (ERK), phosphatidylinositol 3-kinase (AKT) and p38 signalling pathways. mir-128 31-38 mitogen-activated protein kinase 1 Homo sapiens 425-428 25001183-8 2014 These findings suggest that miR-128 could play a role in NSCLC tumourigenesis at least in part by modulation of angiogenesis and lymphangiogenesis through targeting VEGF-C, and could simultaneously block ERK, AKT and p38 signalling pathways. mir-128 28-35 vascular endothelial growth factor C Homo sapiens 165-171 25001183-8 2014 These findings suggest that miR-128 could play a role in NSCLC tumourigenesis at least in part by modulation of angiogenesis and lymphangiogenesis through targeting VEGF-C, and could simultaneously block ERK, AKT and p38 signalling pathways. mir-128 28-35 mitogen-activated protein kinase 1 Homo sapiens 204-207 25001183-8 2014 These findings suggest that miR-128 could play a role in NSCLC tumourigenesis at least in part by modulation of angiogenesis and lymphangiogenesis through targeting VEGF-C, and could simultaneously block ERK, AKT and p38 signalling pathways. mir-128 28-35 AKT serine/threonine kinase 1 Homo sapiens 209-212 25001183-8 2014 These findings suggest that miR-128 could play a role in NSCLC tumourigenesis at least in part by modulation of angiogenesis and lymphangiogenesis through targeting VEGF-C, and could simultaneously block ERK, AKT and p38 signalling pathways. mir-128 28-35 mitogen-activated protein kinase 1 Homo sapiens 217-220 25017996-0 2014 miR-128 and miR-149 enhance the chemosensitivity of temozolomide by Rap1B-mediated cytoskeletal remodeling in glioblastoma. mir-128 0-7 RAP1B, member of RAS oncogene family Homo sapiens 68-73 24959930-13 2014 Moreover, introduction of miR-128 anti-sense oligonucleotide alleviated the cell cycle retardation, proliferation and invasion inhibition induced by SNAI1 shRNA. mir-128 26-33 snail family transcriptional repressor 1 Homo sapiens 149-154 24415783-7 2014 Macrophage colony-stimulating factor (M-CSF) was identified as a target of miR-128, and increased miR-128 levels in epithelial cells due to infection with strain SE2472 significantly decreased the level of cell-secreted M-CSF, leading to impaired M-CSF-mediated macrophage recruitment. mir-128 75-82 colony stimulating factor 1 (macrophage) Mus musculus 38-43 24415783-7 2014 Macrophage colony-stimulating factor (M-CSF) was identified as a target of miR-128, and increased miR-128 levels in epithelial cells due to infection with strain SE2472 significantly decreased the level of cell-secreted M-CSF, leading to impaired M-CSF-mediated macrophage recruitment. mir-128 75-82 colony stimulating factor 1 (macrophage) Mus musculus 220-225 24415783-7 2014 Macrophage colony-stimulating factor (M-CSF) was identified as a target of miR-128, and increased miR-128 levels in epithelial cells due to infection with strain SE2472 significantly decreased the level of cell-secreted M-CSF, leading to impaired M-CSF-mediated macrophage recruitment. mir-128 75-82 colony stimulating factor 1 (macrophage) Mus musculus 220-225 24415783-7 2014 Macrophage colony-stimulating factor (M-CSF) was identified as a target of miR-128, and increased miR-128 levels in epithelial cells due to infection with strain SE2472 significantly decreased the level of cell-secreted M-CSF, leading to impaired M-CSF-mediated macrophage recruitment. mir-128 98-105 colony stimulating factor 1 (macrophage) Mus musculus 38-43 24415783-7 2014 Macrophage colony-stimulating factor (M-CSF) was identified as a target of miR-128, and increased miR-128 levels in epithelial cells due to infection with strain SE2472 significantly decreased the level of cell-secreted M-CSF, leading to impaired M-CSF-mediated macrophage recruitment. mir-128 98-105 colony stimulating factor 1 (macrophage) Mus musculus 220-225 24415783-7 2014 Macrophage colony-stimulating factor (M-CSF) was identified as a target of miR-128, and increased miR-128 levels in epithelial cells due to infection with strain SE2472 significantly decreased the level of cell-secreted M-CSF, leading to impaired M-CSF-mediated macrophage recruitment. mir-128 98-105 colony stimulating factor 1 (macrophage) Mus musculus 220-225 24415783-9 2014 Moreover, intragastric delivery of anti-miR-128 antagomir into mice significantly increased M-CSF-mediated macrophage recruitment and suppressed Salmonella infection. mir-128 40-47 colony stimulating factor 1 (macrophage) Mus musculus 92-97 24311694-3 2013 miR-128 governs motor activity by suppressing the expression of various ion channels and signaling components of the extracellular signal-regulated kinase ERK2 network that regulate neuronal excitability. mir-128 0-7 mitogen-activated protein kinase 1 Mus musculus 155-159 24132591-5 2014 At the molecular level, our results demonstrated that miR-128 overexpression could repress expression of PTEN by directly targeting PTEN 3"-untranslated region. mir-128 54-61 phosphatase and tensin homolog Homo sapiens 105-109 24132591-5 2014 At the molecular level, our results demonstrated that miR-128 overexpression could repress expression of PTEN by directly targeting PTEN 3"-untranslated region. mir-128 54-61 phosphatase and tensin homolog Homo sapiens 132-136 24132591-7 2014 Therefore, our results suggest that miR-128 plays an important role in the proliferation of human osteosarcoma cells by directly regulation of PTEN/AKT signaling. mir-128 36-43 phosphatase and tensin homolog Homo sapiens 143-147 24448358-10 2014 Expression of miR-128, -10b, -502-3p and -192 differed between SCC and AC, and miR-128 and -192 - between NLP and NSCLC. mir-128 14-21 serpin family B member 3 Homo sapiens 63-66 24046120-3 2014 In particular, we examined miR-128 expression, which is thought to target NEK2. mir-128 27-34 NIMA related kinase 2 Homo sapiens 74-78 24046120-6 2014 RESULTS: MiR-128 inhibited NEK2 expression and cancer cell proliferation via cell cycle arrest. mir-128 9-16 NIMA related kinase 2 Homo sapiens 27-31 24046120-12 2014 CONCLUSIONS: NEK2 may be an independent prognostic factor for CRC and was regulated by miR-128, a microRNA that was subjected to epigenetic regulation. mir-128 87-94 NIMA related kinase 2 Homo sapiens 13-17 23835497-5 2013 Target site prediction algorithms indicated that miR-128 binds the 3"-untranslated regions of erythropoietin-producing hepatocellular receptor (Eph)B1 and EphB2 mRNAs. mir-128 49-56 EPH receptor B1 Homo sapiens 94-150 24055866-0 2013 miR-128 regulates non-myocyte hyperplasia, deposition of extracellular matrix and Islet1 expression during newt cardiac regeneration. mir-128 0-7 ISL LIM homeobox 1 Homo sapiens 82-88 24055866-9 2013 From these studies we conclude that miR-128 regulates both cardiac hyperplasia and Islet1 expression during newt heart regeneration and that this information could be translated into future mammalian cardiac studies. mir-128 36-43 ISL LIM homeobox 1 Homo sapiens 83-89 24141048-7 2013 Co-transfection of HspB1 with specific inhibitors of miR-20a or miR-128 prevented the decrease in PDZ-RhoGEF and blocked the neurite growth promoting effects of HspB1. mir-128 64-71 heat shock protein family B (small) member 1 Rattus norvegicus 19-24 24141048-7 2013 Co-transfection of HspB1 with specific inhibitors of miR-20a or miR-128 prevented the decrease in PDZ-RhoGEF and blocked the neurite growth promoting effects of HspB1. mir-128 64-71 heat shock protein family B (small) member 1 Rattus norvegicus 161-166 23835497-5 2013 Target site prediction algorithms indicated that miR-128 binds the 3"-untranslated regions of erythropoietin-producing hepatocellular receptor (Eph)B1 and EphB2 mRNAs. mir-128 49-56 EPH receptor B2 Homo sapiens 155-160 23835497-6 2013 Luciferase reporter assays confirmed that miR-128 binds and regulates EphB1 and EphB2 mRNAs. mir-128 42-49 EPH receptor B1 Homo sapiens 70-75 23835497-6 2013 Luciferase reporter assays confirmed that miR-128 binds and regulates EphB1 and EphB2 mRNAs. mir-128 42-49 EPH receptor B2 Homo sapiens 80-85 23835497-7 2013 Overexpression of EphB2 reduced the ability of miR-128 to promote cell-cell adhesion. mir-128 47-54 EPH receptor B2 Homo sapiens 18-23 23835497-8 2013 The wound-healing assay indicated that miR-128 significantly inhibited cell migration via EphB2. mir-128 39-46 EPH receptor B2 Homo sapiens 90-95 23835497-9 2013 This study revealed the novel functions of miR-128 in cell-cell adhesion and cell migration in glioma cells through the regulation of EphB2, and identified EphB1 and EphB2 as novel miR-128 targets. mir-128 43-50 EPH receptor B2 Homo sapiens 134-139 23835497-9 2013 This study revealed the novel functions of miR-128 in cell-cell adhesion and cell migration in glioma cells through the regulation of EphB2, and identified EphB1 and EphB2 as novel miR-128 targets. mir-128 181-188 EPH receptor B1 Homo sapiens 156-161 23835497-9 2013 This study revealed the novel functions of miR-128 in cell-cell adhesion and cell migration in glioma cells through the regulation of EphB2, and identified EphB1 and EphB2 as novel miR-128 targets. mir-128 181-188 EPH receptor B2 Homo sapiens 166-171 23482671-13 2013 The inhibition of Bmi1 by NPV-LDE-225 was regulated by induction of miR-128. mir-128 68-75 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 18-22 23526655-2 2013 We have confirmed that Bax is the target of miR-128 by negative post-transcriptional regulation. mir-128 44-51 BCL2 associated X, apoptosis regulator Homo sapiens 23-26 23526655-3 2013 miR-128 and Bax were detected in the breast cancer cell line, MDA-MB-231, which was then transfected with miR-128 MIMIC (precursor of miR-128) or AMO (antisense-miR-128 oligonucleotides). mir-128 106-113 BCL2 associated X, apoptosis regulator Homo sapiens 12-15 23526655-3 2013 miR-128 and Bax were detected in the breast cancer cell line, MDA-MB-231, which was then transfected with miR-128 MIMIC (precursor of miR-128) or AMO (antisense-miR-128 oligonucleotides). mir-128 106-113 BCL2 associated X, apoptosis regulator Homo sapiens 12-15 22909061-0 2012 Regulation of colony stimulating factor-1 expression and ovarian cancer cell behavior in vitro by miR-128 and miR-152. mir-128 98-105 colony stimulating factor 1 Homo sapiens 14-41 23492773-10 2013 Pretreatment with PUMA and Bak siRNAs abolished miR-128-induced apoptosis in HCT116 p53+/+ and HCT116 p53-/- cells. mir-128 48-55 tumor protein p53 Homo sapiens 84-87 23492773-10 2013 Pretreatment with PUMA and Bak siRNAs abolished miR-128-induced apoptosis in HCT116 p53+/+ and HCT116 p53-/- cells. mir-128 48-55 tumor protein p53 Homo sapiens 102-105 23492773-11 2013 Taken together, we present the first evidence of miR-128 to be a new component joining the p53 network. mir-128 49-56 tumor protein p53 Homo sapiens 91-94 23492773-12 2013 This study emphasizes that miR-128 is a novel mitochondria-targeted miRNA that can be further evaluated as a chemotherapeutic agent for human cancers as it induces apoptosis irrespective of p53 status. mir-128 27-34 tumor protein p53 Homo sapiens 190-193 23041308-9 2013 In particular, in HSC overexpressing miR-128, many members of the chemokine family were bound to the Ago2 repression complex. mir-128 37-44 argonaute RISC catalytic component 2 Homo sapiens 101-105 23567619-12 2013 The inhibition of Bmi-1 by NVP-LDE-225 was regulated by upregulation of miR-128. mir-128 72-79 Bmi1 polycomb ring finger oncogene Mus musculus 18-23 22614013-6 2013 Furthermore, we identified that miR-26b and miR-128 affected pituitary tumor cell behavior through regulation of their direct targets, PTEN and BMI1, respectively. mir-128 44-51 phosphatase and tensin homolog Homo sapiens 135-139 22614013-6 2013 Furthermore, we identified that miR-26b and miR-128 affected pituitary tumor cell behavior through regulation of their direct targets, PTEN and BMI1, respectively. mir-128 44-51 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 144-148 22614013-7 2013 In addition, we found that miR-128 through BMI1 direct binding on the PTEN promoter affected PTEN expression levels and AKT activity in the pituitary tumor cells. mir-128 27-34 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 43-47 22614013-7 2013 In addition, we found that miR-128 through BMI1 direct binding on the PTEN promoter affected PTEN expression levels and AKT activity in the pituitary tumor cells. mir-128 27-34 phosphatase and tensin homolog Homo sapiens 70-74 22614013-7 2013 In addition, we found that miR-128 through BMI1 direct binding on the PTEN promoter affected PTEN expression levels and AKT activity in the pituitary tumor cells. mir-128 27-34 phosphatase and tensin homolog Homo sapiens 93-97 22614013-7 2013 In addition, we found that miR-128 through BMI1 direct binding on the PTEN promoter affected PTEN expression levels and AKT activity in the pituitary tumor cells. mir-128 27-34 AKT serine/threonine kinase 1 Homo sapiens 120-123 22614013-11 2013 MiR-26b suppression and miR-128 upregulation could have therapeutic potential in GH-producing pituitary tumor patients. mir-128 24-31 growth hormone 1 Homo sapiens 81-83 23492773-0 2013 miR-128 exerts pro-apoptotic effect in a p53 transcription-dependent and -independent manner via PUMA-Bak axis. mir-128 0-7 tumor protein p53 Homo sapiens 41-44 23492773-4 2013 We herein demonstrate that miR-128 positively regulates p53 activity. mir-128 27-34 tumor protein p53 Homo sapiens 56-59 23492773-6 2013 miR-128 inhibition of SIRT1 led to an increase in acetylated p53 and its transcriptional targets. mir-128 0-7 tumor protein p53 Homo sapiens 61-64 23492773-8 2013 We further demonstrated that miR-128 augments the antitumor effect of compounds that target the p53 pathway. mir-128 29-36 tumor protein p53 Homo sapiens 96-99 23492773-9 2013 Furthermore, miR-128 induces apoptosis in wild (WT) p53 as well as in mutant p53-expressing cells in a p53-dependent and -independent manner via induction of PUMA. mir-128 13-20 tumor protein p53 Homo sapiens 52-55 23492773-9 2013 Furthermore, miR-128 induces apoptosis in wild (WT) p53 as well as in mutant p53-expressing cells in a p53-dependent and -independent manner via induction of PUMA. mir-128 13-20 tumor protein p53 Homo sapiens 77-80 23492773-9 2013 Furthermore, miR-128 induces apoptosis in wild (WT) p53 as well as in mutant p53-expressing cells in a p53-dependent and -independent manner via induction of PUMA. mir-128 13-20 tumor protein p53 Homo sapiens 77-80 22922228-3 2012 Here, we demonstrate for the first time that platelet-derived growth factor-B (PDGF-B), a potent angiogenic growth factor involved in GBM development and progression, promotes downregulation of pro-oncogenic (miR-21) and anti-oncogenic (miR-128) miRNAs, as well as upregulation/downregulation of several miRNAs involved in GBM pathology. mir-128 237-244 platelet derived growth factor subunit B Homo sapiens 45-77 22922228-3 2012 Here, we demonstrate for the first time that platelet-derived growth factor-B (PDGF-B), a potent angiogenic growth factor involved in GBM development and progression, promotes downregulation of pro-oncogenic (miR-21) and anti-oncogenic (miR-128) miRNAs, as well as upregulation/downregulation of several miRNAs involved in GBM pathology. mir-128 237-244 platelet derived growth factor subunit B Homo sapiens 79-85 21953503-0 2011 Reduced miR-128 in breast tumor-initiating cells induces chemotherapeutic resistance via Bmi-1 and ABCC5. mir-128 8-15 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 89-94 23056445-9 2012 Overexpression of miR-128 alone or with miR-103 and miR-137 significantly reduced endogenous neurofibromin 1 protein levels, while antisense inhibition of these microRNAs enhanced translation of endogenous neurofibromin 1 and reporter in primary cultures of hippocampal neurons. mir-128 18-25 neurofibromin 1 Homo sapiens 93-108 23056445-9 2012 Overexpression of miR-128 alone or with miR-103 and miR-137 significantly reduced endogenous neurofibromin 1 protein levels, while antisense inhibition of these microRNAs enhanced translation of endogenous neurofibromin 1 and reporter in primary cultures of hippocampal neurons. mir-128 18-25 neurofibromin 1 Homo sapiens 206-221 22442669-3 2012 Overexpression of miR-128 suppressed p70S6K1 and its downstream signaling molecules such as HIF-1 and VEGF expression, and attenuated cell proliferation, tumor growth and angiogenesis. mir-128 18-25 vascular endothelial growth factor A Homo sapiens 102-106 21953503-0 2011 Reduced miR-128 in breast tumor-initiating cells induces chemotherapeutic resistance via Bmi-1 and ABCC5. mir-128 8-15 ATP binding cassette subfamily C member 5 Homo sapiens 99-104 21953503-2 2011 EXPERIMENTAL DESIGN: Lentivirus-mediated miR-128 transduction was done in BT-ICs, enriched by mammosphere cultures or CD44(+)CD24(-) fluorescence-activated cell sorting. mir-128 41-48 CD44 molecule (Indian blood group) Homo sapiens 118-122 21953503-2 2011 EXPERIMENTAL DESIGN: Lentivirus-mediated miR-128 transduction was done in BT-ICs, enriched by mammosphere cultures or CD44(+)CD24(-) fluorescence-activated cell sorting. mir-128 41-48 CD24 molecule Homo sapiens 125-129 21953503-5 2011 RESULTS: MiR-128 was significantly reduced in chemoresistant BT-ICs enriched from breast cancer cell lines and primary breast tumors (P < 0.01), accompanied by an overexpression of Bmi-1 and ABCC5, which were identified as targets of miR-128. mir-128 9-16 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 184-189 21953503-5 2011 RESULTS: MiR-128 was significantly reduced in chemoresistant BT-ICs enriched from breast cancer cell lines and primary breast tumors (P < 0.01), accompanied by an overexpression of Bmi-1 and ABCC5, which were identified as targets of miR-128. mir-128 9-16 ATP binding cassette subfamily C member 5 Homo sapiens 194-199 21953503-6 2011 Ectopic expression of miR-128 reduced the protein levels of Bmi-1 and ABCC5 in BT-ICs, along with decreased cell viability (P < 0.001) and increased apoptosis (P < 0.001) and DNA damage (P < 0.001) in the presence of doxorubicin. mir-128 22-29 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 60-65 21953503-6 2011 Ectopic expression of miR-128 reduced the protein levels of Bmi-1 and ABCC5 in BT-ICs, along with decreased cell viability (P < 0.001) and increased apoptosis (P < 0.001) and DNA damage (P < 0.001) in the presence of doxorubicin. mir-128 22-29 ATP binding cassette subfamily C member 5 Homo sapiens 70-75 21953503-8 2011 CONCLUSIONS: Reduction in miR-128 leading to Bmi-1 and ABCC5 overexpression is a stem cell-like feature of BT-ICs, which contributes to chemotherapeutic resistance in breast cancers. mir-128 26-33 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 45-50 21953503-8 2011 CONCLUSIONS: Reduction in miR-128 leading to Bmi-1 and ABCC5 overexpression is a stem cell-like feature of BT-ICs, which contributes to chemotherapeutic resistance in breast cancers. mir-128 26-33 ATP binding cassette subfamily C member 5 Homo sapiens 55-60 21596314-4 2011 miR-128 represses NMD by targeting the RNA helicase UPF1 and the exon-junction complex core component MLN51. mir-128 0-7 UPF1 RNA helicase and ATPase Homo sapiens 52-56 19010882-5 2008 miR-128 caused a striking decrease in expression of the Bmi-1 oncogene, by direct regulation of the Bmi-1 mRNA 3"-untranslated region, through a single miR-128 binding site. mir-128 0-7 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 56-61 19010882-5 2008 miR-128 caused a striking decrease in expression of the Bmi-1 oncogene, by direct regulation of the Bmi-1 mRNA 3"-untranslated region, through a single miR-128 binding site. mir-128 0-7 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 100-105 19010882-5 2008 miR-128 caused a striking decrease in expression of the Bmi-1 oncogene, by direct regulation of the Bmi-1 mRNA 3"-untranslated region, through a single miR-128 binding site. mir-128 152-159 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 56-61 19010882-5 2008 miR-128 caused a striking decrease in expression of the Bmi-1 oncogene, by direct regulation of the Bmi-1 mRNA 3"-untranslated region, through a single miR-128 binding site. mir-128 152-159 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 100-105 19010882-8 2008 We found that miR-128 expression caused a decrease in histone methylation (H3K27me(3)) and Akt phosphorylation, and up-regulation of p21(CIP1) levels, consistent with Bmi-1 down-regulation. mir-128 14-21 H3 histone pseudogene 16 Homo sapiens 133-136 19010882-8 2008 We found that miR-128 expression caused a decrease in histone methylation (H3K27me(3)) and Akt phosphorylation, and up-regulation of p21(CIP1) levels, consistent with Bmi-1 down-regulation. mir-128 14-21 cyclin dependent kinase inhibitor 1A Homo sapiens 137-141 19010882-8 2008 We found that miR-128 expression caused a decrease in histone methylation (H3K27me(3)) and Akt phosphorylation, and up-regulation of p21(CIP1) levels, consistent with Bmi-1 down-regulation. mir-128 14-21 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 167-172 19010882-10 2008 This showed that miR-128 specifically blocked glioma self-renewal consistent with Bmi-1 down-regulation. mir-128 17-24 BMI1 proto-oncogene, polycomb ring finger Homo sapiens 82-87 21596314-4 2011 miR-128 represses NMD by targeting the RNA helicase UPF1 and the exon-junction complex core component MLN51. mir-128 0-7 CASC3 exon junction complex subunit Homo sapiens 102-107 21372826-10 2011 The up-regulation of miR-128 by Rh2 was further verified in human U251, T98MG and A172 cells using quantitative real-time PCR. mir-128 21-28 Rh associated glycoprotein Homo sapiens 32-35 21372826-11 2011 In U251 cells, transfection of a miR-128 inhibitor (50 nmol/L) prevented the overexpression of miR-128 by Rh2, and significantly blunted Rh2-induced cytotoxicity, apoptosis, caspase 3 activation, transcriptional activation of E2F3a, a miR-128 target gene, as well as E2F3a protein expression. mir-128 33-40 Rh associated glycoprotein Homo sapiens 106-109 21372826-11 2011 In U251 cells, transfection of a miR-128 inhibitor (50 nmol/L) prevented the overexpression of miR-128 by Rh2, and significantly blunted Rh2-induced cytotoxicity, apoptosis, caspase 3 activation, transcriptional activation of E2F3a, a miR-128 target gene, as well as E2F3a protein expression. mir-128 33-40 Rh associated glycoprotein Homo sapiens 137-140 21372826-11 2011 In U251 cells, transfection of a miR-128 inhibitor (50 nmol/L) prevented the overexpression of miR-128 by Rh2, and significantly blunted Rh2-induced cytotoxicity, apoptosis, caspase 3 activation, transcriptional activation of E2F3a, a miR-128 target gene, as well as E2F3a protein expression. mir-128 33-40 caspase 3 Homo sapiens 174-183 21372826-11 2011 In U251 cells, transfection of a miR-128 inhibitor (50 nmol/L) prevented the overexpression of miR-128 by Rh2, and significantly blunted Rh2-induced cytotoxicity, apoptosis, caspase 3 activation, transcriptional activation of E2F3a, a miR-128 target gene, as well as E2F3a protein expression. mir-128 95-102 Rh associated glycoprotein Homo sapiens 106-109 19713529-0 2009 MiR-128 up-regulation inhibits Reelin and DCX expression and reduces neuroblastoma cell motility and invasiveness. mir-128 0-7 reelin Homo sapiens 31-37 19713529-0 2009 MiR-128 up-regulation inhibits Reelin and DCX expression and reduces neuroblastoma cell motility and invasiveness. mir-128 0-7 doublecortin Homo sapiens 42-45 19713529-3 2009 We studied miR-128, a brain-enriched microRNA, in retinoic acid-differentiated neuroblastoma cells, and we found that this microRNA is up-regulated in treated cells, where it down-modulates the expression of two proteins involved in the migratory potential of neural cells: Reelin and DCX. mir-128 11-18 reelin Homo sapiens 274-280 19713529-3 2009 We studied miR-128, a brain-enriched microRNA, in retinoic acid-differentiated neuroblastoma cells, and we found that this microRNA is up-regulated in treated cells, where it down-modulates the expression of two proteins involved in the migratory potential of neural cells: Reelin and DCX. mir-128 11-18 doublecortin Homo sapiens 285-288 19713529-4 2009 Consistently, miR-128 ectopic overexpression suppressed Reelin and DCX, whereas the LNA antisense-mediated miR-128 knockdown caused the two proteins to increase. mir-128 14-21 reelin Homo sapiens 56-62 19713529-4 2009 Consistently, miR-128 ectopic overexpression suppressed Reelin and DCX, whereas the LNA antisense-mediated miR-128 knockdown caused the two proteins to increase. mir-128 14-21 doublecortin Homo sapiens 67-70 18381601-6 2008 In this study, we show that Tat deregulates expression levels of selected microRNAs, including the neuronal mir-128, in primary cortical neurons. mir-128 108-115 tyrosine aminotransferase Homo sapiens 28-31