Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 69
Filter
1.
Biochem Biophys Res Commun ; 725: 150248, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-38870847

ABSTRACT

The excessive migration and proliferation of vascular smooth muscle cells (VSMCs) plays a vital role in vascular intimal hyperplasia. CIRBP is involved in the proliferation of various cancer cells. This study was aimed to explore the role of CIRBP in the proliferation and migration of VSMCs. Adenovirus was used to interfere with cold-inducible RNA-binding protein (CIRBP) expression, while lentivirus was used to overexpress Ras homolog enriched in brain (Rheb). Western blotting and qRT-PCR were used to evaluate the expression of CIRBP, Rheb, and mechanistic target of rapamycin complex 1 (mTORC1) activity. The cell proliferation was determined by Ki67 immunofluorescence staining and CCK-8 assay. The wound healing assay was performed to assess cell migration. Additionally, immunohistochemistry was conducted to explore the role of CIRBP in intimal hyperplasia after vascular injury. We found that silencing CIRBP inhibited the proliferation and migration of VSMCs, decreased the expression of Rheb and mTORC1 activity. Restoration of mTORC1 activity via insulin or overexpression of Rheb via lentiviral transfection both attenuated the inhibitory effects of silencing CIRBP on the proliferation and migration of VSMCs. Moreover, Rheb overexpression abolished the inhibitory effect of silencing CIRBP on mTORC1 activity in VSMCs. CIRBP was upregulated in the injured carotid artery. Silencing CIRBP ameliorated intimal hyperplasia after vascular injury. In the summary, silencing CIRBP attenuates mTORC1 activity via reducing Rheb expression, thereby supressing the proliferation and migration of VSMCs and intimal hyperplasia after vascular injury.


Subject(s)
Cell Movement , Cell Proliferation , Mechanistic Target of Rapamycin Complex 1 , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , RNA-Binding Proteins , Ras Homolog Enriched in Brain Protein , Mechanistic Target of Rapamycin Complex 1/metabolism , Ras Homolog Enriched in Brain Protein/metabolism , Ras Homolog Enriched in Brain Protein/genetics , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/pathology , Animals , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/cytology , Cells, Cultured , Signal Transduction , Male , Rats , Rats, Sprague-Dawley , Humans
2.
Atherosclerosis ; 390: 117470, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38342025

ABSTRACT

BACKGROUND AND AIMS: Myotubularin-related protein 7 (MTMR7) suppresses proliferation in various cell types and is associated with cardiovascular and cerebrovascular diseases. However, whether MTMR7 regulates vascular smooth muscle cell (VSMC) and vascular intimal hyperplasia remains unclear. We explored the role of MTMR7 in phenotypic switching of VSMC and vascular intimal hyperplasia after injury. METHODS AND RESULTS: MTMR7 expression was significantly downregulated in injured arteries. Compared to wild type (WT) mice, Mtmr7-transgenic (Mtmr7-Tg) mice showed reduced intima/media ratio, decreased percentage of Ki-67-positive cells within neointima, and increased Calponin expression in injured artery. In vitro, upregulating MTMR7 by Len-Mtmr7 transfection inhibited platelet derived growth factor (PDGF)-BB-induced proliferation, migration of VSMC and reversed PDGF-BB-induced decrease in expression of Calponin and SM-MHC. Microarray, single cell sequence, and other bioinformatics analysis revealed that MTMR7 is highly related to glucose metabolism and mammalian target of rapamycin complex 1 (mTORC1). Further experiments confirmed that MTMR7 markedly repressed glycolysis and mTORC1 activity in PDGF-BB-challenged VSMC in vitro. Restoring mTORC1 activity abolished MTMR7-mediated suppression of glycolysis, phenotypic shift in VSMC in vitro and protection against vascular intimal hyperplasia in vivo. Furthermore, upregulating MTMR7 in vitro led to dephosphorylation and dissociation of p62 from mTORC1 in VSMC. External expression of p62 in vitro also abrogated the inhibitory effects of MTMR7 on glycolysis and phenotypic switching in PDGF-BB-stimulated VSMC. CONCLUSIONS: Our study demonstrates that MTMR7 inhibits injury-induced vascular intimal hyperplasia and phenotypic switching of VSMC. Mechanistically, the beneficial effects of MTMR7 are conducted via suppressing p62/mTORC1-mediated glycolysis.


Subject(s)
Muscle, Smooth, Vascular , Neointima , Mice , Animals , Becaplermin/pharmacology , Becaplermin/metabolism , Cell Proliferation , Muscle, Smooth, Vascular/pathology , Hyperplasia/pathology , Neointima/metabolism , Mice, Transgenic , Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 1/pharmacology , Glucose/metabolism , Myocytes, Smooth Muscle/pathology , Cell Movement , Cells, Cultured , Mammals
3.
J Cardiovasc Pharmacol ; 82(3): 221-228, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37381169

ABSTRACT

ABSTRACT: Excessive proliferation and migration of vascular smooth muscle cells (VSMCs) cause neointimal hyperplasia after percutaneous vascular interventions. Nuclear receptor subfamily 1 group D member 1 (NR1D1), a crucial member of circadian clock, is involved in the regulation of atherosclerosis and cellular proliferation. However, whether NR1D1 affects vascular neointimal hyperplasia remains unclear. In this study, we found that activating NR1D1 reduced injury-induced vascular neointimal hyperplasia. Overexpression of NR1D1 reduced the number of Ki-67-positive VSMCs and migrated VSMCs after platelet-derived growth factor (PDGF)-BB treatment. Mechanistically, NR1D1 suppressed the phosphorylation of AKT and 2 main effectors of the mammalian target of rapamycin complex 1 (mTORC1), S6, and 4EBP1 in PDGF-BB-challenged VSMCs. Re-activation of mTORC1 by Tuberous sclerosis 1 siRNA (si Tsc1 ) and re-activation of AKT by SC-79 abolished NR1D1-mediated inhibitory effects on proliferation and migration of VSMCs. Moreover, decreased mTORC1 activity induced by NR1D1 was also reversed by SC-79. Simultaneously, Tsc1 knockdown abolished the vascular protective effects of NR1D1 in vivo. In conclusion, NR1D1 reduces vascular neointimal hyperplasia by suppressing proliferation and migration of VSMCs in an AKT/mTORC1-dependent manner.


Subject(s)
Muscle, Smooth, Vascular , Vascular System Injuries , Humans , Hyperplasia/metabolism , Muscle, Smooth, Vascular/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Cell Proliferation , Becaplermin/pharmacology , Vascular System Injuries/pathology , Neointima/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Myocytes, Smooth Muscle/metabolism , Cell Movement , Cells, Cultured
4.
Clin Exp Hypertens ; 45(1): 2178659, 2023 Dec 31.
Article in English | MEDLINE | ID: mdl-36794491

ABSTRACT

BACKGROUND: In-stent restenosis hardly limits the therapeutic effect of the percutaneous vascular intervention. Although the restenosis is significantly ameliorated after the application of new drug-eluting stents, the incidence of restenosis remains at a high level. OBJECTIVE: Vascular adventitial fibroblasts (AFs) play an important role in intimal hyperplasia and subsequent restenosis. The current study was aimed to investigate the role of nuclear receptor subfamily 1, group D, member 1 (NR1D1) in the vascular intimal hyperplasia. METHODS AND RESULTS: We observed increased expression of NR1D1 after the transduction of adenovirus carrying Nr1d1 gene (Ad-Nr1d1) in AFs. Ad-Nr1d1 transduction significantly reduced the numbers of total AFs, Ki-67-positive AFs, and the migration rate of AFs. NR1D1 overexpression decreased the expression level of ß-catenin and attenuated the phosphorylation of the effectors of mammalian target of rapamycin complex 1 (mTORC1), including mammalian target of rapamycin (mTOR) and 4E binding protein 1 (4EBP1). Restoration of ß-catenin by SKL2001 abolished the inhibitory effects of NR1D1 overexpression on the proliferation and migration of AFs. Surprisingly, the restoration of mTORC1 activity by insulin could also reverse the decreased expression of ß-catenin, attenuated proliferation, and migration in AFs induced by NR1D1 overexpression. In vivo, we found that SR9009 (an agonist of NR1D1) ameliorated the intimal hyperplasia at days 28 after injury of carotid artery. We further observed that SR9009 attenuated the increased Ki-67-positive AFs, an essential part of vascular restenosis at days 7 after injury to the carotid artery. CONCLUSION: These data suggest that NR1D1 inhibits intimal hyperplasia by suppressing the proliferation and migration of AFs in a mTORC1/ß-catenin-dependent manner.


Subject(s)
Muscle, Smooth, Vascular , Nuclear Receptor Subfamily 1, Group D, Member 1 , beta Catenin , beta Catenin/metabolism , Cell Movement , Cell Proliferation , Cells, Cultured , Fibroblasts , Hyperplasia/metabolism , Hyperplasia/pathology , Ki-67 Antigen/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Myocytes, Smooth Muscle , Neointima/genetics , Neointima/metabolism , Neointima/pathology , Nuclear Receptor Subfamily 1, Group D, Member 1/metabolism , TOR Serine-Threonine Kinases/metabolism
5.
Mitochondrion ; 65: 150-160, 2022 07.
Article in English | MEDLINE | ID: mdl-35779798

ABSTRACT

Microvascular protection is the main mechanism of metformin against diabetic complications. Cardiac microvascular endothelial cells (CMECs) are the basic component of cardiac microvessels, and they suffer from oxidative stress and mitochondrial dysfunction under type 2 diabetes mellitus (T2DM). Translocase of the outer mitochondrial membrane 70 (Tom70) improves mitochondrial dysfunction, but its role in the hearts of T2DM patients remains unclear. The purpose of this study was to demonstrate the protective effect of metformin on diabetic cardiac microvascular injury and to identify the role of Tom70 in this effect. T2DM mice were established by multiple intraperitoneal injections of low-dose streptozotocin and 12-week high-fat feeding. CMECs were isolated and cultured with normal glucose (NG), high glucose (HG), and HG plus high fat (HG-HF) media. The results indicated that long-term metformin treatment partly reversed cardiovascular complication and mitigated cardiac microvascular injury in T2DM. In addition, exposure to HG-HF led to CMEC damage, aggravated oxidative stress, aggravated mitochondrial dysfunction, and reduced mitochondrial Tom70 expression, whereas upregulation of Tom70 significantly ameliorated these injuries. Furthermore, metformin treatment promoted Tom70 expression and effectively reversed CMEC injury induced by HG-HF. However, all of these effects were interrupted after Tom70 was knocked down. In conclusion, T2DM damages microvascular integrity by activating a cycle of decreased Tom70 expression, mitochondrial dysfunction, and reactive oxygen species (ROS) overload in CMECs. However, metformin suppresses oxidative stress, relieves mitochondrial dysfunction, and promotes the expression of Tom70, ultimately ameliorating diabetic microvascular injury and heart complications.


Subject(s)
Diabetes Mellitus, Type 2 , Metformin , Animals , Mice , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Metformin/metabolism , Metformin/pharmacology , Metformin/therapeutic use , Mitochondrial Membrane Transport Proteins/metabolism , Mitochondrial Precursor Protein Import Complex Proteins , Oxidative Stress
6.
Bioengineered ; 13(4): 10665-10678, 2022 04.
Article in English | MEDLINE | ID: mdl-35470759

ABSTRACT

Abnormal proliferation and migration of vascular smooth muscle cell (VSMC) is a hallmark of vascular neointima hyperplasia. Perilipin 5 (Plin5), a regulator of lipid metabolism, is also confirmed to be involved in vascular disorders, such as microvascular endothelial dysfunction and atherosclerosis. To investigate the regulation and function of plin5 in the phenotypic alteration of VSMC, -an animal model of vascular intima hyperplasia was established in C57BL/6 J and Plin5 knockdown (Plin5±) mice by wire injure. Immunohistochemical staining was used to analyze neointima hyperplasia in artery. Ki-67, dihydroethidium immunofluorescence staining and wound healing assay were used to measure proliferation, reactive oxygen species (ROS) generation and migration of VSMC, respectively. Plin5 was downregulated in artery subjected to vascular injury and in VSMC subjected to platelet-derived growth factor (PDGF)-BB. Plin5 knockdown led to accelerated neointima hyperplasia, excessive proliferation and migration of VSMC after injury. In vitro, we observed increased ROS content in VSMC isolated from Plin5± mice. Antioxidative N-acetylcysteine (NAC) inhibited VSMC proliferation and migration induced by PDGF-BB or plin5 knockdown. More importantly, plin5-peroxlsome proliferator-activated receptor-γ coactivator (PGC)-1α interaction was also attenuated in VSMC after knockdown of plin5. Overexpression of PGC-1α suppressed PDGF-BB-induced ROS generation, proliferation, and migration in VSMC isolated from Plin5± mice. These data suggest that plin5 serves as a potent regulator of VSMC proliferation, migration, and neointima hyperplasia by interacting with PGC-1α and affecting ROS generation.


Subject(s)
Neointima , Transcription Factors/metabolism , Vascular System Injuries , Animals , Becaplermin , Cell Movement/genetics , Cell Proliferation , Cells, Cultured , Hyperplasia/metabolism , Hyperplasia/pathology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/pathology , Neointima/genetics , Neointima/metabolism , Neointima/pathology , Perilipin-5/metabolism , Reactive Oxygen Species/metabolism , Vascular System Injuries/genetics , Vascular System Injuries/metabolism , Vascular System Injuries/pathology
7.
BMB Rep ; 55(5): 244-249, 2022 May.
Article in English | MEDLINE | ID: mdl-35410639

ABSTRACT

Characterized by abnormal proliferation and migration of vascular smooth muscle cells (VSMCs), neointima hyperplasia is a hallmark of vascular restenosis after percutaneous vascular interventions. Vaccinia-related kinase 1 (VRK1) is a stress adaptionassociated ser/thr protein kinase that can induce the proliferation of various types of cells. However, the role of VRK1 in the proliferation and migration of VSMCs and neointima hyperplasia after vascular injury remains unknown. We observed increased expression of VRK1 in VSMCs subjected to platelet-derived growth factor (PDGF)-BB by western blotting. Silencing VRK1 by shVrk1 reduced the number of Ki-67-positive VSMCs and attenuated the migration of VSMCs. Mechanistically, we found that relative expression levels of ß-catenin and effectors of mTOR complex 1 (mTORC1) such as phospho (p)-mammalian target of rapamycin (mTOR), p-S6, and p-4EBP1 were decreased after silencing VRK1. Restoration of ß-catenin expression by SKL2001 and re-activation of mTORC1 by Tuberous sclerosis 1 siRNA (siTsc1) both abolished shVrk1-mediated inhibitory effect on VSMC proliferation and migration. siTsc1 also rescued the reduced expression of ß-catenin caused by VRK1 inhibition. Furthermore, mTORC1 re-activation failed to recover the attenuated proliferation and migration of VSMC resulting from shVrk1 after silencing ß-catenin. We also found that the vascular expression of VRK1 was increased after injury. VRK1 inactivation in vivo inhibited vascular injury-induced neointima hyperplasia in a ß-catenin-dependent manner. These results demonstrate that inhibition of VRK1 can suppress the proliferation and migration of VSMC and neointima hyperplasia after vascular injury via mTORC1/ß-catenin pathway. [BMB Reports 2022; 55(5): 244-249].


Subject(s)
Intracellular Signaling Peptides and Proteins , Muscle, Smooth, Vascular , Neointima , Protein Serine-Threonine Kinases , TOR Serine-Threonine Kinases , Vascular System Injuries , beta Catenin , Becaplermin/pharmacology , Carotid Intima-Media Thickness , Cell Movement , Cell Proliferation , Cells, Cultured , Humans , Hyperplasia , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Neointima/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism , Vascular System Injuries/metabolism , Vascular System Injuries/pathology , beta Catenin/metabolism
8.
J Cardiovasc Pharmacol ; 79(5): 739-748, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35275098

ABSTRACT

ABSTRACT: This study was designed to investigate the role and mechanism of PIKfyve in the proliferation and migration of vascular smooth muscle cells (VSMCs) and vascular intima hyperplasia. We first observed increased protein levels of PIKfyve, phospho (p)-S6 Ribosomal Protein (S6)Ser235/236, p-4EBP1Thr37/46 in VSMCs after 24 hours of platelet-derived growth factor (PDGF)-BB treatment. By using cell counting kit-8 assay, Ki-67 immunofluorescence staining and wound healing assay, we found that PIKfyve inhibition ameliorated the enhanced activity of VSMC proliferation and migration induced by PDGF-BB. Silencing PIKfyve also suppressed the phosphorylation of S6 and 4EBP1 (2 major effectors of mammalian target of rapamycin complex 1), glucose consumption, activity of hexokinase, and LDH in PDGF-BB-challenged VSMCs. After rescuing the phosphorylation of S6 and 4EBP1 by silencing Tsc1, the suppressive effects of PIKfyve inhibition on glucose utilization, proliferation, and migration in VSMCs were abolished. The animal model of vascular restenosis was established in C57BL/6J mice by wire injury. We found the expression of PIKfyve was increased in carotid artery at day 28 after injury. Reducing the activity of PIKfyve alleviated vascular neointima hyperplasia after injury. In conclusion, targeting PIKfyve might be a novel effective method to reduce the proliferation and migration of VSMCs and vascular restenosis by affecting mammalian target of rapamycin complex 1-mediated glucose utilization.


Subject(s)
Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Animals , Becaplermin/pharmacology , Cell Movement , Cell Proliferation , Cells, Cultured , Glucose/metabolism , Hyperplasia/metabolism , Hyperplasia/pathology , Mammals , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Neointima/metabolism , Neointima/pathology , Phosphatidylinositol 3-Kinases/metabolism , Tunica Intima
9.
Tohoku J Exp Med ; 256(2): 131-139, 2022 02.
Article in English | MEDLINE | ID: mdl-35197406

ABSTRACT

Type 2 diabetes mellitus (T2DM)-associated mitochondrial impairment may a key factor leading to liver injury. Transient receptor potential receptor vanilloid 1 (TRPV1) regulates the energy expenditure and cholesterol metabolism in hepatocytes and protects against oxidative toxicity. Optic atrophy 1 (OPA1) is involved in the protection of TRPV1 on cardiac microvascular and lung injury. The aim of this study is to identify the role of TRPV1 in redox signals and liver protection via OPA1. TRPV1 knockout (TRPV1-/-) mice were used. And T2DM associated liver injury was induced by high glucose and high fatty acid (HG/HF) treatment. Mechanisms were studied by TUNEL staining, transmission electron microscope (TEM) analysis, reverse transcription polymerase chain reaction (RT-PCR) and Western blotting in vivo and in vitro. We determined that HG/HF treatment increased TRPV1 expression in liver tissues and AML12 cells. The knockout of TRPV1 increased the apoptotic hepatocytes rate. The inhibition of TRPV1 by 5'-iRTX in HG/HF group elevated the reactive oxygen species (ROS) levels, whereas TRPV1 agonist capsaicin reduced ROS. Our studies also showed that the OPA1 expression was lower in livers from HG/HF treated mice than the control, and genetic ablation of TRPV1 decreased OPA1 expression to a greater extent than the HG/HF mice. The protective effects of TRPV1 on mitochondrial were blocked by OPA1 siRNA. In conclusion, our study showed that the identified regulation of TRPV1 to OPA1 has important implication to the pathogenesis of T2DM-associated liver injury. Targeting the action of TRPV1 and OPA1 presents a potential therapeutic intervention.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Diabetes Mellitus, Type 2 , GTP Phosphohydrolases , Hyperglycemia , Hyperlipidemias , TRPV Cation Channels , Animals , Apoptosis , Hyperglycemia/complications , Mice , TRPV Cation Channels/genetics , TRPV Cation Channels/metabolism
10.
Cardiovasc Diagn Ther ; 10(6): 1770-1784, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33381422

ABSTRACT

BACKGROUND: Antithrombotic therapy is a cornerstone of acute myocardial infarction (AMI) treatment and is thought to be associated with an increased risk of chronic subdural hematoma (CSDH). However, no well-established model exists to predict subsequent antithrombotic treatment outcomes after CSDH in patients with recent AMI. We aimed to identify a prognostic model to predict the 6-month outcome of treatment with antithrombotic therapy. METHODS: This multicenter retrospective analysis involved 553 patients with recent AMI with antithrombotic-related CSDH. Several candidate clinical variables and biomarkers were examined in the training cohort (Chengdu training cohort; n=368). Patients with unfavorable outcomes had experienced at least 1 of the following: major adverse cardiovascular events (MACE), recurrence, or a modified Rankin scale (mRS) score of 2 to 6. To develop a 6-month outcome prediction model, three approaches were used: (I) a demographic variable model, (II) a clinical marker model and (III) a decision-driven model. A clinical outcome prediction model based on the superior predictors was assessed by logistic regression analysis. The nomogram for the final model was internally validated using a bootstrap procedure and externally validated in an independent cohort (Anhui cohort; n=185). RESULTS: Model A produced 7 predictors of unfavorable outcomes, while models B and C yielded 2 and 1 predictors, respectively. The areas under the curve (AUC) increased from 0.743 [model A; 95% confidence interval (CI): 0.680-0.782] to 0.889 (model A + B + C; 95% CI: 0.851-0.916). The final prediction model included age, systolic blood pressure (SBP), body mass index (BMI), the Glasgow Coma Scale (GCS), the estimated glomerular filtration rate (eGFR), the early resumption of antithrombotic therapy, hematoma thickness and the presence of abdominal obesity, frailty and previous bleeding. Internal and external validation of the selected final model revealed adequate C-statistics and calibration slope values (internal validation: 0.81 and 0.78; external validation: 0.80 and 0.76, respectively). CONCLUSIONS: This model provided a risk stratification tool to predict unfavorable outcomes in patients with recent AMI with antithrombotic-related CSDH. Because the study was based on ten readily practical and available variables, it may be widely applicable to guide management and complement clinical assessment.

11.
Aging (Albany NY) ; 13(1): 493-515, 2020 11 26.
Article in English | MEDLINE | ID: mdl-33259334

ABSTRACT

Mitochondrial calcium uptake 1 (MICU1) is a pivotal molecule in maintaining mitochondrial homeostasis under stress conditions. However, it is unclear whether MICU1 attenuates mitochondrial stress in angiotensin II (Ang-II)-induced cardiac hypertrophy or if it has a role in the function of melatonin. Here, small-interfering RNAs against MICU1 or adenovirus-based plasmids encoding MICU1 were delivered into left ventricles of mice or incubated with neonatal murine ventricular myocytes (NMVMs) for 48 h. MICU1 expression was depressed in hypertrophic myocardia and MICU1 knockdown aggravated Ang-II-induced cardiac hypertrophy in vivo and in vitro. In contrast, MICU1 upregulation decreased cardiomyocyte susceptibility to hypertrophic stress. Ang-II administration, particularly in NMVMs with MICU1 knockdown, led to significantly increased reactive oxygen species (ROS) overload, altered mitochondrial morphology, and suppressed mitochondrial function, all of which were reversed by MICU1 supplementation. Moreover, peroxisome proliferator-activated receptor gamma coactivator 1-α (PGC-1α)/MICU1 expression in hypertrophic myocardia increased with melatonin. Melatonin ameliorated excessive ROS generation, promoted mitochondrial function, and attenuated cardiac hypertrophy in control but not MICU1 knockdown NMVMs or mice. Collectively, our results demonstrate that MICU1 attenuates Ang-II-induced cardiac hypertrophy by inhibiting mitochondria-derived oxidative stress. MICU1 activation may be the mechanism underlying melatonin-induced protection against myocardial hypertrophy.


Subject(s)
Antioxidants/pharmacology , Calcium-Binding Proteins/genetics , Cardiomegaly/genetics , Melatonin/pharmacology , Mitochondria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Oxidative Stress/genetics , Angiotensin II/toxicity , Animals , Calcium-Binding Proteins/drug effects , Calcium-Binding Proteins/metabolism , Cardiomegaly/chemically induced , Cardiomegaly/metabolism , Disease Models, Animal , Gene Knockdown Techniques , Heart/drug effects , In Vitro Techniques , Mice , Mitochondria/drug effects , Mitochondrial Membrane Transport Proteins/drug effects , Mitochondrial Membrane Transport Proteins/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Oxidative Stress/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Reactive Oxygen Species/metabolism , Vasoconstrictor Agents/toxicity
12.
Life Sci ; 262: 118496, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32987060

ABSTRACT

The infection epidemic event of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) was formally declared a pandemic by World Health Organization on March 11th, 2020. Corona Virus Disease 2019 (COVID-19) is caused by SARS-CoV-2, a new type of coronavirus, which has high contagion and mainly causes respiratory symptoms. With the increase in confirmed cases, however, the infection symptoms turn to be diverse with secondary or first clinical symptoms relating to damage of the cardiovascular system and changes of myocardial enzyme spectrum, cardiac troponin I, electrocardiogram, cardiac function. The occurrence of extra-pulmonary manifestations, including immediately and long-term damage, means that the overall health burden caused by SARS-CoV-2 infection may be under-estimated because COVID-19 patients developed cardiovascular system injury are more likely to become serious. The factors such as directly pathogen-mediated damage to cardiomyocytes, down-regulated angiotensin-converting enzyme 2 (ACE2) expression, excessive inflammatory response, hypoxia and adverse drug reaction, are closely related to the occurrence and development of the course of COVID-19. In combination with recently published medical data of patients having SARS-CoV-2 infection and the latest studies, the manifestations of damage to cardiovascular system by COVID-19, possible pathogenic mechanisms and advances of the treatment are proposed in this article.


Subject(s)
COVID-19 Drug Treatment , COVID-19/complications , Cardiomyopathies/complications , Cardiomyopathies/drug therapy , SARS-CoV-2/drug effects , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Down-Regulation/drug effects , Humans , Pandemics
13.
Aging (Albany NY) ; 12(10): 9726-9744, 2020 05 18.
Article in English | MEDLINE | ID: mdl-32422606

ABSTRACT

Neurofibromin 2 (NF2), a potent tumor suppressor, is reported to inhibit proliferation in several cell types. The role of NF2 in neointima hyperplasia after vascular injury is unknown. We explored the role of NF2 in proliferation, migration of vascular smooth muscle cell (VSMC) and neointima hyperplasia after vascular injury. NF2 phosphorylation was elevated in VSMC subjected to platelet-derived growth factor (PDGF)-BB and in artery subjected to vascular injury. Mice deficient for Nf2 in VSMC showed enhanced neointima hyperplasia after injury, increased proliferation and migration of VSMC after PDGF-BB treatment. Mechanistically, we observed increased nuclear p-NF2, declined p-Yes-Associated Protein (YAP), nuclear translocation of YAP after PDGF-BB treatment or injury. NF2 knockdown or YAP overexpression showed similar phenotype in VSMC proliferation, migration and neointima hyperplasia. YAP inhibition abolished the above effects mediated by NF2 knockdown. Finally, NF2 knockdown further promoted YAP-TEA Domain Transcription Factor 1 (TEAD1) interaction after PDGF-BB treatment. Inhibition of TEAD1 blocked PDGF-BB-induced VSMC proliferation and migration, which were not reversed by either NF2 knockdown or YAP overexpression. In conclusion, NF2 knockdown promotes VSMC proliferation, migration and neointima hyperplasia after vascular injury via inducing YAP-TEAD1 interaction.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Neointima/genetics , Neurofibromin 2/deficiency , Transcription Factors/metabolism , Vascular Remodeling/genetics , Vascular System Injuries/genetics , Animals , Cell Proliferation/genetics , Hyperplasia/genetics , Mice , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , TEA Domain Transcription Factors , YAP-Signaling Proteins
14.
Atherosclerosis ; 301: 44-53, 2020 05.
Article in English | MEDLINE | ID: mdl-32325260

ABSTRACT

BACKGROUND AND AIMS: TRPA1 is a calcium permeable non-selective cation channel, its expression is up-regulated in atherosclerosis plaque, yet its function in macrophages activation is unknown. We sought to establish the role of TRPA1 in inflammatory macrophages activation. METHODS: TRPA1-/-ApoE-/- mice and C57BL/6 J control were treated with a high-fat diet (HFD) and the TRPA1 agonist cinnamaldehyde (CIN). Third-order branches of superior aorta of patients and mice were collected. Oil Red O staining and hematoxylin and eosin staining were performed to measure atherosclerotic lesions. The RNA-seq was performed to identify TRPA1 function in atherosclerosis. The expression of bone marrow-derived macrophages (BMDMs) markers was tested by Western blot. In addition, the levels of inflammatory factors were checked by ELISA. Chromatin immunoprecipitation (ChIP)-PCR and luciferase reporter gene assays were used to explore if TRPA1 could regulate histone modifications. RESULTS: TRPA1-/-ApoE-/- mice showed a significant increase in atherosclerosis plaques compared to ApoE-/- mice after HFD treatment. Conversely, activation of TRPA1 by CIN sharply reduced atherosclerosis progression. Atherosclerosis was associated with a significant change in macrophage polarization toward the M1 proinflammatory phenotype. We found that inhibition of TRPA1 remarkably stimulated M1 marker genes expression, while repressed M2 marker genes expression. The interaction between TRPA1 and Ezh2, a subunit of polycomb repressive complex 2, suppressed the proteasome-dependent degradation of Ezh2. Thus, TRPA1 epigenetically regulated H3K27 trimethylation level in macrophages. CONCLUSIONS: Our results demonstrate that TRPA1, up-regulated in atherosclerosis plaque, could regulate the macrophages toward an inflammatory phenotype, thereby modulating atherosclerosis progression. Activation of TRPA1 might serve as an atherosclerosis therapeutic target.


Subject(s)
Atherosclerosis , Plaque, Atherosclerotic , Animals , Apolipoproteins E/genetics , Atherosclerosis/genetics , Disease Models, Animal , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , TRPA1 Cation Channel/genetics
15.
J Cardiovasc Pharmacol ; 75(6): 556-563, 2020 06.
Article in English | MEDLINE | ID: mdl-32195756

ABSTRACT

Ischemia and anoxia-induced mitochondrial impairment may be a key factor leading to heart injury during myocardial infarction (MI). Calpain 1 and 2 are involved in the MI-induced mitochondria injury. G protein-coupled receptor 35 (GPR35) could be triggered by hypoxia. Whether or not GPR35 regulates calpain 1/2 in the pathogenesis of MI is still unclear. In this study, we determined that MI increases GPR35 expression in myocardial tissue. Suppression of GPR35 protects heart from MI injury in mice through reduction of reactive oxygen species activity and mitochondria-dependent apoptosis. Further studies show that GPR35 regulates calpain 1/2. Suppression of GPR35 reduces the expression and activity of calpain 1/2, and alleviates calpain 1/2-associated mitochondrial injury to preserve cardiac function. Based on these data, we conclude that a functional inhibition of GPR35 downregulates calpain 1/2 and contributes to maintenance of cardiac function under pathologic conditions with mitochondrial disorder. In conclusion, our study showed that the identified regulation by GPR35 of calpain 1/2 has important implications for the pathogenesis of MI. Targeting the action of GPR35 and calpain 1/2 in mitochondria presents a potential therapeutic intervention for MI.


Subject(s)
Calpain/metabolism , Mitochondria, Heart/enzymology , Myocardial Infarction/therapy , Myocytes, Cardiac/enzymology , RNA, Small Interfering/administration & dosage , RNAi Therapeutics , Receptors, G-Protein-Coupled/metabolism , Animals , Apoptosis , Calpain/genetics , Cells, Cultured , Disease Models, Animal , Male , Mice, Inbred C57BL , Mitochondria, Heart/pathology , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Oxidative Stress , Reactive Oxygen Species/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction
16.
Am J Hypertens ; 33(3): 223-233, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31648306

ABSTRACT

BACKGROUND: Transient receptor potential melastatin subtype 8 (TRPM8) is a cold-sensing cation channel, mainly localized in the sensory neurons, which can be activated by menthol, a compound with a naturally cold sensation in mint. However, the effect of TRPM8 activation in inflammation and cardiac remodeling after myocardial infarction (MI) is not well defined. METHODS: TRPM8 knockout (KO) mice (TRPM8-/-) and their wild-type littermates, aged 8 weeks, were randomly divided into sham and MI groups and were fed with chow or chow plus menthol. RESULTS: Dietary menthol significantly attenuated MI injury, evidenced by decreased survival rates and plasma cardiac troponion I levels, reduced infarct size and cardiomyocytes, declined collagen deposition, and rescued cardiac function and hemodynamics. However, these effects of menthol disappeared when mice were lacking TRPM8. Furthermore, feeding of menthol ameliorated elevated expression of inflammatory cytokines and chemokines, and aggravated inflammation infiltration in the MI mice but not in TRPM8-/- mice. In addition, menthol treatment increased the release of calcitonin gene-related peptide (CGRP), which were absent in TRPM8-/- mice. CONCLUSIONS: In conclusion, our results suggest that dietary menthol can protect against inflammation and cardiac remodeling after MI through activation of TRPM8.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Inflammation Mediators/metabolism , Inflammation/prevention & control , Menthol/administration & dosage , Myocardial Infarction/drug therapy , Myocardium/metabolism , TRPM Cation Channels/agonists , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Diet , Disease Models, Animal , Fibrosis , Hemodynamics/drug effects , Inflammation/metabolism , Inflammation/physiopathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocardium/pathology , Signal Transduction , TRPM Cation Channels/genetics , TRPM Cation Channels/metabolism
17.
Oxid Med Cell Longev ; 2019: 8690746, 2019.
Article in English | MEDLINE | ID: mdl-31772713

ABSTRACT

BACKGROUND: Hyper-free fatty acidemia (HFFA) impairs cardiac capillaries, as well as type 2 diabetes mellitus (T2DM). Perilipin 5 (Plin5) maintains metabolic balance of free fatty acids (FFAs) in high oxidative tissues via the states of nonphosphorylation and phosphorylation. However, when facing to T2DM-HFFA, Plin5's role in cardiac microvascular endothelial cells (CMECs) is not defined. METHODS: In mice of WT or Plin5-/-, T2DM models were rendered by high-fat diet combined with intraperitoneal injection of streptozocin. CMECs isolated from left ventricles were incubated with high glucose (HG) and high FFAs (HFFAs). Plin5 phosphorylation was stimulated by isoproterenol. Plin5 expression was knocked down by small interfering RNA (siRNA). We determined cardiac function by small animal ultrasound, apoptotic rate by flow cytometry, microvessel quantity by immunohistochemistry, microvascular integrity by scanning electron microscopy, intracellular FFAs by spectrophotometry, lipid droplets (LDs) by Nile red staining, mRNAs by quantitative real-time polymerase chain reaction, proteins by western blots, nitric oxide (NO) and reactive oxygen species (ROS) by fluorescent dye staining and enzyme-linked immunosorbent assay kits. RESULTS: In CMECs, HFFAs aggravated cell injury induced by HG and activated Plin5 expression. In mice with T2DM-HFFA, Plin5 deficiency reduced number of cardiac capillaries, worsened structural incompleteness, and enhanced diastolic dysfunction. Moreover, in CMECs treated with HG-HFFAs, both ablation and phosphorylation of Plin5 reduced LDs content, increased intracellular FFAs, stimulated mitochondrial ß-oxidation, added ROS generation, and reduced the expression and activity of endothelial nitric oxide synthase (eNOS), eventually leading to increased apoptotic rate and decreased NO content, all of which were reversed by N-acetyl-L-cysteine. CONCLUSION: Plin5 preserves lipid balance and cell survival in diabetic CMECs by regulating FFAs metabolism bidirectionally via the states of nonphosphorylation and phosphorylation.


Subject(s)
CME-Carbodiimide/metabolism , Diabetes Mellitus, Type 2/drug therapy , Fatty Acids, Nonesterified/metabolism , Gene Expression/genetics , Perilipin-5/therapeutic use , Reactive Oxygen Species/metabolism , Animals , Mice , Perilipin-5/pharmacology , Transfection
18.
Toxicol Appl Pharmacol ; 383: 114747, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31499192

ABSTRACT

Abnormal proliferation of vascular smooth muscle cells (VSMCs) is a hallmark of vascular restenosis. We investigated whether polypyrimidine tract-binding protein 1 (PTBP1), a novel regulator of cell proliferation and differentiation, is implicated in VSMC proliferation and neointima hyperplasia responding to injury. C57BL/6 J mice of 10-12 weeks old were randomly divided into sham and carotid artery injury group. Primary VSMCs obtained from thoracic aortas of 10- to 12-week-old mice were treated with physiological saline and platelet derived growth factor-BB (PDGF-BB). Adenovirus expressing shCon, shPTBP1 or shYY2 were transfected into the injured common carotid artery or VSMCs. qRT-PCR and immunoblotting were used to determine the mRNA and protein expression levels, respectively. Immunohistochemical staining of H&E and Ki-67 were used to evaluate restenosis of vessels. Cell counting kit-8 assay and Ki-67 immunofluorescent staining were utilized to evaluate the rate of VSMC proliferation. The expression of PTBP1 were upregulated both in injured arteries and in PDGF-BB-treated VSMCs. PTBP1 inhibition significantly attenuated neointima hyperplasia and Ki-67 positive area induced by injury. Knockdown of PTBP1 in vitro also suppressed VSMC proliferation after PDGF-BB treatment. The effects of PTBP1 inhibition mentioned above were all abolished by knockdown of YY2. Finally, we identified four cell cycle regulators (p53, p21, Cdkn1c, Cdkn2b) that were regulated by PTBP1/YY2 axis both in vitro and in vivo. These findings demonstrated that PTBP1 is a critical regulator of VSMC proliferation and neointima hyperplasia via modulating the expression of YY2.


Subject(s)
Cell Proliferation/physiology , Heterogeneous-Nuclear Ribonucleoproteins/physiology , Hyperplasia/metabolism , Muscle, Smooth, Vascular/metabolism , Neointima/metabolism , Polypyrimidine Tract-Binding Protein/physiology , Transcription Factors/biosynthesis , Animals , Becaplermin/pharmacology , Cell Proliferation/drug effects , Heterogeneous-Nuclear Ribonucleoproteins/antagonists & inhibitors , Hyperplasia/pathology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Neointima/pathology , Polypyrimidine Tract-Binding Protein/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/physiology
19.
Oxid Med Cell Longev ; 2019: 6408352, 2019.
Article in English | MEDLINE | ID: mdl-31217840

ABSTRACT

Cardiac fibroblasts (CFs) are a critical cell population responsible for myocardial extracellular matrix homeostasis. After stimulation by myocardial infarction (MI), CFs transdifferentiate into cardiac myofibroblasts (CMFs) and play a fundamental role in the fibrotic healing response. Transient receptor potential ankyrin 1 (TRPA1) channels are cationic ion channels with a high fractional Ca2+ current, and they are known to influence cardiac function after MI injury; however, the molecular mechanisms regulating CMF transdifferentiation remain poorly understood. TRPA1 knockout mice, their wild-type littermates, and mice pretreated with the TRPA1 agonist cinnamaldehyde (CA) were subjected to MI injury and monitored for survival, cardiac function, and fibrotic remodeling. TRPA1 can drive myofibroblast transdifferentiation initiated 1 week after MI injury. In addition, we explored the underlying mechanisms via in vitro experiments through gene transfection alone or in combination with inhibitor treatment. TRPA1 overexpression fully activated CMF transformation, while CFs lacking TRPA1 were refractory to transforming growth factor ß- (TGF-ß-) induced transdifferentiation. TGF-ß enhanced TRPA1 expression, which promoted the Ca2+-responsive activation of calcineurin (CaN). Moreover, dual-specificity tyrosine-regulated kinase-1a (DYRK1A) regulated CaN-mediated NFAT nuclear translocation and TRPA1-dependent transdifferentiation. These findings suggest a potential therapeutic role for TRPA1 in the regulation of CMF transdifferentiation in response to MI injury and indicate a comprehensive pathway driving CMF formation in conjunction with TGF-ß, Ca2+ influx, CaN, NFATc3, and DYRK1A.


Subject(s)
Calcineurin/metabolism , Fibroblasts/metabolism , Myocardial Infarction/genetics , Myofibroblasts/metabolism , Animals , Cell Transdifferentiation , Disease Models, Animal , Male , Mice , Myocardial Infarction/pathology , Signal Transduction , Transfection
20.
Mol Med Rep ; 20(2): 1436-1442, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31173198

ABSTRACT

Neointimal hyperplasia could be one of the most important complications after balloon angioplasty. Since calcium signaling has several physiologic effects on the regulation of the proliferation and migration of vascular smooth muscle cells (VSMCs), it was hypothesized that transmembrane protein 66 (TMEM66), a store operated calcium entry (SOCE)­associated regulatory factor, possesses vascular protection against balloon injury. The rat balloon­induced carotid artery injury model was performed. Histological analysis was used to check neointimal hyperplasia. TMEM66 expression was measured by PCR and immunoblotting. The results revealed that TMEM66 was expressed in the medial and neointimal layers of the injured artery, and the expression of TMEM66 was markedly decreased. TMEM66 overexpression attenuated neointimal hyperplasia via VSMC proliferation/migration inhibition, and restored expression of VSMC phenotypic markers. Moreover, TMEM66 overexpression reduced the increased expression of Stim1 and Orai1 and PDGF­BB treatment­enhanced [Ca2+]i. In conclusion, TMEM66 protects against balloon injury­induced neointimal hyperplasia, and may be a pharmacological target for the treatment of restenosis.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Carotid Artery Injuries/genetics , Hyperplasia/genetics , Intracellular Calcium-Sensing Proteins/genetics , Membrane Proteins/metabolism , Neointima/genetics , Angioplasty, Balloon/adverse effects , Animals , Becaplermin/pharmacology , Calcium Signaling , Calcium-Binding Proteins/genetics , Carotid Arteries/metabolism , Carotid Arteries/pathology , Carotid Artery Injuries/etiology , Carotid Artery Injuries/metabolism , Carotid Artery Injuries/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Gene Expression Regulation , Hyperplasia/etiology , Hyperplasia/metabolism , Hyperplasia/pathology , Intracellular Calcium-Sensing Proteins/metabolism , Ion Transport , Male , Membrane Proteins/genetics , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima/etiology , Neointima/metabolism , Neointima/pathology , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL