Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Hepatology ; 75(5): 1218-1234, 2022 05.
Article in English | MEDLINE | ID: mdl-34591986

ABSTRACT

BACKGROUND AND AIMS: NAFLD is considered as the hepatic manifestation of the metabolic syndrome, which includes insulin resistance, obesity and hyperlipidemia. NASH is a progressive stage of NAFLD with severe hepatic steatosis, hepatocyte death, inflammation, and fibrosis. Currently, no pharmacological interventions specifically tailored for NASH are approved. Ovarian tumor domain, ubiquitin aldehyde binding 1 (OTUB1), the founding member of deubiquitinases, regulates many metabolism-associated signaling pathways. However, the role of OTUB1 in NASH is unclarified. METHODS AND RESULTS: We demonstrated that mice with Otub1 deficiency exhibited aggravated high-fat diet-induced and high-fat high-cholesterol (HFHC) diet-induced hyperinsulinemia and liver steatosis. Notably, hepatocyte-specific overexpression of Otub1 markedly alleviated HFHC diet-induced hepatic steatosis, inflammatory responses, and liver fibrosis. Mechanistically, we identified apoptosis signal-regulating kinase 1 (ASK1) as a key candidate target of OTUB1 through RNA-sequencing analysis and immunoblot analysis. Through immunoprecipitation-mass spectrometry analysis, we further found that OTUB1 directly bound to tumor necrosis factor receptor-associated factor 6 (TRAF6) and suppressed its lysine 63-linked polyubiquitination, thus inhibiting the activation of ASK1 and its downstream pathway. CONCLUSIONS: OTUB1 is a key suppressor of NASH that inhibits polyubiquitinations of TRAF6 and attenuated TRAF6-mediated ASK1 activation. Targeting the OTUB1-TRAF6-ASK1 axis may be a promising therapeutic strategy for NASH.


Subject(s)
Cysteine Endopeptidases/metabolism , Non-alcoholic Fatty Liver Disease , Animals , Diet, High-Fat , Disease Models, Animal , Liver , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/drug therapy , Signal Transduction , TNF Receptor-Associated Factor 6
2.
J Cell Mol Med ; 25(16): 7867-7877, 2021 08.
Article in English | MEDLINE | ID: mdl-34190420

ABSTRACT

Cardiac hypertrophy and the resultant heart failure are among the most common causes of morbidity and mortality worldwide; thus, identifying the key factor mediating pathological cardiac hypertrophy is critically important for developing the strategy to protect against heart failure. Runx1 (Runt-related transcription factor 1) acts as an essential transcription factor that functions in a variety of cellular processes including differentiation, proliferation, tissue growth and DNA damage response. However, relatively little is known about the role of Runx1 in heart, especially cardiac hypertrophy and heart failure. In the present study, we investigated the role of Runx1 in experimentally pathological cardiac hypertrophy. The in vitro model was induced by Ang II exposure to cultured neonatal rat cardiomyocytes, and the in vivo pathological cardiac hypertrophy models were induced by chronic pressure overload in mice. Runx1 expression is increased in heart tissues from mice with pressure overload-induced cardiac hypertrophy and in neonatal rat cardiomyocytes in response to Ang II stimulation. Moreover, knockdown of cardiac Runx1 alleviates the pressure overload-induced cardiac hypertrophy. Mechanistically, Runx1 activates the p53 signalling by binding to the p53 gene and promotes its transcription. Rescue experiments indicate that Runx1 promotes cardiac hypertrophy in a p53-dependent manner. Remarkably, we demonstrated that Ro5-3335 (a Runx1 inhibitor) acts as a potential therapeutic drug for treating pathological cardiac hypertrophy. In summary, we conclude that Runx1 is a novel mediator and therapeutic target for pathological cardiac hypertrophy.


Subject(s)
Cardiomegaly/pathology , Core Binding Factor Alpha 2 Subunit/metabolism , Gene Expression Regulation , Gene Knockdown Techniques/methods , Myocytes, Cardiac/pathology , Tumor Suppressor Protein p53/metabolism , Animals , Cardiomegaly/genetics , Cardiomegaly/metabolism , Cells, Cultured , Core Binding Factor Alpha 2 Subunit/antagonists & inhibitors , Core Binding Factor Alpha 2 Subunit/genetics , Disease Models, Animal , Mice , Mice, Inbred BALB C , Myocytes, Cardiac/metabolism , Rats , Signal Transduction , Tumor Suppressor Protein p53/genetics
3.
Hepatology ; 70(4): 1099-1118, 2019 10.
Article in English | MEDLINE | ID: mdl-30820969

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) has become the most common cause of chronic liver disease worldwide. Due to the growing economic burden of NAFLD on public health, it has become an emergent target for clinical intervention. DUSP12 is a member of the dual specificity phosphatase (DUSP) family, which plays important roles in brown adipocyte differentiation, microbial infection, and cardiac hypertrophy. However, the role of DUSP12 in NAFLD has yet to be clarified. Here, we reveal that DUSP12 protects against hepatic steatosis and inflammation in L02 cells after palmitic acid/oleic acid treatment. We demonstrate that hepatocyte specific DUSP12-deficient mice exhibit high-fat diet (HFD)-induced and high-fat high-cholesterol diet-induced hyperinsulinemia and liver steatosis and decreased insulin sensitivity. Consistently, DUSP12 overexpression in hepatocyte could reduce HFD-induced hepatic steatosis, insulin resistance, and inflammation. At the molecular level, steatosis in the absence of DUSP12 was characterized by elevated apoptosis signal-regulating kinase 1 (ASK1), which mediates the mitogen-activated protein kinase (MAPK) pathway and hepatic metabolism. DUSP12 physically binds to ASK1, promotes its dephosphorylation, and inhibits its action on ASK1-related proteins, JUN N-terminal kinase, and p38 MAPK in order to inhibit lipogenesis under high-fat conditions. Conclusion: DUSP12 acts as a positive regulator in hepatic steatosis and offers potential therapeutic opportunities for NAFLD.


Subject(s)
Apoptosis/genetics , Dual-Specificity Phosphatases/genetics , Gene Expression Regulation , MAP Kinase Kinase Kinase 5/genetics , Non-alcoholic Fatty Liver Disease/genetics , Analysis of Variance , Animals , Cells, Cultured , Diet, High-Fat , Disease Models, Animal , Down-Regulation , Humans , Insulin Resistance/genetics , Lipid Metabolism/genetics , Lipogenesis/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Non-alcoholic Fatty Liver Disease/physiopathology , Random Allocation , Reference Values , Signal Transduction/genetics
4.
Water Sci Technol ; 82(3): 440-453, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32960790

ABSTRACT

A polyamine functionalized polystyrene resin (PSATA) was prepared via condensation reaction of acetylated polystyrene resin with triethylenetetramine, which, upon NaBH4 reduction, produced PSATAR. In comparison with the PSATA, the PSATAR with more flexible amine groups shows improved structural properties, and the equilibrium adsorption capacities of phenol, 2-nitrophenol (ONP) and 2,4-dinitrophenol (DNP) in wastewater were up to 1.073, 1.832 and 1.901 mmol/g, respectively. Their adsorption isotherms fit well with the Freundlich model, indicating a multilayer, heterogeneous adsorption nature. Kinetic studies indicated that the adsorption of phenolic compounds conforms to the pseudo-second-order kinetics with the adsorption rate controlled by film diffusion for ONP and DNP, and intra-particle diffusion in the later stage for phenol.


Subject(s)
Water Pollutants, Chemical/analysis , Water Purification , Adsorption , Kinetics , Phenols , Polystyrenes , Trientine , Wastewater
5.
Exp Cell Res ; 370(1): 78-86, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29902536

ABSTRACT

Metabolic dysfunction is a hallmark of cardiac hypertrophy and heart failure. During cardiac failure, the metabolism of cardiomyocyte switches from fatty acid oxidation to glycolysis. However, the roles of key metabolic enzymes in cardiac hypertrophy are not understood fully. Here in the present work, we identified Aldolase A (AldoA) as a core regulator of cardiac hypertrophy. The mRNA and protein levels of AldoA were significantly up-regulated in transverse aortic constriction (TAC)- and isoproterenol (ISO)-induced hypertrophic mouse hearts. Overexpression of AldoA in cardiomyocytes promoted ISO-induced cardiomyocyte hypertrophy, whereas AldoA knockdown repressed cardiomyocyte hypertrophy. In addition, adeno-associated virus 9 (AAV9)-mediated in vivo knockdown of AldoA in the hearts rescued ISO-induced decrease in cardiac ejection fraction and fractional shortening and repressed cardiac hypertrophy. Mechanism study revealed that AldoA repressed the activation of AMP-dependent protein kinase (AMPK) signaling in a liver kinase B1 (LKB1)-dependent and AMP-independent manner. Inactivation of AMPK is a core mechanism underlying AldoA-mediated promotion of ISO-induced cardiomyocyte hypertrophy. By contrast, activation of AMPK with metformin and AICAR blocked AldoA function during cardiomyocyte hypertrophy. In summary, our data support the notion that AldoA-AMPK axis is a core regulatory signaling sensing energetic status and participates in cardiac hypertrophy.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Cardiomegaly/metabolism , Cardiomegaly/pathology , Fructose-Bisphosphate Aldolase/metabolism , Signal Transduction/physiology , Aminoimidazole Carboxamide/analogs & derivatives , Aminoimidazole Carboxamide/metabolism , Animals , Heart/physiopathology , Heart Failure/metabolism , Heart Failure/pathology , Male , Mice , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Protein Serine-Threonine Kinases/metabolism , Ribonucleotides/metabolism , Up-Regulation/physiology
6.
Mol Cell Biochem ; 429(1-2): 167-177, 2017 May.
Article in English | MEDLINE | ID: mdl-28176246

ABSTRACT

Isorhamnetin, a flavonoid compound extracted from the Chinese herb Hippophae rhamnoides L., is well known for its anti-inflammatory, anti-oxidative, anti-adipogenic, anti-proliferative, and anti-tumor activities. However, the role of isorhamnetin in cardiac hypertrophy has not been reported. The aims of the present study were to find whether isorhamnetin could alleviate cardiac hypertrophy and to define the underlying molecular mechanisms. Here, we investigated the effects of isorhamnetin (100 mg/kg/day) on cardiac hypertrophy induced by aortic banding in mice. Cardiac hypertrophy was evaluated by echocardiographic, hemodynamic, pathological, and molecular analyses. Our data demonstrated that isorhamnetin could inhibit cardiac hypertrophy and fibrosis 8 weeks after aortic banding. The results further revealed that the effect of isorhamnetin on cardiac hypertrophy was mediated by blocking the activation of phosphatidylinositol 3-kinase-AKT signaling pathway. In vitro studies performed in neonatal rat cardiomyocytes confirmed that isorhamnetin could attenuate cardiomyocyte hypertrophy induced by angiotensin II, which was associated with phosphatidylinositol 3-kinase-AKT signaling pathway. In conclusion, these data indicate for the first time that isorhamnetin has protective potential for targeting cardiac hypertrophy by blocking the phosphatidylinositol 3-kinase-AKT signaling pathway. Thus, our study suggests that isorhamnetin may represent a potential therapeutic strategy for the treatment of cardiac hypertrophy and heart failure.


Subject(s)
Cardiomegaly/drug therapy , Cardiotonic Agents/administration & dosage , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quercetin/analogs & derivatives , Angiotensin II/adverse effects , Animals , Cardiomegaly/diagnostic imaging , Cardiomegaly/metabolism , Cardiotonic Agents/pharmacology , Echocardiography , Gene Expression Regulation/drug effects , Male , Mice , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Quercetin/administration & dosage , Quercetin/pharmacology , Rats , Signal Transduction/drug effects
8.
J Am Heart Assoc ; 12(24): e029745, 2023 Dec 19.
Article in English | MEDLINE | ID: mdl-38084712

ABSTRACT

BACKGROUND: Cardiac hypertrophy (CH) is a well-established risk factor for many cardiovascular diseases and a primary cause of mortality and morbidity among older adults. Currently, no pharmacological interventions have been specifically tailored to treat CH. OTUD7B (ovarian tumor domain-containing 7B) is a member of the ovarian tumor-related protease (OTU) family that regulates many important cell signaling pathways. However, the role of OTUD7B in the development of CH is unclear. Therefore, we investigated the role of OTUD7B in CH. METHODS AND RESULTS: OTUD7B knockout mice were used to assay the role of OTUD7B in CH after transverse aortic coarctation surgery. We further assayed the specific functions of OTUD7B in isolated neonatal rat cardiomyocytes. We found that OTUD7B expression decreased in hypertrophic mice hearts and phenylephrine-stimulated neonatal rat cardiomyocytes. Furthermore, OTUD7B deficiency exacerbated transverse aortic coarctation surgery-induced myocardial hypertrophy, abnormal cardiac function, and fibrosis. In cardiac myocytes, OTUD7B knockdown promoted phenylephrine stimulation-induced myocardial hypertrophy, whereas OTUD7B overexpression had the opposite effect. An immunoprecipitation-mass spectrometry analysis showed that OTUD7B directly binds to KLF4 (Krüppel-like factor 4). Additional molecular experiments showed that OTUD7B impedes KLF4 degradation by inhibiting lysine residue at 48 site-linked ubiquitination and suppressing myocardial hypertrophy by activating the serine/threonine kinase pathway. CONCLUSIONS: These results demonstrate that the OTUD7B-KLF4 axis is a novel molecular target for CH treatment.


Subject(s)
Aortic Coarctation , Kruppel-Like Factor 4 , Mice , Rats , Animals , Cardiomegaly/genetics , Cardiomegaly/prevention & control , Cardiomegaly/metabolism , Phenylephrine/pharmacology , Phenylephrine/metabolism , Mice, Knockout , Ubiquitination , Myocytes, Cardiac/metabolism , Mice, Inbred C57BL , Endopeptidases/metabolism , Endopeptidases/pharmacology
9.
RSC Adv ; 12(44): 28637, 2022 Oct 04.
Article in English | MEDLINE | ID: mdl-36320532

ABSTRACT

[This retracts the article DOI: 10.1039/C9RA06282C.].

10.
Free Radic Biol Med ; 192: 13-24, 2022 11 01.
Article in English | MEDLINE | ID: mdl-36108935

ABSTRACT

Diabetic cardiomyopathy (DCM) is ventricular dysfunction that occurs in patients with diabetes mellitus (DM), independent of recognized risk factors, such as coronary artery disease, hypertension, and valvular heart disease. Dual-specificity phosphatase 12 (DUSP12) is a dual-specificity phosphatase expressed in all tissues. Genome-wide linkage studies have found an association between DUSP12 and type 2 diabetes (T2D). However, the role of DUSP12 in DCM remains largely unknown. Ubiquitously expressed DUSP12 is involved in nonalcoholic fatty liver disease, bacterial infection, and myocardial hypertrophy and plays a critical role in tumorigenesis. Herein, we observed an increased expression of DUSP12 in a hyperglycemia cell model and a high-fat diet (HFD) mouse model. Heart-specific DUSP12-deficient mice showed severe cardiac dysfunction and remodeling induced by an HFD. DUSP12 deficiency exacerbated oxidative stress injury and apoptosis, whereas DUSP12 overexpression had the opposite effect. At the molecular level, DUSP12 physically bound to apoptotic signal-regulated kinase 1 (ASK1), promoted its dephosphorylation, and inhibited its action on c-Jun N-terminal kinase and p38 mitogen-activated protein kinase. Rescue experiments have shown that oxidative stress injury and apoptosis, exacerbated by DUSP12 deficiency, are alleviated by ASK1 inhibition. Therefore, we consider DUSP12 an important signaling pathway in DCM.


Subject(s)
Diabetes Mellitus, Type 2 , Diabetic Cardiomyopathies , Dual-Specificity Phosphatases , Oxidative Stress , Animals , Apoptosis , Diabetes Mellitus, Type 2/complications , Diabetes Mellitus, Type 2/genetics , Diabetic Cardiomyopathies/genetics , Dual-Specificity Phosphatases/genetics , Dual-Specificity Phosphatases/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Mice , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Cell Death Dis ; 13(10): 889, 2022 10 21.
Article in English | MEDLINE | ID: mdl-36270989

ABSTRACT

Ring-finger protein 5 (RNF5) is an E3 ubiquitin ligase which is expressed in a variety of human tissues. RNF5 is involved in the regulation of endoplasmic reticulum stress, inflammation, and innate immunity and plays an important role in the occurrence and development of various tumors. However, the role of RNF5 in cardiac hypertrophy has not been reported. In this study, we found the expression of RNF5 was increased in the hearts of mice with pathological cardiac hypertrophy. The loss-of-function research demonstrated that RNF5 deficiency exacerbated cardiac hypertrophy, whereas gain-of-function studies revealed that overexpression of RNF5 had opposite effects. The stimulator of interferon genes (STING) is a signaling molecule that can activate type I interferon immunity, which can meditate inflammation and immune response in many diseases. The protein-protein interaction experiments confirmed that STING interacted with RNF5. Further studies showed that RNF5 inhibited cardiac hypertrophy by promoting STING degradation through K48-linked polyubiquitination. Therefore, we defined RNF5 as importantly regulated signaling for cardiac hypertrophy.


Subject(s)
Interferon Type I , Ubiquitin-Protein Ligases , Animals , Humans , Mice , Cardiomegaly/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Inflammation , Interferon Type I/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
12.
J Am Heart Assoc ; 11(21): e026174, 2022 11.
Article in English | MEDLINE | ID: mdl-36314496

ABSTRACT

Background Restenosis is one of the main bottlenecks in restricting the further development of cardiovascular interventional therapy. New signaling molecules involved in the progress have continuously been discovered; however, the specific molecular mechanisms remain unclear. MTMR14 (myotubularin-related protein 14) is a novel phosphoinositide phosphatase that has a variety of biological functions and is involved in diverse biological processes. However, the role of MTMR14 in vascular biology remains unclear. Herein, we addressed the role of MTMR14 in neointima formation and vascular smooth muscle cell (VSMC) proliferation after vessel injury. Methods and Results Vessel injury models were established using SMC-specific conditional MTMR14-knockout and -transgenic mice. Neointima formation was assessed by histopathological methods, and VSMC proliferation and migration were assessed using fluorescence ubiquitination-based cell cycle indicator, transwell, and scratch wound assay. Neointima formation and the expression of MTMR14 was increased after injury. MTMR14 deficiency accelerated neointima formation and promoted VSMC proliferation after injury, whereas MTMR14 overexpression remarkably attenuated this process. Mechanistically, we demonstrated that MTMR14 suppressed the activation of PLK1 (polo-like kinase 1) by interacting with it, which further leads to the inhibition of the activation of MEK/ERK/AKT (mitogen-activated protein kinase kinase/extracellular-signal-regulated kinase/protein kinase B), thereby inhibiting the proliferation of VSMC from the medial to the intima and thus preventing neointima formation. Conclusions MTMR14 prevents neointima formation and VSMC proliferation by inhibiting PLK1. Our findings reveal that MTMR14 serves as an inhibitor of VSMC proliferation and establish a link between MTMR14 and PLK1 in regulating VSMC proliferation. MTMR14 may become a novel potential therapeutic target in the treatment of restenosis.


Subject(s)
Phosphoric Monoester Hydrolases , Protein Serine-Threonine Kinases , Vascular System Injuries , Animals , Mice , Cell Movement , Cell Proliferation , Cells, Cultured , Mice, Transgenic , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Neointima/pathology , Phosphoric Monoester Hydrolases/metabolism , Vascular System Injuries/genetics , Vascular System Injuries/prevention & control , Vascular System Injuries/metabolism , Protein Serine-Threonine Kinases/metabolism , Polo-Like Kinase 1
13.
Endocrine ; 72(1): 179-189, 2021 04.
Article in English | MEDLINE | ID: mdl-32770440

ABSTRACT

PURPOSE: The study aimed to disclose mortality pattern and quantitatively evaluate risks for cause-specific mortality among thyroid cancer survivors. METHODS: We included 173,710 patients from the Surveillance, Epidemiology, and End Results (SEER) database with thyroid cancer diagnosed between 1975 and 2015. Standardized mortality ratio (SMR) was calculated using general US population as the reference. Cumulative incidence function curves were constructed to elaborate crude cause-specific mortality by histology. Cox proportional hazards regression model was adopted to identify predictors for cause-specific mortality, expressed as hazard ratio (HR) and 95% confidence interval (CI). RESULTS: After a median follow-up of 101 months, 23,040 (13.3%) deaths occurred, of which 29.1% and 21.7% were attributable to thyroid cancer and cardiovascular disease (CVD), respectively. CVD SMRs were 1.14, 1.47, 1.21, and 5.66 in patients with follicular, Hürthle cell, medullary and anaplastic histology, respectively. The adjusted HRs of thyroid cancer-specific mortality were 1.59 (95% CI: 1.46-1.74), 1.87 (95% CI: 1.65-2.12), 3.66 (95% CI: 3.31-4.05), and 12.65 (95% CI: 11.50-13.92) for follicular, Hürthle cell, medullary, and anaplastic histology, respectively, as compared with papillary histology; HRs of CVD-specific mortality were 1.23 (95% CI: 1.12-1.34), 1.27 (95% CI: 1.11-1.46), 1.13 (95% CI: 0.96-1.33), and 1.60 (95% CI: 1.19-2.16), respectively. Older age, male sex, nonwhite race, unmarried status, and advanced stage were independent predictors of CVD-specific mortality, while receiving surgery and radiotherapy were protective against CVD-specific mortality. CONCLUSIONS: We disclosed distinct mortality patterns by histology and identified predictors of CVD-specific mortality in thyroid cancer survivors, supporting CVD intervention for aggressive thyroid cancer.


Subject(s)
Cancer Survivors , Cardiovascular Diseases , Thyroid Neoplasms , Aged , Cause of Death , Humans , Male , Risk Factors
14.
Life Sci ; 259: 118251, 2020 Oct 15.
Article in English | MEDLINE | ID: mdl-32795540

ABSTRACT

AIMS: Circular RNAs (circRNAs) are relevant to atherosclerosis progression. However, the role and mechanism of circRNA hsa_circ_0029589 (circ_0029589) in atherosclerosis are not fully understood. This research aims to explore the function and mechanism of circ_0029589 in oxidized low-density lipoprotein (ox-LDL)-caused vascular smooth muscle cells (VSMCs) injury in vitro. MAIN METHODS: VSMCs were challenged via ox-LDL to mimic atherosclerosis-like injury in vitro. Circ_0029589, microRNA-214-3p (miR-214-3p) and stromal interaction molecule 1 (STIM1) abundances were detected via quantitative reverse transcription polymerase chain reaction or western blot. Cell proliferation was investigated via cell viability, cycle, apoptosis and proliferation-associated protein levels. Cell migration and invasion were assessed via transwell analysis. The relationship between miR-214-3p and circ_0029589 or STIM1 was tested via dual-luciferase reporter analysis and RNA immunoprecipitation. KEY FINDINGS: Circ_0029589 level was enhanced in ox-LDL-challenged VSMCs. Circ_0029589 interference constrained cell proliferation, migration and invasion in ox-LDL-challenged VSMCs. miR-214-3p was targeted by circ_0029589 and miR-214-3p knockdown weakened the suppressive function of circ_0029589 silence on VSMCs proliferation, migration and invasion. STIM1 was targeted via miR-214-3p and miR-214-3p could suppress VSMCs proliferation, migration and invasion via decreasing STIM1. Moreover, circ_0029589 modulated STIM1 level by miR-214-3p. SIGNIFICANCE: Circ_0029589 knockdown repressed proliferation, migration and invasion of VSMCs challenged via ox-LDL by regulating miR-214-3p and STIM1, indicating that circ_0029589 might play important role in atherosclerosis.


Subject(s)
Atherosclerosis/genetics , MicroRNAs/metabolism , Neoplasm Proteins/metabolism , RNA, Circular/metabolism , Stromal Interaction Molecule 1/metabolism , Apoptosis/physiology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cell Movement/physiology , Cell Proliferation/physiology , Cell Survival/physiology , Disease Progression , Humans , Lipoproteins, LDL/metabolism , MicroRNAs/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/physiology , Neoplasm Invasiveness , Neoplasm Proteins/genetics , RNA, Circular/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Signal Transduction , Stromal Interaction Molecule 1/genetics
15.
J Am Heart Assoc ; 9(22): e017751, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33170082

ABSTRACT

Background Cardiac hypertrophy (CH) is a physiological response that compensates for blood pressure overload. Under pathological conditions, hypertrophy can progress to heart failure as a consequence of the disorganized growth of cardiomyocytes and cardiac tissue. USP10 (ubiquitin-specific protease 10) is a member of the ubiquitin-specific protease family of cysteine proteases, which are involved in viral infection, oxidative stress, lipid drop formation, and heat shock. However, the role of USP10 in CH remains largely unclear. Here, we investigated the roles of USP10 in CH. Methods and Results Cardiac-specific USP10 knockout (USP10-CKO) mice and USP10-transgenic (USP10-TG) mice were used to examined the role of USP10 in CH following aortic banding. The specific functions of USP10 were further examined in isolated cardiomyocytes. USP10 expression was increased in murine hypertrophic hearts following aortic banding and in isolated cardiomyocytes in response to hypertrophic agonist. Mice deficient in USP10 in the heart exhibited exaggerated cardiac hypertrophy and fibrosis following pressure overload stress, which resulted in worsening of cardiac contractile function. In contrast, cardiac overexpression of USP10 protected against pressure overload-induced maladaptive CH. Mechanistically, we demonstrated that USP10 activation and interaction with Sirt6 in response to angiotensin II led to a marked increase in the ubiquitination of Sirt6 and resulted in Akt signaling downregulation and attenuation of cardiomyocyte hypertrophy. Accordingly, inactivation of USP10 reduced Sirt6 abundance and stability and diminished Sirt6-induced downstream signaling in cardiomyocytes. Conclusions USP10 functions as a Sirt6 deubiquitinase that induces cardiac myocyte hypertrophy and triggers maladaptive CH.


Subject(s)
Cardiomegaly/etiology , Sirtuins/metabolism , Ubiquitin Thiolesterase/physiology , Angiotensin II , Animals , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Culture Techniques , Disease Models, Animal , Male , Mice , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Signal Transduction/physiology
16.
Cell Death Dis ; 11(2): 140, 2020 02 20.
Article in English | MEDLINE | ID: mdl-32080168

ABSTRACT

Cardiac hypertrophy (CH) is an independent risk factor for many cardiovascular diseases, and is one of the primary causes of morbidity and mortality in elderly people. Pathological CH involves excessive protein synthesis, increased cardiomyocyte size, and ultimately the development of heart failure. Myotubularin-related protein 14 (MTMR14) is a member of the myotubularin (MTM)-related protein family, which is involved in apoptosis, aging, inflammation, and autophagy. However, its exact function in CH is still unclear. Herein, we investigated the roles of MTMR14 in CH. We show that MTMR14 expression was increased in hypertrophic mouse hearts. Mice deficient in heart MTMR14 exhibited an aggravated aortic-banding (AB)-induced CH phenotype. In contrast, MTMR14 overexpression prevented pressure overload-induced hypertrophy. At the molecular level, prevention of CH in the absence of MTMR14 involved elevations in Akt pathway components, which are key elements that regulate apoptosis and cell proliferation. These results demonstrate that MTMR14 is a new molecular target for the treatment of CH.


Subject(s)
Hypertrophy, Left Ventricular/enzymology , Myocytes, Cardiac/enzymology , Phosphoric Monoester Hydrolases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Cell Proliferation , Cell Size , Disease Models, Animal , HEK293 Cells , Humans , Hypertrophy, Left Ventricular/pathology , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Male , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/pathology , Phosphoric Monoester Hydrolases/deficiency , Phosphoric Monoester Hydrolases/genetics , Rats, Sprague-Dawley , Signal Transduction , Ventricular Function, Left , Ventricular Remodeling
17.
Genes Genomics ; 41(10): 1223-1231, 2019 10.
Article in English | MEDLINE | ID: mdl-31350734

ABSTRACT

BACKGROUND: Postoperative cognitive dysfunction (POCD) is a neurodegenerative disorder with impairment of cognition. Sevoflurane anesthesia has been found to lead to CD and microRNAs (miRNAs) were reported to affect cognitive function. This study investigates the neuroprotective effect against sevoflurane anesthesia-induced CD. METHODS: HE staining was used to detect the pathological change of hippocampal neuron. Morris water maze test was used to analyze latency time, platform crossing and swimming speed. Quantitative real-time PCR (qRT-PCR) and western blotting were performed to examine the mRNA and protein expression of miR-410-3p, IL-6, TNF-α, IL-1ß and C-X-C motif chemokine receptor 5 (CXCR5). Dual-luciferase reporter assay was used to detect the relationship between miR-410-3p and CXCR5. RESULTS: MiR-410-3p was downregulated in sevoflurane anesthesia-induced rats and cells and act as a suppressor in sevoflurane anesthesia-induced hippocampal neuron apoptosis and inflammation. Furthermore, miR-410-3p was identified to bind with CXCR5. Further analysis showed that CXCR5 expression was increased by sevoflurane treatment, whereas was repressed by miR-410-3p overexpression. Moreover, miR-410-3p could inhibit sevoflurane anesthesia-induced hippocampal neuron apoptosis by phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt) pathway. CONCLUSION: These data indicated that miR-410-3p exhibited its neuroprotective effect on sevoflurane anesthesia-induced CD by targeting CXCR5 via PI3K/Akt signaling pathway. Our study may potentially provide a new light on the pathogenesis and therapeutic method for sevoflurane anesthesia-induced CD.


Subject(s)
Anesthesia/adverse effects , Cognitive Dysfunction/drug therapy , MicroRNAs/pharmacology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Sevoflurane/pharmacology , Signal Transduction/drug effects , Animals , Apoptosis/drug effects , Cognitive Dysfunction/chemically induced , Cognitive Dysfunction/pathology , Disease Models, Animal , Down-Regulation , Hippocampus/pathology , Inflammation , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Male , MicroRNAs/genetics , Neurons/drug effects , Postoperative Cognitive Complications , Rats , Receptors, CXCR5/drug effects , Receptors, CXCR5/metabolism , Receptors, Chemokine/metabolism , Tumor Necrosis Factor-alpha/metabolism
18.
RSC Adv ; 9(48): 27856-27864, 2019 Sep 03.
Article in English | MEDLINE | ID: mdl-35530470

ABSTRACT

Non-protein-coding RNAs (lncRNAs) are emerging as important regulators in disease pathogenesis, including atherosclerosis (AS). Here, we investigated the role and underlying mechanisms of nexilin F-actin binding protein antisense RNA 1 (NEXN-AS1) on the proliferation and migration of vascular smooth muscle cells (VSMCs). Our data revealed that ox-LDL treatment resulted in decreased NEXN-AS1 expression and increased miR-33a/b levels in human aorta VSMCs (HA-VSMCs) in dose- and time-dependent manners. Overexpression of NEXN-AS1 mitigated the proliferation and migration of HA-VSMCs under ox-LDL stimulation using CCK-8 and wound-healing assays. Moreover, dual-luciferase reporter and RNA immunoprecipitation assays verified that NEXN-AS1 acted as molecular sponges of miR-33a and miR-33b in HA-VSMCs. MiR-33a or miR-33b silencing attenuated the proliferation and migration of ox-LDL-treated HA-VSMCs. Furthermore, miR-33a or miR-33b mediated the inhibitory effects of NEXN-AS1 overexpression on the proliferation and migration of ox-LDL-treated HA-VSMCs. Our study suggested that high level of NEXN-AS1 mitigated VSMC proliferation and migration under ox-LDL stimulation at least partly through sponging miR-33a and miR-33b, illuminating NEXN-AS1 as a novel therapeutic approach for AS treatment.

19.
Int J Biol Sci ; 15(12): 2615-2626, 2019.
Article in English | MEDLINE | ID: mdl-31754334

ABSTRACT

Background/Aims: Vascular smooth muscle cell (VSMC) hyperplasia plays important roles in the pathogenesis of many vascular diseases, such as atherosclerosis and restenosis. Many microRNAs (miRs) have recently been reported to regulate the proliferation and migration of VSMC. In the current study, we aimed to explore the function of miR-93 in VSMCs and its molecular mechanism. Methods: First, qRT-PCR and immunofluorescence assays were performed to determine miR-93 expression in rat VSMCs following carotid artery injury in vivo and platelet-derived growth factor-BB (PDGF-BB) stimulation in vitro. Next, the biological role of miR-93 in rat VSMC proliferation and migration was examined in vivo and vitro. EdU incorporation assay and MTT assay for measuring cell proliferation, Transwell cell invasion assay and Cell scratch wound assay for measuring cell migration. Then, the targets of miR-93 were identified. Finally, the expression levels of proteins in the Raf-ERK1/2 pathway were measured by western blot. Results: MiR-93 was upregulated in rat VSMCs following carotid artery injury in vivo. Similar results were observed in ex vivo cultured VSMCs after PDGF-BB treatment. MiR-93 inhibition suppressed neointimal formation after carotid artery injury. Moreover, our results demonstrated that a miR-93 inhibitor suppressed the PDGF-BB induced proliferation and migration of in VSMC. This inhibitor also decreased the expression levels of MMP2 and cyclin D1. Mechanistically, we discovered that mitofusin 2(Mfn2) is a direct target of miR-93. Furthermore, an analysis of the signaling events revealed that miR-93-mediated VSMC proliferation and migration occurred via the Raf-ERK1/2 pathway. Conclusions: Our findings suggest that miR-93 promotes VSMCs proliferation and migration by targeting Mfn2. MiR-93 may be a new target for treating in-stent restenosis.


Subject(s)
Cell Movement/genetics , Cell Proliferation/genetics , GTP Phosphohydrolases/physiology , MicroRNAs/physiology , Mitochondrial Proteins/physiology , Muscle, Smooth, Vascular/cytology , Neointima/genetics , Animals , Cells, Cultured , GTP Phosphohydrolases/genetics , GTP Phosphohydrolases/metabolism , Male , MicroRNAs/genetics , MicroRNAs/metabolism , Mitochondrial Proteins/genetics , Mitochondrial Proteins/metabolism , Muscle, Smooth, Vascular/metabolism , Neointima/metabolism , Rats , Rats, Sprague-Dawley
20.
Free Radic Biol Med ; 121: 215-230, 2018 06.
Article in English | MEDLINE | ID: mdl-29733904

ABSTRACT

RATIONALE: Complement C1q tumor necrosis factor related proteins (C1QTNFs) have been reported to have diverse biological influence on the cardiovascular system. C1QTNF1 is a member of the CTRP superfamily. C1QTNF1 is expressed in the myocardium; however, its function in myocytes has not yet been investigated. OBJECTIVE: To systematically investigate the roles of C1QTNF1 in angiotensin II (Ang II)-induced cardiac hypertrophy. METHODS AND RESULTS: C1QTNF1 knock-out mice were used with the aim of determining the role of C1QTNF1 in cardiac hypertrophy in the adult heart. Data from experiments showed that C1QTNF1 was up-regulated during cardiac hypertrophic processes, which were triggered by increased reactive oxygen species. C1QTNF1 deficiency accelerated cardiac hypertrophy, fibrosis, inflammation responses, and oxidative stress with deteriorating cardiac dysfunction in the Ang II-induced cardiac hypertrophy mouse model. We identified C1QTNF1 as a negative regulator of cardiomyocyte hypertrophy in Ang II-stimulated neonatal rat cardiomyocytes using the recombinant human globular domain of C1QTNF1 and C1QTNF1 siRNA. Injection of the recombinant human globular domain of C1QTNF1 also suppressed the Ang II-induced cardiac hypertrophic response in vivo. The anti-hypertrophic effects of C1QTNF1 rely on AMPKa activation, which inhibits mTOR P70S6K phosphorylation. An AMPKa inhibitor abrogated the anti-hypertrophic effects of the recombinant human globular domain of C1QTNF1 both in vivo and vitro. Moreover, C1QTNF1-mediated AMPKa activation was triggered by the inhibition of PDE1-4, which subsequently activated the cAMP/PKA/LKB1 pathway. CONCLUSION: Our results demonstrated that C1QTNF1 improves cardiac function and inhibits cardiac hypertrophy and fibrosis by increasing and activating AMPKa, suggesting that C1QTNF1 could be a therapeutic target for cardiac hypertrophy and heart failure.


Subject(s)
Adipokines/physiology , Angiotensin II/toxicity , Cardiomegaly/prevention & control , Disease Models, Animal , Fibrosis/prevention & control , Protein Kinases/metabolism , Vasoconstrictor Agents/toxicity , AMP-Activated Protein Kinase Kinases , Animals , Cardiomegaly/chemically induced , Cardiomegaly/enzymology , Cardiomegaly/pathology , Cells, Cultured , Fibrosis/chemically induced , Fibrosis/enzymology , Fibrosis/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Phosphorylation , Rats , Rats, Sprague-Dawley , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL