Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters










Publication year range
1.
Acta Pharm Sin B ; 11(6): 1592-1606, 2021 Jun.
Article in English | MEDLINE | ID: mdl-34221870

ABSTRACT

HYD-PEP06, an endostatin-modified polypeptide, has been shown to produce effective anti-colorectal carcinoma effects through inhibiting epithelial-mesenchymal transition (EMT). However, whether HYD-PEP06 has similar suppressive effect on hepatocellular carcinoma (HCC) remained unknown. In this study, HYD-PEP06 inhibited metastasis and EMT but not proliferation in vitro. Cignal finder pathway reporter array and Western blot analysis revealed that HYD-PEP06 suppressed HCCLM3 cell metastasis and EMT by inhibiting the PI3K/AKT pathway. Moreover, HYD-PEP06 exerted anti-metastasis effects in HepG2 cancer stem-like cells (CSCs) via suppressing the WNT/ß-catenin signaling pathway. Finally, in HCCLM3 tumor-bearing BALB/c nu/nu nude mice, HYD-PEP06 substantially suppressed tumor growth, lung metastasis and HCC progress. Our results suggest that HYD-PEP06 inhibits the metastasis and EMT of HCC and CSCs as well, and thus has the potential as an agent for HCC treatment.

2.
Toxicol Appl Pharmacol ; 410: 115363, 2021 01 01.
Article in English | MEDLINE | ID: mdl-33290780

ABSTRACT

Tongue cancer is one of the most common oral malignancies. Quisinostat is a histone deacetylase inhibitor with antitumor activity. The aim of this study was to evaluate the effects of quisinostat on the viability of tongue squamous cell carcinoma (TSCC) cells (CAL-27, TCA-8113) in vitro and in vivo. Cell viability, cell morphological observation, scratch wound-healing assay, transwell migration assay, transmission electron microscope, flow cytometry and cellular reactive oxygen species were assessed in vitro. The results showed that quisinostat can significantly inhibit the viability, growth and migration of TSCC cells. And quisinostat could significantly induce TSCC cells apoptosis, pyroptosis, and ferroptosis. Quisinostat significantly inhibited tumor tissue growth in animal experiments. Up-regulation of the expression of Bax, cleaved-caspase3, caspase-1, p53, phospho-p53 and down-regulated of the expression of caspase-3, Bcl-2, GPX4 in cell lines and tumor tissues of nude mice were observed by Western blotting analysis. Up-regulation of the expression of caspase-1, Bax, cleaved-caspase3, p53 and down-regulated of the expression of ki67, caspase-3, Bcl-2, GPX4 in tumor tissues of nude mice were observed by immunohistochemistry. TUNEL analysis showed that quisinostat could increase the apoptosis rate in the tumor tissues of nude mice. Up-regulation of the expression of p53 and down-regulated expression of GPX4 in cell lines were observed by immunofluorescent staining, and the expression locations of p53 and GPX4 proteins in TSCC cells were observed. Based on these findings, quisinostat may be a potential drug for the treatment of tongue squamous cell carcinoma.


Subject(s)
Apoptosis/drug effects , Ferroptosis/drug effects , Histone Deacetylase Inhibitors/therapeutic use , Hydroxamic Acids/therapeutic use , Pyroptosis/drug effects , Tongue Neoplasms/drug therapy , Animals , Apoptosis/physiology , Cell Death/drug effects , Cell Death/physiology , Cell Line, Tumor , Dose-Response Relationship, Drug , Ferroptosis/physiology , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Pyroptosis/physiology , Tongue Neoplasms/metabolism , Tongue Neoplasms/pathology , Tumor Burden/drug effects , Tumor Burden/physiology , Xenograft Model Antitumor Assays/methods
3.
J Cell Mol Med ; 24(17): 9545-9559, 2020 09.
Article in English | MEDLINE | ID: mdl-32722882

ABSTRACT

RNF4, a poly-SUMO-specific E3 ubiquitin ligase, is associated with protein degradation, DNA damage repair and tumour progression. However, the effect of RNF4 in cardiomyocytes remains to be explored. Here, we identified the alteration of RNF4 from ischaemic hearts and oxidative stress-induced apoptotic cardiomyocytes. Upon myocardial infarction (MI) or H2 O2 /ATO treatment, RNF4 increased rapidly and then decreased gradually. PML SUMOylation and PML nuclear body (PML-NB) formation first enhanced and then degraded upon oxidative stress. Reactive oxygen species (ROS) inhibitor was able to attenuate the elevation of RNF4 expression and PML SUMOylation. PML overexpression and RNF4 knockdown by small interfering RNA (siRNA) enhanced PML SUMOylation, promoted p53 recruitment and activation and exacerbated H2 O2 /ATO-induced cardiomyocyte apoptosis which could be partially reversed by knockdown of p53. In vivo, knockdown of endogenous RNF4 via in vivo adeno-associated virus infection deteriorated post-MI structure remodelling including more extensive interstitial fibrosis and severely fractured and disordered structure. Furthermore, knockdown of RNF4 worsened ischaemia-induced cardiac dysfunction of MI models. Our results reveal a novel myocardial apoptosis regulation model that is composed of RNF4, PML and p53. The modulation of these proteins may provide a new approach to tackling cardiac ischaemia.


Subject(s)
Apoptosis/genetics , Ischemia/genetics , Myocytes, Cardiac/metabolism , Nuclear Proteins/genetics , Transcription Factors/genetics , Animals , Fibrosis/genetics , Male , Mice , Myocardial Infarction/genetics , Oxidative Stress/genetics , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Sumoylation/genetics , Tumor Suppressor Protein p53/genetics , Ubiquitin-Protein Ligases/genetics
4.
Front Pharmacol ; 11: 123, 2020.
Article in English | MEDLINE | ID: mdl-32153415

ABSTRACT

Arsenic trioxide (ATO) is an effective therapeutic agent against acute promyelocytic leukemia (APL); however, its anti-tumor effect on solid tumors such as colorectal cancer (CRC) is still in debate. Ascorbic acid (AA) also produces a selective cytotoxic activity against tumor cells. Here, we exploit the potential benefit of ATO/AA combination in generating cytotoxicity to CRC cells, which may lay the groundwork for the potential combinational chemotherapy of CRCs. According to the results, we found that ATO and AA effectively inhibited the viability of human CRC cells in a synergistic manner. AA and ATO corporately activated caspase-3 to trigger apoptosis and upregulated the expression of caspase-1 and promoted formation of inflammasomes to induce pyroptosis. Furthermore, the stimulation of reactive oxygen species (ROS) overproduction was demonstrated as a subcellular mechanism for apoptosis and pyroptosis induced by ATO/AA combination treatment. Our findings suggest that ATO combination with a conventional dosage of AA offers an advantage for killing CRC cells. The synergistic action of ATO/AA combination might be considered a plausible strategy for the treatment of CRC and perhaps other solid tumors as well.

5.
Mol Ther Oncolytics ; 16: 86-99, 2020 Mar 27.
Article in English | MEDLINE | ID: mdl-31970286

ABSTRACT

Small ubiquitin-related modifiers (SUMO) represent a class of ubiquitin-like proteins that are conjugated, like ubiquitin, by a set of enzymes to form cellular regulatory proteins, and play key roles in the control of cell proliferation, differentiation, and apoptosis. We found that ginkgolic acid (GA) can significantly reduce cell vitality in a dose- and time-dependent manner and can also accelerate cyto-apoptosis in both Tca8113 and Cal-27 cells. Migration and wound-healing assays were executed to determine the anti-migration effect of GA in oral squamous cell carcinoma (OSCC) cell lines. GA represses transforming growth factor-ß1 (TGF-ß1)-induced epithelial-mesenchymal transition (EMT) markers in OSCC cell lines. This investigation is the first evidence that GA suppresses TGF-ß1-induced SUMOylation of SMAD4. We show that GA affects the phosphorylation of SMAD2/3 protein and the release of SMAD4. In the xenograft mouse model, the OSCC progression was reduced by GA, effectively suppressing the growth of tumors. In addition, siSMAD4 improved cell migration and viability, which was inhibited by GA in Tca8113 cells. GA suppresses tumorigenicity and tumor progression of OSCC through inhibition of TGF-ß1-induced enhancement of SUMOylation of SMAD4. Thus, GA could be a promising therapeutic for OSCC.

6.
J Cell Physiol ; 234(5): 6263-6273, 2019 05.
Article in English | MEDLINE | ID: mdl-30246389

ABSTRACT

Transforming growth factor-ß (TGF-ß) signaling pathway is involved in fibrosis in most, if not all forms of cardiac diseases. Here, we evaluate a positive feedback signaling the loop of TGF-ß1/promyelocytic leukemia (PML) SUMOylation/Pin1 promoting the cardiac fibrosis. To test this hypothesis, the mice underwent transverse aortic constriction (3 weeks) were developed and the morphological evidence showed obvious interstitial fibrosis with TGF-ß1, Pin1 upregulation, and increase in PML SUMOylation. In neonatal mouse cardiac fibroblasts (NMCFs), we found that exogenous TGF-ß1 induced the upregulation of TGF-ß1 itself in a time- and dose-dependent manner, and also triggered the PML SUMOylation and the formation of PML nuclear bodies (PML-NBs), and consequently recruited Pin1 into nuclear to colocalize with PML. Pharmacological inhibition of TGF-ß signal or Pin1 with LY364947 (3 µM) or Juglone (3 µM), the TGF-ß1-induced PML SUMOylation was reduced significantly with downregulation of the messenger RNA and protein for TGF-ß1 and Pin1. To verify the cellular function of PML by means of gain- or loss-of-function, the positive feedback signaling loop was enhanced or declined, meanwhile, TGF-ß-Smad signaling pathway was activated or weakened, respectively. In summary, we uncovered a novel reciprocal loop of TGF-ß1/PML SUMOylation/Pin1 leading to myocardial fibrosis.


Subject(s)
Myocardium/pathology , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Promyelocytic Leukemia Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Feedback, Physiological , Fibrosis , Heart , Heart Diseases/metabolism , Heart Diseases/pathology , Mice , Sumoylation
7.
J Cell Physiol ; 233(12): 9575-9583, 2018 12.
Article in English | MEDLINE | ID: mdl-29943817

ABSTRACT

The epithelial-mesenchymal transition (EMT) is a key event associated with metastasis and dissemination in breast tumor pathogenesis. Promyelocytic leukemia (PML) gene produces several isoforms due to alternative splicing; however, the biological function of each specific isoform has yet to be identified. In this study, we report a previously unknown role for PMLIV, the most intensely studied nuclear isoform, in transforming growth factor-ß (TGF-ß) signaling-associated EMT and migration in breast cancer. This study demonstrates that PMLIV overexpression promotes a more aggressive mesenchymal phenotype and increases the migration of MCF-7 cancer cells. This event is associated with activation of the TGF-ß canonical signaling pathway through the induction of Smad2/3 phosphorylation and the translocation of phospho-Smad2/3 to the nucleus. In this study, we report a previously unknown role for PMLIV in TGF-ß signaling-induced regulation of breast cancer-associated EMT and migration. Targeting this pathway may be therapeutically beneficial.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Epithelial-Mesenchymal Transition , Promyelocytic Leukemia Protein/metabolism , Transforming Growth Factor beta/metabolism , Cell Nucleus/metabolism , Female , HEK293 Cells , Humans , MCF-7 Cells , Models, Biological , Phosphorylation , Promyelocytic Leukemia Protein/chemistry , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Signal Transduction , Smad2 Protein/metabolism , Smad3 Protein/metabolism
8.
Br J Pharmacol ; 175(15): 3111-3130, 2018 08.
Article in English | MEDLINE | ID: mdl-29722931

ABSTRACT

BACKGROUND AND PURPOSE: PEP06, a polypeptide modified from endostatin, was investigated for its antitumour effects on colorectal cancer (CRC) and the possible mechanisms of this antitumour activity were examined in in vitro and in vivo models. EXPERIMENTAL APPROACH: After PEP06 treatment, cell proliferation and migration assays were performed in CRC cells. Epithelial-mesenchymal transition (EMT) progression was determined by Western blotting, immunofluorescent staining and immunohistochemistry in vitro and in a residual xenograft model. MiRNAs regulated by PEP06 were identified by miRNA microarray and verified by in situ hybridization and quantitative real-time PCR. The interactions between PEP06 and integrin αvß3 were determined with Biacore SA biochips. The cellular function of miR-146b-5p was validated by gain-of-function and loss-of-function approaches. A mouse model of lung metastasis was used to determine the effect of PEP06 on metastatic growth. KEY RESULTS: PEP06 did not affect cell viability but reduced migration and EMT in SW620 and HCT116 cells. PEP06 significantly repressed the expression of miR-146b-5p in these two cell lines through binding to integrin αvß3. MiR-146b-5p was shown to increase EMT by targeting Smad4, and the miR-146b-5p-Smad4 cascade regulated EMT in CRC. PEP06 also suppressed CRC pulmonary metastasis, increased survival of mice and hampered residual tumour growth by inhibiting EMT through down-regulating miR-146b-5p. CONCLUSIONS AND IMPLICATIONS: PEP06 is a polypeptide that inhibits the growth and metastasis of colon cancer through its RGD motif binding to integrin αvß3, thereby down-regulating miR-146b-5p to inhibit EMT in vitro and in vivo. It might have potential as a therapeutic for CRC.


Subject(s)
Antineoplastic Agents/therapeutic use , Colorectal Neoplasms/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Animals , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Endostatins , Female , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/physiology , Humans , Lung Neoplasms/secondary , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Peptide Fragments , Vimentin/metabolism , Wound Healing/drug effects
9.
Toxicol Appl Pharmacol ; 345: 1-9, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29524504

ABSTRACT

BACKGROUND AND PURPOSE: Protein modification by small ubiquitin-like modifier (SUMO) plays a critical role in the pathogenesis of heart diseases. The present study was designed to determine whether ginkgolic acid (GA) as a SUMO-1 inhibitor exerts an inhibitory effect on cardiac fibrosis induced by myocardial infarction (MI). EXPERIMENTAL APPROACH: GA was delivered by osmotic pumps in MI mice. Masson staining, electron microscopy (EM) and echocardiography were used to assess cardiac fibrosis, ultrastructure and function. Expression of SUMO-1, PML, TGF-ß1 and Pin1 was measured with Western blot or Real-time PCR. Collagen content, cell viability and myofibroblast transformation were measured in neonatal mouse cardiac fibroblasts (NMCFs). Promyelocytic leukemia (PML) protein was over-expressed by plasmid transfection. KEY RESULTS: GA improved cardiac fibrosis and dysfunction, and decreased SUMO-1 expression in MI mice. GA (>20 µM) inhibited NMCF viability in a dose-dependent manner. Nontoxic GA (10 µM) restrained angiotensin II (Ang II)-induced myofibroblast transformation and collagen production. GA also inhibited expression of TGF-ß1 mRNA and protein in vitro and in vivo. GA suppressed PML SUMOylation and PML nuclear body (PML-NB) organization, and disrupted expression and recruitment of Pin1 (a positive regulator of TGF-ß1 mRNA), whereas over-expression of PML reversed that. CONCLUSIONS AND IMPLICATIONS: Inhibition of SUMO-1 by GA alleviated MI-induced heart dysfunction and fibrosis, and the SUMOylated PML/Pin1/TGF-ß1 pathway is crucial for GA-inhibited cardiac fibrosis.


Subject(s)
Myocardial Infarction/drug therapy , SUMO-1 Protein/antagonists & inhibitors , Salicylates/therapeutic use , Animals , Animals, Newborn , Cell Survival/drug effects , Cell Survival/physiology , Dose-Response Relationship, Drug , Fibrosis/drug therapy , Fibrosis/metabolism , Fibrosis/pathology , Male , Mice , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , SUMO-1 Protein/metabolism , Salicylates/pharmacology , Stroke Volume/drug effects , Stroke Volume/physiology
10.
J Am Heart Assoc ; 6(6)2017 May 30.
Article in English | MEDLINE | ID: mdl-28559372

ABSTRACT

BACKGROUND: Myocardial infarction (MI) is often accompanied by cardiomyocyte apoptosis, which decreases heart function and leads to an increased risk of heart failure. The aim of this study was to examine the effects of transforming growth factor-ß receptor III (TGFßR3) on cardiomyocyte apoptosis during MI. METHODS AND RESULTS: An MI mouse model was established by left anterior descending coronary artery ligation. Cell viability, apoptosis, TGFßR3, and mitogen-activated protein kinase signaling were assessed by methylthiazolyldiphenyl-tetrazolium bromide assay, terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling assay, immunofluorescence, electron microscopy, and Western blotting. Our results demonstrated that TGFßR3 expression in the border region of the heart was dynamically changed during MI. After stimulation with H2O2, TGFßR3 overexpression in cardiomyocytes led to increased cell apoptosis and activation of p38 signaling, whereas TGFßR3 knockdown had the opposite effect. ERK1/2 and JNK1/2 signaling was not altered by TGFßR3 modulation, and p38 inhibitor (SB203580) reduced the effect of TGFßR3 on apoptosis, suggesting that p38 has a nonredundant function in activating apoptosis. Consistent with the in vitro observations, cardiac TGFßR3 transgenic mice showed augmented cardiomyocyte apoptosis, enlarged infarct size, increased injury, and enhanced p38 signaling upon MI. Conversely, cardiac loss of function of TGFßR3 by adeno-associated viral vector serotype 9-TGFßR3 short hairpin RNA attenuated the effects of MI in mice. CONCLUSIONS: TGFßR3 promotes apoptosis of cardiomyocytes via a p38 pathway-associated mechanism, and loss of TGFßR3 reduces MI injury, which suggests that TGFßR3 may serve as a novel therapeutic target for MI.


Subject(s)
Apoptosis , Myocardial Infarction/metabolism , Myocytes, Cardiac/metabolism , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Animals , Apoptosis/drug effects , Cells, Cultured , Disease Models, Animal , Hydrogen Peroxide/pharmacology , Male , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Proteoglycans/genetics , RNA Interference , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Time Factors , Transfection , p38 Mitogen-Activated Protein Kinases/metabolism
11.
Oncotarget ; 8(28): 45447-45458, 2017 Jul 11.
Article in English | MEDLINE | ID: mdl-28525371

ABSTRACT

The human ether-a-go-go-related gene (HERG) channel is a novel target for the treatment of drug-induced long QT syndrome, which causes lethal cardiotoxicity. This study is designed to explore the possible role of PML SUMOylation and its associated nuclear bodies (NBs) in the regulation of HERG protein expression. Both arsenic trioxide (ATO) and angiotensin II (Ang II) were able to significantly reduce HERG protein expression, while also increasing PML SUMOylation and accelerating the formation of PML-NBs. Pre-exposure of cardiomyocytes to a SUMOylation chemical inhibitor, ginkgolic acid, or the silencing of UBC9 suppressed PML SUMOylation, subsequently preventing the downregulation of HERG induced by ATO or Ang II. Conversely, knockdown of RNF4 led to a remarkable increase in PML SUMOylation and the function of PML-NBs, further promoting ATO- or Ang II-induced HERG protein downregulation. Mechanistically, an increase in PML SUMOylation by ATO or Ang II dramatically enhanced the formation of PML and Pin1 complexes in PML-NBs, leading to the upregulation of TGF-ß1 protein, eventually inhibiting HERG expression through activation of protein kinase A. The present work uncovered a novel molecular mechanism underlying HERG protein expression and indicated that PML SUMOylation is a critical step in the development of drug-acquired arrhythmia.


Subject(s)
Angiotensin II/pharmacology , Arsenicals/pharmacology , ERG1 Potassium Channel/metabolism , Oxides/pharmacology , Animals , Arsenic Trioxide , Cyclic AMP-Dependent Protein Kinases/metabolism , ERG1 Potassium Channel/genetics , Gene Expression Regulation/drug effects , Gene Knockdown Techniques , Humans , Intranuclear Inclusion Bodies/metabolism , Mice , Models, Biological , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , NIMA-Interacting Peptidylprolyl Isomerase/genetics , NIMA-Interacting Peptidylprolyl Isomerase/metabolism , Promyelocytic Leukemia Protein/metabolism , Sumoylation/drug effects , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism
12.
Mol Ther ; 25(3): 666-678, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28143738

ABSTRACT

The promyelocytic leukemia protein (PML) is essential in the assembly of dynamic subnuclear structures called PML nuclear bodies (PML-NBs), which are involved in regulating diverse cellular functions. However, the possibility of PML being involved in cardiac disease has not been examined. In mice undergoing transverse aortic constriction (TAC) and arsenic trioxide (ATO) injection, transforming growth factor ß1 (TGF-ß1) was upregulated along with dynamic alteration of PML SUMOylation. In cultured neonatal mouse cardiac fibroblasts (NMCFs), ATO, angiotensin II (Ang II), and fetal bovine serum (FBS) significantly triggered PML SUMOylation and the assembly of PML-NBs. Inhibition of SUMOylated PML by silencing UBC9, the unique SUMO E2-conjugating enzyme, reduced the development of cardiac fibrosis and partially improved cardiac function in TAC mice. In contrast, enhancing SUMOylated PML accumulation, by silencing RNF4, a poly-SUMO-specific E3 ubiquitin ligase, accelerated the induction of cardiac fibrosis and promoted cardiac function injury. PML colocalized with Pin1 (a positive regulator for TGF-ß1 mRNA expression in PML-NBs) and increased TGF-ß1 activity. These findings suggest that the UBC9/PML/RNF4 axis plays a critical role as an important SUMO pathway in cardiac fibrosis. Modulating the protein levels of the pathway provides an attractive therapeutic target for the treatment of cardiac fibrosis and heart failure.


Subject(s)
Gene Silencing , Myocardium/metabolism , Myocardium/pathology , Nuclear Proteins/genetics , Promyelocytic Leukemia Protein/metabolism , Transcription Factors/genetics , Ubiquitin-Conjugating Enzymes/genetics , Angiotensin II/pharmacology , Animals , Arsenic Trioxide , Arsenicals/pharmacology , Collagen/biosynthesis , Fibrosis , Mice , Myofibroblasts/drug effects , Myofibroblasts/metabolism , Oxides/pharmacology , Protein Binding , Sumoylation , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Ubiquitin-Protein Ligases
13.
Cell Physiol Biochem ; 41(2): 835-848, 2017.
Article in English | MEDLINE | ID: mdl-28214884

ABSTRACT

BACKGROUND/AIMS: The present study investigated whether the transient receptor potential melastatin 4 (TRPM4) channel plays a role in high salt diet (HSD)-induced endothelial injuries. METHODS: Western blotting and immunofluorescence were used to examine TRPM4 expression in the mesenteric endothelium of Dahl salt-sensitive (SS) rats fed a HSD. The MTT, TUNEL, and transwell assays were used to evaluate the cell viability, cell apoptosis, and cell migration, respectively, of human umbilical vein endothelial cells (HUVECs). Enzyme-linked immunosorbent assays were used to determine the concentrations of intercellular adhesion molecule 1 (ICAM-1), vascular cell adhesion protein 1 (VCAM-1), and E-selectin. Carboxy-H2DCFDA, a membrane-permeable reactive oxygen species (ROS)-sensitive fluorescent probe, was used to detect intracellular ROS levels. RESULTS: TRPM4 was mainly expressed near the plasma membrane of mesenteric artery endothelial cells, and its expression level increased in SS hypertensive rats fed a HSD. Its protein expression was significantly upregulated upon treatment with exogenous hydrogen peroxide (H2O2) and aldosterone in cultured HUVECs. Cell viability decreased upon treatment with both agents in a concentration-dependent manner, which could be partially reversed by 9-phenanthrol, a specific TRPM4 inhibitor. Exogenous H2O2 induced apoptosis, enhanced cell migration, and increased the release of adhesion molecules, including ICAM-1, VCAM-1, and E-selectin, all of which were significantly attenuated upon treatment with 9-phenanthrol. Aldosterone and H2O2 induced the accumulation of intracellular ROS, which was significantly inhibited by 9-phenanthrol, suggesting that oxidative stress is one of the mechanisms underlying aldosterone-induced endothelial injury. CONCLUSIONS: Given the fact that oxidative stress and high levels of circulating aldosterone are present in hypertensive patients, we suggest that the upregulation of TRPM4 in the vascular endothelium may be involved in endothelial injuries caused by these stimuli.


Subject(s)
Diet , Endothelium, Vascular/metabolism , TRPM Cation Channels/metabolism , Aldosterone/toxicity , Animals , Apoptosis/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Cells, Cultured , E-Selectin/analysis , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Hydrogen Peroxide/toxicity , Male , Mesenteric Arteries/cytology , Oxidative Stress/drug effects , Phenanthrenes/pharmacology , RNA, Small Interfering/metabolism , Rats , Rats, Inbred Dahl , Sodium Chloride/pharmacology , TRPM Cation Channels/antagonists & inhibitors , TRPM Cation Channels/genetics , Up-Regulation/drug effects
14.
Sci Rep ; 7: 42657, 2017 02 15.
Article in English | MEDLINE | ID: mdl-28198439

ABSTRACT

A long non-coding RNA (lncRNA), named myocardial infarction associated transcript (MIAT), has been documented to confer risk of myocardial infarction (MI). The aim of this study is to elucidate the pathophysiological role of MIAT in regulation of cardiac fibrosis. In a mouse model of MI, we found that MIAT was remarkably up-regulated, which was accompanied by cardiac interstitial fibrosis. MIAT up-regulation in MI was accompanied by deregulation of some fibrosis-related regulators: down-regulation of miR-24 and up-regulation of Furin and TGF-ß1. Most notably, knockdown of endogenous MIAT by its siRNA reduced cardiac fibrosis and improved cardiac function and restored the deregulated expression of the fibrosis-related regulators. In cardiac fibroblasts treated with serum or angiotensin II, similar up-regulation of MIAT and down-regulation of miR-24 were consistently observed. These changes promoted fibroblasts proliferation and collagen accumulation, whereas knockdown of MIAT by siRNA or overexpression of miR-24 with its mimic abrogated the fibrogenesis. Our study therefore has identified MIAT as the first pro-fibrotic lncRNA in heart and unraveled the role of MIAT in the pathogenesis of MI. These findings also promise that normalization of MIAT level may prove to be a therapeutic option for the treatment of MI-induced cardiac fibrosis and the associated cardiac dysfunction.


Subject(s)
Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardium/metabolism , Myocardium/pathology , RNA, Long Noncoding/genetics , Animals , Cell Proliferation , Cell Survival , Collagen/metabolism , Disease Models, Animal , Echocardiography , Fibroblasts/metabolism , Fibrosis , Gene Expression Regulation , Gene Knockdown Techniques , Heart Function Tests , Male , Mice , MicroRNAs , Myocardial Infarction/diagnosis , Myocardial Infarction/physiopathology , RNA, Small Interfering/genetics
15.
Cell Physiol Biochem ; 39(3): 827-36, 2016.
Article in English | MEDLINE | ID: mdl-27497782

ABSTRACT

BACKGROUND/AIMS: Deregulated myocardial fibrosis is associated with a wide spectrum of cardiac conditions, being considered one of the major causes for heart disease. Our study was designed to investigate the role of microRNA-328 (miR-328) in regulating cardiac fibrosis. METHODS: We induced cardiac fibrosis following MI by occlusion of the left coronary artery in C57BL/6 mice. Real-time PCR was employed to evaluate the level of miR-328. Masson's Trichrome stain was used to evaluate the development of fibrosis. Luciferase activity assay was performed to confirm the miRNA's binding site in the TGFßRIII gene. Western blot analysis was used to examine TGFßRIII, p-smad2/3 and TGF-ß1 at protein level. RESULTS: In this study, we found that miR-328 was significantly upregulated in the border zone of infarcted myocardium of wild type (WT) mice; TGFßRIII was downregulated whereas TGF-ß1 was upregulated along with increased cardiac fibrosis. And miR-328 stimulated TGF-ß1 signaling and promoted collagen production in cultured fibroblasts. We further found that the pro-fibrotic effect of miR-328 was mediated by targeting TGFßRIII. Additionally, cardiac fibrosis was significantly reduced in infarcted heart when treated with miR-328 antisense. CONCLUSIONS: These data suggest that miR-328 is a potent pro-fibrotic miRNA and an important determinant of cardiac fibrosis in diseased heart.


Subject(s)
Fibroblasts/metabolism , MicroRNAs/genetics , Myocardial Infarction/genetics , Myocardium/metabolism , Proteoglycans/genetics , Receptors, Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1/genetics , Animals , Collagen/genetics , Collagen/metabolism , Coronary Occlusion/pathology , Coronary Occlusion/surgery , Coronary Vessels/pathology , Coronary Vessels/surgery , Fibroblasts/pathology , Fibrosis , Gene Expression Regulation , Genes, Reporter , Luciferases/genetics , Luciferases/metabolism , Male , Mice , Mice, Inbred C57BL , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardium/pathology , Oligonucleotides, Antisense/genetics , Oligonucleotides, Antisense/metabolism , Primary Cell Culture , Proteoglycans/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction , Smad2 Protein/genetics , Smad2 Protein/metabolism , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism
16.
Hypertension ; 68(3): 654-66, 2016 09.
Article in English | MEDLINE | ID: mdl-27432858

ABSTRACT

The role of type III transforming growth factor-ß receptor (TßRIII) in the pathogenesis of heart diseases remains largely unclear. Here, we investigated the functional role and molecular mechanisms of TßRIII in the development of myocardial hypertrophy. Western blot and quantitative real time-polymerase chain reaction analyses revealed that the expression of TßRIII was significantly elevated in human cardiac hypertrophic samples. Consistently, TßRIII expression was substantially increased in transverse aortic constriction (TAC)- and isoproterenol-induced mouse cardiac hypertrophy in vivo and in isoproterenol-induced cardiomyocyte hypertrophy in vitro. Overexpression of TßRIII resulted in cardiomyocyte hypertrophy, whereas isoproterenol-induced cardiomyocyte hypertrophy was greatly attenuated by knockdown of TßRIII in vitro. Cardiac-specific transgenic expression of TßRIII independently led to cardiac hypertrophy in mice, which was further aggravated by isoproterenol and TAC treatment. Cardiac contractile function of the mice was not altered in TßRIII transgenic mice; however, TAC led to significantly decreased cardiac contractile function in TßRIII transgenic mice compared with control mice. Conversely, isoproterenol- and TAC-induced cardiac hypertrophy and TAC-induced cardiac contractile function impairment were partially reversed by suppression of TßRIII in vivo. Our data suggest that TßRIII mediates stress-induced cardiac hypertrophy through activation of Ca(2+)/calmodulin-dependent protein kinase II, which requires a physical interaction of ß-arrestin2 with both TßRIII and calmodulin-dependent protein kinase II. Our findings indicate that stress-induced increase in TßRIII expression results in cardiac hypertrophy through ß-arrestin2-dependent activation of calmodulin-dependent protein kinase II and that transforming growth factor-ß and ß-adrenergic receptor signaling are not involved in spontaneous cardiac hypertrophy in cardiac-specific transgenic expression of TßRIII mice. Our findings may provide a novel target for control of myocardial hypertrophy.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cardiomegaly/genetics , Transforming Growth Factor beta/metabolism , beta-Arrestin 2/metabolism , Analysis of Variance , Animals , Biopsy, Needle , Cardiomegaly/pathology , Cells, Cultured , Disease Models, Animal , Humans , Immunohistochemistry , Isoproterenol/pharmacology , Mice , Mice, Transgenic , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Random Allocation , Sensitivity and Specificity , T-Box Domain Proteins/genetics
17.
Br J Pharmacol ; 172(15): 3779-92, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25884615

ABSTRACT

BACKGROUND AND PURPOSE: Statins decrease heart disease risk, but their mechanisms are not completely understood. We examined the role of the TGF-ß receptor III (TGFBR3) in the inhibition of cardiac fibrosis by simvastatin. EXPERIMENTAL APPROACH: Myocardial infarction (MI) was induced by ligation of the left anterior descending coronary artery in mice given simvastatin orally for 7 days. Cardiac fibrosis was measured by Masson staining and electron microscopy. Heart function was evaluated by echocardiography. Signalling through TGFBR3, ERK1/2, JNK and p38 pathways was measured using Western blotting. Collagen content and cell viability were measured in cultures of neonatal mouse cardiac fibroblasts (NMCFs). Interactions between TGFBR3 and the scaffolding protein, GAIP-interacting protein C-terminus (GIPC) were detected using co-immunoprecipitation (co-IP). In vivo, hearts were injected with lentivirus carrying shRNA for TGFBR3. KEY RESULTS: Simvastatin prevented fibrosis following MI, improved heart ultrastructure and function, up-regulated TGFBR3 and decreased ERK1/2 and JNK phosphorylation. Simvastatin up-regulated TGFBR3 in NMCFs, whereas silencing TGFBR3 reversed inhibitory effects of simvastatin on cell proliferation and collagen production. Simvastatin inhibited ERK1/2 and JNK signalling while silencing TGFBR3 opposed this effect. Co-IP demonstrated TGFBR3 binding to GIPC. Overexpressing TGFBR3 inhibited ERK1/2 and JNK signalling which was abolished by knock-down of GIPC. In vivo, suppression of cardiac TGFBR3 abolished anti-fibrotic effects, improvement of cardiac function and changes in related proteins after simvastatin. CONCLUSIONS AND IMPLICATIONS: TGFBR3 mediated the decreased cardiac fibrosis, collagen deposition and fibroblast activity, induced by simvastatin, following MI. These effects involved GIPC inhibition of the ERK1/2/JNK pathway.


Subject(s)
Fibrosis/drug therapy , Myocardial Infarction/drug therapy , Proteoglycans/biosynthesis , Receptors, Transforming Growth Factor beta/biosynthesis , Simvastatin/therapeutic use , Up-Regulation/drug effects , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Echocardiography , Fibrosis/pathology , Gene Knockdown Techniques , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , MAP Kinase Signaling System/drug effects , Male , Mice , Myocardial Infarction/pathology , Simvastatin/pharmacology
18.
J Cell Mol Med ; 18(7): 1334-43, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24758696

ABSTRACT

Beta-adrenoceptor (ß-AR) exerts critical regulation of cardiac function. MicroRNAs (miRNAs) are potentially involved in a variety of biological and pathological processes. This study aimed to investigate the role of miRNA let-7e in the up-regulation of ß(1) -AR and arrhythmogenesis in acute myocardial infarction (AMI) in rats. ß(1) -AR expression was significantly up-regulated and let-7a, c, d, e and i were markedly down-regulated in the infarcted heart after 6 and 24 hrs myocardial infarction. Forced expression of let-7e suppressed ß(1) -AR expression at the protein level, without affecting ß(1) -AR mRNA level, in neonatal rat ventricular cells (NRVCs). Silencing of let-7e by let-7e antisense inhibitor (AMO-let-7e) enhanced ß(1) -AR expression at the protein level in NRVCs. Administration of the lentivirus vector containing precursor let-7e (len-pre-let-7e) significantly inhibited ß(1) -AR expression in rats, whereas len-AMO-let-7e up-regulated ß(1) -AR relative to the baseline control level, presumably as a result of depression of tonic inhibition of ß(1) -AR by endogenous let-7e. Len-negative control (len-NC) did not produce significant influence on ß(1) -AR expression. Len-pre-let-7e also profoundly reduced the up-regulation of ß(1) -AR induced by AMI and this effect was abolished by len-AMO-let-7e. Importantly, len-pre-let-7e application significantly reduced arrhythmia incidence after AMI in rats and its anti-arrhythmic effect was cancelled by len-AMO-let-7e. Notably, anti-arrhythmic efficacy of len-pre-let-7e was similar to propranolol, a non-selective ß-AR blocker and metoprolol, a selective ß(1) -AR blocker. Down-regulation of let-7e contributes to the adverse increase in ß(1) -AR expression in AMI and let-7e supplement may be a new therapeutic approach for preventing adverse ß(1) -AR up-regulation and treating AMI-induced arrhythmia.


Subject(s)
Anti-Arrhythmia Agents , Arrhythmias, Cardiac/prevention & control , MicroRNAs/genetics , Myocardial Infarction/prevention & control , Myocytes, Cardiac/metabolism , Receptors, Adrenergic, beta-1/chemistry , 3' Untranslated Regions/genetics , Animals , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/pathology , Biomarkers/analysis , Blotting, Western , Cells, Cultured , Gene Expression Profiling , Gene Expression Regulation , Heart Rate , Male , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , RNA, Messenger/genetics , Rats , Rats, Wistar , Real-Time Polymerase Chain Reaction , Receptors, Adrenergic, beta-1/genetics , Receptors, Adrenergic, beta-1/metabolism , Reverse Transcriptase Polymerase Chain Reaction
19.
Eur J Pharmacol ; 735: 202-10, 2014 Jul 15.
Article in English | MEDLINE | ID: mdl-24769415

ABSTRACT

Cisapride has been shown to have electrophysiological effects on the heart. The aim of this study was to investigate whether cisapride has effects on cardiac hypertrophy. Rat and cellular models of cardiac hypertrophy were used in this study. Cell surface area (CSA), mRNA and protein expression were used to evaluate cardiac hypertrophy. Cardiac function was measured by echocardiography. Cisapride attenuated ISO-induced increase in CSA in a dose-dependent manner in cultured neonatal rat cardiomyocytes. A significant anti-hypertrophic effect was achieved by cisapride 0.01µM (P<0.05). Cisapride repressed the increased mRNA levels of ANP, BNP, ß-MHC in ISO-treated cells (P<0.05). However, mallotoxin or GR113808 did not influence anti-hypertrophic effects of cisapride. In addition, cisapride inhibited the increase of intracellular Ca(2+) ([Ca(2+)]i) and the upregulation of protein levels of calcineurin and NFATc-3 (P<0.05) as well as prevented the downregulation of p-NFATc-3 (P<0.01) induced by ISO. Consistently, cisapride (0.5mg/kg/day) produced inhibitory effects on cardiac hypertrophy, including the suppression of ANP, BNP, ß-MHC, calcineurin, and NFATc-3; elevation of p-NFATc-3; reduction of cross-sectional area of cardiomyocytes in rat heart; and restoration of cardiac dysfunction by improving left ventricular diastolic and systolic performance. Importantly, cisapride 0.5 and 5.0mg/kg/day did not cause prolongation of QT and QTc intervals in rats. In conclusion, cisapride possesses a prominent anti-hypertrophic property which is likely to be conferred by its ability to downregulate Ca(2+)/calcineurin/NFAT and the present data provide new insight into this drug action.


Subject(s)
Calcineurin/metabolism , Cardiomegaly/metabolism , Cardiotonic Agents/pharmacology , Cisapride/pharmacology , NFATC Transcription Factors/metabolism , Animals , Cardiomegaly/drug therapy , Cardiomegaly/physiopathology , Cardiotonic Agents/therapeutic use , Cisapride/therapeutic use , Male , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Sprague-Dawley , Up-Regulation/drug effects
20.
J Cardiovasc Med (Hagerstown) ; 15(7): 579-86, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24583668

ABSTRACT

OBJECTIVES: A great number of studies indicate that cardiac fibroblasts are essential for maintaining the structure and function of heart. Hypoxia-inducible factor-1 alpha (HIF-1α) is a central transcriptional regulator of hypoxic response. The present study examined whether over-expression of HIF-1α could prevent hypoxia-induced injury in neonatal rat cardiac fibroblasts and, if so, its possible molecular targets. METHODS: Western blotting was used to detect protein level. MTT, electron microscopy, TUNEL staining and confocal microscopy were used to identify cell viability, cell apoptosis and intracellular calcium ([Ca]i) in cardiac fibroblasts, respectively. RESULTS: When cardiac fibroblasts were exposed to hypoxia, HIF-1α protein in nuclei was transiently accumulated at 1 h, and then gradually degraded within 24 h of hypoxia exposure. Over-expression of HIF-1α enhanced nucleus expression of HIF-1α in cardiac fibroblasts, and significantly abolished the decrease of cell viability and cell apoptosis caused by 24-h hypoxia. Accordingly, hypoxia-induced Bax up-regulation, Bcl-2 down-regulation, caspase-3 activation and overload of [Ca]i in cardiac fibroblasts were reversed by HIF-1α over-expression, but were promoted by 30 µmol/l SC205346, a specific HIF-1α blocker. CONCLUSIONS: Our results indicate that HIF-1α may act as a protective factor in the apoptotic process of cardiac fibroblasts and represent a potential therapeutic target for heart remodeling after hypoxia injury.


Subject(s)
Apoptosis/genetics , DNA/genetics , Fibroblasts/ultrastructure , Gene Expression Regulation , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Myocardial Ischemia/genetics , Myocytes, Cardiac/ultrastructure , Animals , Blotting, Western , Calcium/metabolism , Caspase 3/metabolism , Cell Survival , Enzyme-Linked Immunosorbent Assay , Fibroblasts/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , In Situ Nick-End Labeling , Microscopy, Confocal , Microscopy, Electron , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocytes, Cardiac/metabolism , Rats , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...