Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Medicine (Baltimore) ; 103(4): e36799, 2024 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-38277535

RESUMO

Pyroptosis plays a key role in the death of cells including cardiomyocytes, and it is associated with a variety of cardiovascular diseases. However, the role of pyroptosis-related genes (PRGs) in hypertrophic cardiomyopathy (HCM) is not well characterized. This study aimed to identify key biomarkers and explore the molecular mechanisms underlying the functions of the PRGs in HCM. The differentially expressed genes were identified by GEO2R, and the differentially expressed pyroptosis-related genes (DEPRGs) of HCM were identified by combining with PRGs. Enrichment analysis was performed using the "clusterProfiler" package of the R software. Protein-protein interactions (PPI) network analysis was performed using the STRING database, and hub genes were screened using cytoHubba. TF-miRNA coregulatory networks and protein-chemical interactions were analyzed using NetworkAnalyst. RT-PCR/WB was used for expression validation of HCM diagnostic markers. Quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western Blot (WB) were used to measure and compare the expression of the identified genes in the cardiac hypertrophy model and the control group. A total of 20 DEPRGs were identified, which primarily showed enrichment for the positive regulation of cytokine production, regulation of response to biotic stimulus, tumor necrosis factor production, and other biological processes. These processes primarily involved pathways related to Renin-angiotensin system, Adipocytokine signaling pathway and NF-kappa B signaling pathway. Then, a PPI network was constructed, and 8 hub genes were identified. After verification analysis, the finally identified HCM-related diagnostic markers were upregulated gene protein tyrosine phosphatase non-receptor type 11 (PTPN11), downregulated genes interleukin-1 receptor-associated kinase 3 (IRAK3), and annexin A2 (ANXA2). Further GSEA analysis revealed these 3 biomarkers primarily related to cardiac muscle contraction, hypertrophic cardiomyopathy, fatty acid degradation and ECM - receptor interaction. Moreover, we also elucidated the interaction network of these biomarkers with the miRNA network and known compounds, respectively. RT-PCR/WB results indicated that PTPN11 expression was significantly increased, and IRAK3 and ANXA2 expressions were significantly decreased in HCM. This study identified PTPN11, IRAK3, and ANXA2 as pyroptosis-associated biomarkers of HCM, with the potential to reveal the development and pathogenesis of HCM and could be potential therapeutic targets.


Assuntos
Cardiomiopatia Hipertrófica , MicroRNAs , Humanos , Redes Reguladoras de Genes , Perfilação da Expressão Gênica/métodos , Piroptose/genética , Biomarcadores , MicroRNAs/genética , MicroRNAs/metabolismo , Cardiomiopatia Hipertrófica/diagnóstico , Cardiomiopatia Hipertrófica/genética , Biologia Computacional/métodos
2.
Int Urol Nephrol ; 53(11): 2321-2332, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33895976

RESUMO

PURPOSE: Hyperuricemia (HUA) and hypertriglyceridemia (HTG) were very common in chronic kidney disease (CKD) and associated with accelerated progression of CKD. This was a retrospective, cross-sectional study which aimed to explore the relationship between serum uric acid levels or triglyceride levels and tubular atrophy/interstitial fibrosis (proven by renal biopsy). METHODS: The present study enrolled 229 CKD individuals who included 127 biopsy-proven primary IgA nephrology (IgAN) patients and 102 biopsy-proven primary membranous nephropathy (MN) patients. The baseline characteristics at the time of the kidney biopsy were collected. According to the serum uric acid (UA) or triglyceride (TG) whether it exceeds the normal reference range, patients were divided into non-HUA (n = 127), HUA (n = 102), non-HTG (n = 119), and HTG group (n = 110). Based on the extent of tubular atrophy/interstitial fibrosis, patients were divided into no/mild injury (T0, n = 127), moderate injury (T1, n = 102). Multivariable logistic regression for factors predicting moderate tubular atrophy/interstitial fibrosis was performed. RESULTS: There were 127 IgAN and 102 MN cases among 229 patients in the present study. The prevalence of HUA was 44.5% (n = 102), 40.9% (n = 52), and 49.0% (n = 50) in all patients, IgAN patients and MN patients, respectively (P = 1.49). The prevalence of HTG was 48.0% (n = 110), 29.9% (n = 38), and 70.6% (n = 72) (P < 0.001), respectively, as well. Multivariate logistic regression analysis showed that HUA and HTG were independent risk factors for moderate tubular atrophy/interstitial fibrosis (HUA OR = 2.335, 95% CI = 1.147-4.755, P = 0.019; HTG OR = 2.646, 95% CI = 1.289-5.432, P = 0.008). The area under curve (AUC) of model 1 (HUA + eGFR + HTG + age + serum globulin + 24 h urinary protein, AUC = 0.876) was larger than the other two models; however, there was no significant difference among these models (all P > 0.05). CONCLUSIONS: Hyperuricemia and hypertriglyceridemia, which were prevalent in CKD patients, were the independent risk factors for moderate tubular atrophy/interstitial fibrosis. HUA together with HTG could improve the value of diagnosis for moderate tubular atrophy/interstitial fibrosis to some extent.


Assuntos
Glomerulonefrite por IGA/complicações , Glomerulonefrite Membranosa/complicações , Hipertrigliceridemia/complicações , Hiperuricemia/complicações , Rim/patologia , Adulto , Atrofia/etiologia , Estudos Transversais , Feminino , Fibrose/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
3.
Theranostics ; 10(6): 2522-2537, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32194817

RESUMO

Objective: The current antiangiogenic therapy for atherosclerotic plaques was mainly achieved by the use of antiangiogenic drugs, but serious side effects have limited the clinical application. The present study investigated whether therapeutic ultrasound (TUS) treatment with appropriate pressure could selectively deplete the neovasculature in vulnerable plaques to improve its stability with no side effects on the body; the underlying mechanisms were also explored. Methods and Results: A mouse model of advanced atherosclerosis was generated by maintaining apolipoprotein E-deficient (ApoE-/-) mice on a hypercholesterolemic diet (HCD). Plaque, skeletal muscle, mesentery and skin tissue from 114 atheroma-bearing mice were subjected to sham therapy, an ultrasound application combined with microbubbles at four different ultrasound pressures (1.0, 2.0, 3.0, 5.0 MPa), or ultrasound at 5.0 MPa alone. Microvessel density (MVD) was assessed by immunofluorescence and immunohistochemical methods. The plaque necrotic center/fiber cap (NC/FC) ratio and vulnerability index were calculated to evaluate plaque vulnerability. Twenty-four hours after TUS treatment at 3.0 MPa, the MVD in the plaque was substantially decreased by 84% (p < 0.05), while there was almost no change in MVD and neovessel density (NVD) in normal tissues, including skeletal muscle, mesentery and skin. Additionally, a marked reduction in the number of immature vessels was observed in the plaques (reduced by 90%, p < 0.05), whereas the number of mature vessels was not significantly decreased. Furthermore, TUS treatment at 3.0 MPa significantly improved plaque stability, as reflected by the NC/FC ratio and vulnerability index, which may be due to the selective destruction of intraplaque neovascularization by TUS treatment, thereby decreasing the extravasation of erythrocytes and leading to vascular inflammation alleviation and thin-cap fibroatheroma reduction. Conclusions: TUS treatment at 3.0 MPa selectively depleted plaque neovessels and improved the stability of vulnerable plaques through a reduction in erythrocyte extravasation and inflammatory mediator influx, with no significant effect on normal tissue.


Assuntos
Aterosclerose/terapia , Microbolhas/uso terapêutico , Neovascularização Patológica/terapia , Placa Aterosclerótica/terapia , Terapia por Ultrassom/métodos , Inibidores da Angiogênese/uso terapêutico , Animais , Aterosclerose/patologia , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica/métodos , Masculino , Camundongos , Camundongos Knockout para ApoE , Microscopia de Fluorescência/métodos , Densidade Microvascular/efeitos dos fármacos , Neovascularização Patológica/patologia , Placa Aterosclerótica/patologia , Ultrassonografia/métodos
4.
Theranostics ; 9(19): 5558-5576, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31534503

RESUMO

Objective: Long noncoding RNAs (lncRNAs) may serve as specific targets for the treatment of abdominal aortic aneurysms (AAAs). LncRNA GAS5, functionally associated with smooth muscle cell (SMC) apoptosis and proliferation, is likely involved in AAA formation, but the exact role of GAS5 in AAA is unknown. We thus explored the contribution of GAS5 to SMC-regulated AAA formation and its underlying mechanisms. Methods: Human specimens were used to verify the diverse expression of GAS5 in normal and AAA tissues. The angiotensin II (Ang II)-induced AAA model in ApoE-/- mice and the CaCl2-induced AAA model in wild-type C57BL/6 mice were used. RNA pull-down and luciferase reporter gene assays were performed in human aortic SMCs to detect the interaction between GAS5 and its downstream targets of protein or microRNA (miR). Results: GAS5 expression was significantly upregulated in human AAA specimens and two murine AAA models compared to human normal aortas and murine sham-operated controls. GAS5 overexpression induced SMC apoptosis and repressed its proliferation, thereby promoting AAA formation in two murine AAA models. Y-box-binding protein 1 (YBX1) was identified as a direct target of GAS5 while it also formed a positive feedback loop with GAS5 to regulate the downstream target p21. Furthermore, GAS5 acted as a miR-21 sponge to release phosphatase and tensin homolog from repression, which blocked the activation and phosphorylation of Akt to inhibit proliferation and promote apoptosis in SMCs. Conclusion: The LncRNA GAS5 contributes to SMC survival during AAA formation. Thus, GAS5 might serve as a novel target against AAA.


Assuntos
Aneurisma da Aorta Abdominal/metabolismo , Aneurisma da Aorta Abdominal/parasitologia , Músculo Liso Vascular/citologia , RNA Longo não Codificante/metabolismo , Idoso , Animais , Aorta/metabolismo , Aneurisma da Aorta Abdominal/genética , Apolipoproteínas E/genética , Apoptose , Feminino , Humanos , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/citologia , Miócitos de Músculo Liso/metabolismo , RNA Longo não Codificante/genética
5.
Clin Sci (Lond) ; 133(13): 1439-1455, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31235554

RESUMO

The long non-coding RNA (lncRNA) PTENP1 is a pseudogene of phosphatase and tensin homologue deleted on chromosome ten (PTEN), has been implicated in smooth muscle cell (SMC) proliferation and apoptosis. PTENP1 is the pseudogene of PTEN. However, it is unclear whether and how PTENP1 functions in the proliferation and apoptosis of human aortic SMCs (HASMCs). Here, we hypothesised that PTENP1 inhibits HASMC proliferation and enhances apoptosis by promoting PTEN expression. PCR analysis and Western blot assays respectively showed that both PTENP1 and PTEN were up-regulated in human aortic dissection (AD) samples. PTENP1 overexpression significantly increased the protein expression of PTEN, promoted apoptosis and inhibited the proliferation of HASMCs. PTENP1 silencing exhibited the opposite effects and mitigated H2O2-induced apoptosis of HASMCs. In an angiotensin II (Ang II)-induced mouse aortic aneurysm (AA) model, PTENP1 overexpression potentiated aortic SMC apoptosis, exacerbated aneurysm formation. Mechanistically, RNA pull-down assay and a series of luciferase reporter assays using miR-21 mimics or inhibitors identified PTENP1 as a molecular sponge for miR-21 to endogenously compete for the binding between miR-21 and the PTEN transcript, releasing PTEN expression. This finding was further supported by in vitro immunofluorescent evidence showing decreased cell apoptosis upon miR-21 mimic administration under baseline PTENP1 overexpression. Ex vivo rescue of PTEN significantly mitigated the SMC apoptosis induced by PTENP1 overexpression. Finally, Western blot assays showed substantially reduced Akt phosphorylation and cyclin D1 and cyclin E levels with up-regulated PTENP1 in HASMCs. Our study identified PTENP1 as a mediator of HASMC homeostasis and suggests that PTENP1 is a potential target in AD or AA intervention.


Assuntos
Aneurisma Aórtico/metabolismo , Dissecção Aórtica/metabolismo , Apoptose , Proliferação de Células , Músculo Liso Vascular/metabolismo , Miócitos de Músculo Liso/metabolismo , Pseudogenes , RNA Longo não Codificante/metabolismo , Dissecção Aórtica/genética , Dissecção Aórtica/patologia , Animais , Aorta/metabolismo , Aorta/patologia , Aneurisma Aórtico/genética , Aneurisma Aórtico/patologia , Ciclo Celular , Células Cultivadas , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Liso Vascular/patologia , Miócitos de Músculo Liso/patologia , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais
6.
Circulation ; 139(25): 2857-2876, 2019 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-30947518

RESUMO

BACKGROUND: circRNAs (circular RNAs) are emerging as powerful regulators of cardiac development and disease, but their roles in cardiac regeneration are still unknown. This study used superenhancers to distinguish key circRNAs in the regulation of cardiac regeneration and explored the mechanisms underlying circRNA functions. METHODS: We used integrated bioinformatics analysis of RNA sequencing data and superenhancer catalogs to identify superenhancer-associated circRNAs. Quantitative polymerase chain reactions and in situ hybridization were performed to determine the circRNA expression patterns in hearts. Gain- and loss-of-function assays were conducted to detect the role of circRNAs in cardiomyocyte proliferation and cardiac repair after myocardial infarction. Chromatin immunoprecipitation (ChIP) and electrophoretic mobility shift assays were used to determine the binding of Meis1 (Meis homeobox 1) on circNfix-associated superenhancers. RNA pulldown and luciferase reporter assays were used to study circRNA interactions with proteins and miRNAs (micro RNAs). RESULTS: We identified a circRNA, Nfix circRNA (circNfix), that was regulated by a superenhancer and overexpressed in the adult heart in humans, rats, and mice. The transcription factor Meis1 bound to the superenhancer at the circNfix locus, and increased its expression. In vitro and in vivo, cardiomyocyte proliferation was increased by knockdown of circNfix, whereas it was inhibited by circNfix overexpression. Moreover, circNfix downregulation promoted cardiomyocyte proliferation and angiogenesis and inhibited cardiomyocyte apoptosis after myocardial infarction, attenuating cardiac dysfunction and improving the prognosis. Mechanistically, circNfix reinforced the interaction of Ybx1 (Y-box binding protein 1) with Nedd4l (an E3 ubiquitin ligase), and induced Ybx1 degradation through ubiquitination, repressing cyclin A2 and cyclin B1 expression. In addition, circNfix acted as a sponge for miR-214 to promote Gsk3ß (glycogen synthase kinase 3 ß) expression and repress ß-catenin activity. CONCLUSIONS: Loss of superenhancer-regulated circNfix promotes cardiac regenerative repair and functional recovery after myocardial infarction by suppressing Ybx1 ubiquitin-dependent degradation and increasing miR-214 activity and thus may be a promising strategy for improving the prognosis after MI.


Assuntos
Proliferação de Células , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , RNA Circular/metabolismo , Regeneração , Animais , Apoptose , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Humanos , Camundongos Endogâmicos C57BL , MicroRNAs/genética , MicroRNAs/metabolismo , Proteína Meis1/genética , Proteína Meis1/metabolismo , Infarto do Miocárdio/genética , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/patologia , Ubiquitina-Proteína Ligases Nedd4/genética , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Neovascularização Fisiológica , RNA Circular/genética , Ratos Sprague-Dawley , Transdução de Sinais , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , beta Catenina/genética , beta Catenina/metabolismo
7.
Arterioscler Thromb Vasc Biol ; 39(1): e10-e25, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30580562

RESUMO

Objective- Vascular smooth muscle cell phenotypic transition plays a critical role in the formation of abdominal aortic aneurysms (AAAs). SM22α (smooth muscle 22α) has a vital role in maintaining the smooth muscle cell phenotype and is downregulated in AAA. However, whether manipulation of the SM22α gene influences the pathogenesis of AAA is unclear. Here, we investigated whether SM22α prevents AAA formation and explored the underlying mechanisms. Approach and Results- In both human and animal AAA tissues, a smooth muscle cell phenotypic switch was confirmed, as manifested by the downregulation of SM22α and α-SMA (α-smooth muscle actin) proteins. The methylation level of the SM22α gene promoter was dramatically higher in mouse AAA tissues than in control tissues. SM22α knockdown in ApoE-/- (apolipoprotein E-deficient) mice treated with Ang II (angiotensin II) accelerated the formation of AAAs, as evidenced by a larger maximal aortic diameter and more medial elastin degradation than those found in control mice, whereas SM22α overexpression exerted opposite effects. Similar results were obtained in a calcium chloride-induced mouse AAA model. Mechanistically, SM22α deficiency significantly increased reactive oxygen species production and NF-κB (nuclear factor-κB) activation in AAA tissues, whereas SM22α overexpression produced opposite effects. NF-κB antagonist SN50 or antioxidant N-acetyl-L-cysteine partially abrogated the exacerbating effects of SM22α silencing on AAA formation. Conclusions- SM22α reduction in AAAs because of the SM22α promoter hypermethylation accelerates AAA formation through the reactive oxygen species/NF-κB pathway, and therapeutic approaches to increase SM22α expression are potentially beneficial for preventing AAA formation.


Assuntos
Aneurisma da Aorta Abdominal/prevenção & controle , Proteínas dos Microfilamentos/fisiologia , Proteínas Musculares/fisiologia , Músculo Liso Vascular/fisiologia , Miócitos de Músculo Liso/fisiologia , NF-kappa B/fisiologia , Espécies Reativas de Oxigênio/metabolismo , Animais , Células Cultivadas , Metilação de DNA , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculo Liso Vascular/citologia , NADPH Oxidases/metabolismo , Fenótipo , Fosforilação , Regiões Promotoras Genéticas
8.
EBioMedicine ; 39: 69-82, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30545799

RESUMO

BACKGROUND: We previously found that loss of lncRNA-AZIN2 splice variant (AZIN2-sv) increases cardiomyocyte (CM) proliferation and attenuates adverse ventricular remodelling post-myocardial infarction (MI). However, whether inhibition of AZIN2-sv can simultaneously induce angiogenesis and thus improve prognosis after MI is unclear. METHODS: We used in situ hybridization and quantitative PCR to determine AZIN2-sv expression in endothelial cells. Knockdown and overexpression were performed to detect the role of AZIN2-sv in endothelial cell function, angiogenesis and prognosis after MI. RNA pulldown, RNA immunoprecipitation and luciferase reporter assays were used to determine the interaction with talin1 (Tln1) protein and miRNA-214 (miR-214). DNA pulldown and chromatin immunoprecipitation (ChIP) assays were used to study AZIN2-sv binding to upstream transcription factors. FINDINGS: AZIN2-sv was enriched in cardiac endothelial cells. The loss of AZIN2-sv reduced endothelial cell apoptosis and promoted endothelial sprouting and capillary network formation in vitro. Moreover, in vivo, the loss of AZIN2-sv induced angiogenesis and improved cardiac function after MI. Mechanistically, AZIN2-sv reduced Tln1 and integrin ß1 (ITGB1) protein levels to inhibit neovascularization. AZIN2-sv activated the ubiquitination-dependent degradation of Tln1 mediated by proteasome 26S subunit ATPase 5 (PSMC5). In addition, AZIN2-sv could bind to miR-214 and suppress the phosphatase and tensin homologue (PTEN)/Akt pathway to inhibit angiogenesis. With regard to the upstream mechanism, Bach1, a negative regulator of angiogenesis, bound to the promoter of AZIN2-sv and increased its expression. INTERPRETATION: Bach1-activated AZIN2-sv could participate in angiogenesis by promoting the PSMC5-mediated ubiquitination-dependent degradation of Tln1 and blocking the miR-214/PTEN/Akt pathway. Inhibition of AZIN2-sv induced angiogenesis and myocardial regeneration simultaneously, thus, AZIN2-sv could be an ideal therapeutic target for improving myocardial repair after MI. FUND: National Natural Science Foundations of China.


Assuntos
ATPases Associadas a Diversas Atividades Celulares/metabolismo , MicroRNAs/genética , Infarto do Miocárdio/genética , Neovascularização Fisiológica , Complexo de Endopeptidases do Proteassoma/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais , Talina/genética , Processamento Alternativo , Animais , Apoptose , Fatores de Transcrição de Zíper de Leucina Básica/metabolismo , Movimento Celular , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Testes de Função Cardíaca , Células Endoteliais da Veia Umbilical Humana , Humanos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/citologia , Miócitos Cardíacos/metabolismo , PTEN Fosfo-Hidrolase/metabolismo , Prognóstico , Proteólise , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Talina/metabolismo
9.
J Exp Clin Cancer Res ; 37(1): 232, 2018 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-30231924

RESUMO

BACKGROUND: Multidrug resistance (MDR) is often responsible for the failure of chemotherapy treatment, and current strategies for cancer MDR are not adequately satisfying as to their efficacy and safety. In this study, we sought to determine the anti-MDR effects of ultrasound (US) irradiation and its underlying mechanisms against drug-resistance. METHODS: MDR variant MCF-7/ADR cell lines and endothelial cell lines were used to determine the appropriate ultrasound intensity for in vitro experiments. MCF-7/ADR cell and HEPG2/ADM cells were used to assess the anti-MDR effect of US irradiation. Intracellular adriamycin (ADM) accumulation, Cell viability, cell proliferation and cell apoptosis were evaluated after ADM + US treatment or ADM treatment alone. MCF-7/ADR xenograft mice were used to investigate the appropriate ultrasound intensity for in vivo experiments and its effect on the long-term prognosis. Underlining mechanisms by which ultrasound exposure reversing MDR phenotype were investigated both in vitro and in vivo. RESULTS: Combination of ADM and 0.74 W/cm2 US irradiation enhanced ADM intracellular concentration and nuclear accumulation in MCF-7/ADR and HEPG2/ADM cells, compared to those treated with ADM alone. Enhanced cellular ADM uptake and nuclei localization was associated with increased cytotoxicity of ADM to ADM-resistant cells, lower ADM-resistant cell viability and proliferative cell ratio, and higher apoptotic cell ratio. More importantly, US exposure increased the effectiveness of ADM to inhibit tumor growth in MCF-7/ADR xenograft mice. Mechanistically, US exposure promoted ADM accumulation in MDR cells mainly through down-regulation of P-glycoprotein (P-gp), which is dependent on US-induced intracellular reactive oxygen species (ROS) production. US-induced oxidative stress promoted miR-200c-3p and miR-34a-3p expression by forming miR-200c/34a/ZEB1 double-negative feedback loop. Finally, US-induced miR-200c/34a overexpression decreased P-gp expression and reversed MDR phenotype. CONCLUSION: US irradiation could reverse MDR phenotype by activating ROS-ZEB1-miR200c/34a-P-gp signal pathway. Our findings offer a new and promising strategy for sensitizing cells to combat MDR and to improve the therapeutic index of chemotherapy.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/genética , MicroRNAs/genética , Homeobox 1 de Ligação a E-box em Dedo de Zinco/genética , Animais , Apoptose/genética , Apoptose/efeitos da radiação , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/efeitos da radiação , Sobrevivência Celular/genética , Sobrevivência Celular/efeitos da radiação , Doxorrubicina/administração & dosagem , Resistência a Múltiplos Medicamentos/genética , Resistencia a Medicamentos Antineoplásicos/efeitos da radiação , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos da radiação , Células Hep G2 , Humanos , Células MCF-7 , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , Estresse Oxidativo/efeitos da radiação , Espécies Reativas de Oxigênio/efeitos da radiação , Ondas Ultrassônicas/efeitos adversos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Int J Clin Exp Pathol ; 8(10): 12368-78, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26722423

RESUMO

BACKGROUNDS: Acute ischemia reperfusion-induced kidney injury is a common cause of acute renal failure, and it is also an important cause of delayed recovery of transplanted kidney functions and even loss of function. However, there is no effective treatment method in clinical applications presently. OBJECTIVE: The objective was to investigate effects of transforming growth factor-ß1 on homing of bone marrow mesenchymal stem cells in renal ischemia-reperfusion injury. METHODS: Effects of TGF-ß1 over-expression in MSCs on expression of CXCR4 and chemotactic effect to SDF-1 were investigated by in vitro transmembrane chemotaxis. Anti-TGF-ß1 antibody was incubated with ischemia reperfusion injury renal tissue homogenate and effects of anti-TGF-ß1 antibody were observed. In addition, effects of TGF-ß1 gene transfection and anti-CXCR4 antibody treatment in MSCs on expression of SDF-1/CXCR4 axis of renal tissues and damage repair were further explored. RESULTS: Expression of TGF-ß1 mRNA in the IRI group increased significantly, and MSCs transplantation could enhance expression of CXCR4 mRNA in rats of the IRI group, the expression of CXCR4 can be decreased by the anti-TGF-ß1 antibody and the anti-CXCR4 antibody. TGF-ß1 induced homing of MSCs in repair of renal ischemic reperfusion injury by regulating expression of CXCR4 on cell membranes. Blue fluorescence of DAPI-positive MSCs cells of renal parenchyma in the IRI+MSC group was enhanced significantly, which was significantly inhibited by anti-TGF-ß1 and anti-CXCR4 antibody, and the inhibitory effect of anti-CXCR4 antibody was more obvious than that of anti-TGF-ß1 antibody. CONCLUSION: Transforming growth factor-ß1 promotes homing of bone marrow mesenchymal stem cells in renal ischemia-reperfusion injury, which will provide useful data on role of TGF-ß1 in regulating SDF-1/CXCR4 axis-induced MSCs homing.


Assuntos
Injúria Renal Aguda/metabolismo , Movimento Celular/fisiologia , Transplante de Células-Tronco Mesenquimais/métodos , Traumatismo por Reperfusão/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Transplante de Medula Óssea/métodos , Modelos Animais de Doenças , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Receptores CXCR4/biossíntese , Transfecção
11.
Mol Med Rep ; 9(5): 1794-8, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24573381

RESUMO

Acute renal failure is one of the most common complications observed in hospitals. Although extensive studies have been carried out to search for therapeutic treatments, no effective cure has been established. In recent years, stem cell therapy for tissue engineering and repair has become a key area of study. Bone marrow mesenchymal stem cells (MSCs) have been demonstrated to exhibit a reparative role in ischemia reperfusion-injured kidney tissue, and the stromal cell-derived factor-1 (SDF-1)/chemokine (C-X-C motif) receptor 4 (CXCR4) axis has been found to play an important role in the migration and homing of MSCs. In the present study, transforming growth factor-ß1 (TGF-ß1) in the homogenate of the acute ischemia reperfusion-injured renal tissue was found to markedly increase the CXCR4 surface expression of MSCs, which contributes to the migration of MSCs to SDF-1. Neutralization of TGF-ß1 inhibited the migration in an antibody concentration-dependent manner, through downregulation of CXCR4 localized to the membrane. These observations suggest a potential mechanism for MSC migration to the kidney which may provide a possible therapeutic target for curing acute renal failure.


Assuntos
Injúria Renal Aguda/metabolismo , Microambiente Celular , Quimiocina CXCL12/metabolismo , Células-Tronco Mesenquimais/metabolismo , Receptores CXCR/metabolismo , Traumatismo por Reperfusão/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Injúria Renal Aguda/genética , Animais , Movimento Celular/genética , Células Cultivadas , Modelos Animais de Doenças , Feminino , Expressão Gênica , Imunofenotipagem , Células-Tronco Mesenquimais/efeitos dos fármacos , Fenótipo , Ratos , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Traumatismo por Reperfusão/genética , Fator de Crescimento Transformador beta1/farmacologia
12.
Mol Biol Rep ; 37(4): 1749-54, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19578979

RESUMO

Proteinuria is a well-established exacerbating factor in chronic kidney disease. Although the mechanisms of albumin-induced tubulointerstitial damage have been extensively studied, the influence of mycophenolic acid (MPA) on tubular epithelial cells has not been sufficiently elucidated. MPA, the active metabolite of mycophenolate mofetil, is a potent, non-competitive, and reversible inhibitor of inosine-5'-monophosphate dehydrogenase, the rate-limiting enzyme for de novo purine synthesis. Monocyte chemoattractant protein 1 (MCP-1) is a 76-amino-acid chemokine thought to be the major chemotactic factor for monocytes. MCP-1 is found in macrophage-rich areas of atherosclerotic lesions. However, the mechanisms regulating MCP-1 expression by MPA in renal tubular epithelial cells were still unclear. In this study, the inhibitory effect of MPA on MCP-1 expression by albumin-induced renal tubular epithelial cells was investigated, and the roles of p38 mitogen-activated protein kinase (p38 MAPK) pathway were explored. MPA attenuated albumin-induced expression of MCP-1 mRNA and protein. The experiment suggested that MPA actively inhibited protein of MCP-1. The inhibitory effect of MPA on MCP-1 expression was mediated by the sequential attenuation of p38 MAPK expression. These inhibitory effects were partially inhibited by SB203580, a specific inhibitor of p38 MAPK. Taken together, these results suggest that the negative modulation of MCP-1 by MPA is partly dependent on p38 MAPK pathway.


Assuntos
Albuminas/farmacologia , Quimiocina CCL2/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/enzimologia , Túbulos Renais Proximais/citologia , Ácido Micofenólico/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Linhagem Celular , Quimiocina CCL2/genética , Ativação Enzimática/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Imidazóis/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Piridinas/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA