Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Cell Mol Life Sci ; 80(9): 267, 2023 Aug 26.
Article in English | MEDLINE | ID: mdl-37626241

ABSTRACT

Previous studies show a woman's pregnancy is correlated with post-reproductive longevity, and nulliparity is associated with higher risk of incident heart failure, suggesting pregnancy likely exerts a cardioprotection. We previously reported a cardioprotective phenomenon termed myocardial hypertrophic preconditioning, but it is unknown whether pregnancy-induced physiological hypertrophic preconditioning (PHP) can also protect the heart against subsequent pathological hypertrophic stress. We aimed to clarify the phenomenon of PHP and its mechanisms. The pluripara mice whose pregnancy-induced physiological hypertrophy regressed and the nulliparous mice underwent angiotensin II (Ang II) infusion or transverse aortic constriction (TAC). Echocardiography, invasive left ventricular hemodynamic measurement and histological analysis were used to evaluate cardiac remodeling and function. Silencing or overexpression of Foxo3 by adeno-associated virus was used to investigate the role of FoxO3a involved in the antihypertrophic effect. Compared with nulliparous mice, pathological cardiac hypertrophy induced by Ang II infusion, or TAC was significantly attenuated and heart failure induced by TAC was markedly improved in mice with PHP. Activation of FoxO3a was significantly enhanced in the hearts of postpartum mice. FoxO3a inhibited myocardial hypertrophy by suppressing signaling pathway of phosphorylated glycogen synthase kinase-3ß (p-GSK3ß)/ß-catenin/Cyclin D1. Silencing or overexpression of Foxo3 attenuated or enhanced the anti-hypertrophic effect of PHP in mice with pathological stimulation. Our findings demonstrate that PHP confers resistance to subsequent hypertrophic stress and slows progression to heart failure through activation of FoxO3a/GSK3ß pathway.


Subject(s)
Aortic Valve Stenosis , Heart Failure , Peptide Hormones , Animals , Female , Mice , Pregnancy , Angiotensin II , Cardiomegaly/genetics , Glycogen Synthase Kinase 3 beta/genetics , Heart
2.
Front Cell Infect Microbiol ; 13: 1038472, 2023.
Article in English | MEDLINE | ID: mdl-37033478

ABSTRACT

Background: The imbalance of gut microbiota (GM) is associated with a higher risk of thrombosis in patients with atrial fibrillation (AF). Oral anticoagulants (OACs) have been found to significantly reduce the risk of thromboembolism and increase the risk of bleeding. However, the OAC-induced alterations in gut microbiota in patients with AF remain elusive. Methods: In this study, the microbial composition in 42 AF patients who received long-term OAC treatment (AF-OAC group), 47 AF patients who did not (AF group), and 40 volunteers with the risk of AF (control group) were analyzed by 16S rRNA gene sequencing of fecal bacterial DNA. The metagenomic functional prediction of major bacterial taxa was performed using the Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) software package. Results: The gut microbiota differed between the AF-OAC and AF groups. The abundance of Bifidobacterium and Lactobacillus decreased in the two disease groups at the genus level, but OACs treatment mitigated the decreasing tendency and increased beneficial bacterial genera, such as Megamonas. In addition, OACs reduced the abundance of pro-inflammatory taxa on the genus Ruminococcus but increased certain potential pathogenic taxa, such as genera Streptococcus, Escherichia-Shigella, and Klebsiella. The Subgroup Linear discriminant analysis effect size (LEfSe) analyses revealed that Bacteroidetes, Brucella, and Ochrobactrum were more abundant in the anticoagulated bleeding AF patients, Akkermansia and Faecalibacterium were more abundant in the non-anticoagulated-bleeding-AF patients. The neutrophil-to-lymphocyte ratio (NLR) was lower in the AF-OAC group compared with the AF group (P < 0.05). Ruminococcus was positively correlated with the NLR and negatively correlated with the CHA2DS2-VASc score (P < 0.05), and the OACs-enriched species (Megamonas and Actinobacteria) was positively correlated with the prothrombin time (PT) (P < 0.05). Ruminococcus and Roseburia were negatively associated with bleeding events (P < 0.05). Conclusions: Our study suggested that OACs might benefit AF patients by reducing the inflammatory response and modulating the composition and abundance of gut microbiota. In particular, OACs increased the abundance of some gut microbiota involved in bleeding and gastrointestinal dysfunction indicating that the exogenous supplementation with Faecalibacterium and Akkermansia might be a prophylactic strategy for AF-OAC patients to lower the risk of bleeding after anticoagulation.


Subject(s)
Atrial Fibrillation , Gastrointestinal Microbiome , Stroke , Humans , Atrial Fibrillation/complications , Atrial Fibrillation/drug therapy , Atrial Fibrillation/chemically induced , Phylogeny , RNA, Ribosomal, 16S/genetics , Risk Factors , Anticoagulants/therapeutic use , Hemorrhage/chemically induced , Hemorrhage/complications , Administration, Oral , Risk Assessment
3.
J Trace Elem Med Biol ; 74: 127084, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36182726

ABSTRACT

BACKGROUND: Hypertension is a major contributor to cardiovascular morbidity and mortality, affecting over 17.1 million individuals worldwide. Environmental exposure such as toxic trace elements could be risk factors for hypertension, but the associations of toxic metal exposure with hypertension are not well understood. METHODS: We recruited 400 volunteers consisting of 200 patients with hypertension (cases) and 200 healthy individuals without hypertension (controls). In the case or control group, half of the subjects came from the rare earth mining (REM) areas and the other half from non-REM areas. Serum levels of 8 rare earth elements (REEs) and 13 non-REEs were determined. RESULTS: The concentrations of Ce and La were significant higher in the cases than in the controls in all comparisons. Serum concentrations of Mg, Mn, Dy, Ce and La were positively correlated with blood pressure, while those of concentrations K and Se were negatively correlated with blood pressure (p < 0.05). Compared with the lowest quartiles, participants in the highest quartiles of Sm, Gd, Dy, Yb, La and Ce had a 6.01-fold (95 % CI: 2.28, 15.8), 3.29-fold (95 % CI: 1.18, 9.16), 4.07-fold (95 % CI: 1.51,10.9), 7.83-fold (95 % CI: 2.78, 22.4), 20.00-fold (95 % CI: 5.48-72.9) and 6.13-fold (95 % CI: 2.13-17.6) increase in the probability of having hypertension respectively. Among all the detected metals, the univariate odds ratios (UORs) and adjusted odds ratios (AORs) of hypertension for highest vs. lowest quartile serum concentrations of Sm, Gd, Dy, Yb, La and Ce were significantly > 1 (p < 0.05), with the positive dose-response relationships observed between their serum levels and ORs associated with hypertension risk. CONCLUSIONS: Collectively, there appears to be a positive correlation between hypertension and environmental exposure to REEs, especially La and Ce. Further studies are warranted to investigate the underlying mechanisms responsible for the risk.


Subject(s)
Hypertension , Metals, Rare Earth , Trace Elements , Environmental Exposure/adverse effects , Humans , Metals, Rare Earth/analysis , Mining
4.
Gene ; 831: 146576, 2022 Jul 15.
Article in English | MEDLINE | ID: mdl-35568340

ABSTRACT

Abdominal aortic aneurysms (AAA) are pathological dilations in local aortic wall. The inflammatory infiltrates of the perivascular adipose tissue (PAT) surrounding AAAs were associated with AAAs and have been shown to contribute vascular pathology. However, the mechanism by which PAT inflammation contributes to vascular pathology in AAA remains to be clarified. This study aimed to explore the association between immune cell infiltration and key gene expression profile in PAT of AAA. For that, a gene expression dataset of human dilated perivascular adipose tissue (dPAT), non-dilated perivascular adipose tissue (ndPAT), subcutaneous abdominal fat (SAF) and omental-visceral fat (OVF) samples, as well as another microarray dataset of the abdominal perivascular adipose tissue in peripheral artery disease patients were downloaded from GEO database for analysis in this study. The CIBERSORT algorithm, weighted gene co-expression network analysis (WGCNA) and LASSO algorithm were used for the identification of immune infiltration, immune-related genes and the development of diagnostic signature. Our data discovered a significant higher proportion of activated mast cells and follicular helper T (Tfh) cells in dPAT than ndPAT, OVT and SAF samples. Moreover, AP-1 family members (FOS, FOSB, ATF3, JUN and JUNB) were found to compose the hub genes of purple module in WGCNA. Among them, FOS gene acts as a higher efficient marker to discriminate dPAT from ndPAT, OVT and SAF in AAA. Meanwhile, the expression profiles of the AP-1 family members are all significantly positive correlated with activated mast cell, plasma cell and Tfh cell infiltration in dPAT of AAA. Therefore, in the PAT surrounding AAA, the signature of inflammatory infiltration might be represented by a FOS-dominated cell network consist of activated mast cell, plasma cell and Tfh cell. Given the complicated etiology of AAA, our results are likely to shed new light on the pathophysiologic mechanism of AAA influenced by the local dPAT.


Subject(s)
Aortic Aneurysm, Abdominal , Proto-Oncogene Proteins c-fos/genetics , Adipose Tissue/metabolism , Aortic Aneurysm, Abdominal/metabolism , Genes, fos , Humans , Transcription Factor AP-1/genetics , Transcriptome
5.
BMC Nephrol ; 22(1): 257, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34238243

ABSTRACT

BACKGROUND: Few studies have evaluated the clinical presentation, management, and outcomes of patients with end-stage renal disease (ESRD) presenting with acute aortic dissection (AAD) in real-world clinical practice. Thus, this study investigated the clinical characteristics, management, and outcomes of AAD patients with ESRD. METHODS: A total of 217 patients were included. We evaluated the differences in the clinical features, management, and in-hospital outcomes of patients with and without a history of ESRD presenting with AAD. RESULTS: A history of ESRD was present in 71 of 217 patients. Patients with ESRD had atypical clinical manifestations (p < 0.001) and were more likely to be managed medically compared with patients without ESRD (p = 0.002). Hypertension and type B aortic dissection were significantly more common among patients with ESRD. Moreover, patients with ESRD had lower leucocyte and platelet counts than patients without ESRD in laboratory findings (p < 0.001). However, hospitalization days and in-hospital mortality were similar between the two groups (p > 0.05). Multivariate analysis identified Type A aortic dissection as an independent predictor of in-hospital mortality among patients without ESRD (OR, 13.68; 95% CI, 1.92 to 98.90; P = 0.006). CONCLUSIONS: This study highlights differences in the clinical characteristics, management, and outcomes of AAD patients with ESRD. These patients usually have atypical symptoms and more comorbid conditions and are managed more conservatively. However, these patients have no in-hospital survival disadvantage over those without ESRD. Further studies are needed to better understand and optimize care for patients with ESRD presenting with AAD.


Subject(s)
Aortic Aneurysm/complications , Aortic Aneurysm/therapy , Aortic Dissection/complications , Aortic Dissection/therapy , Kidney Failure, Chronic/complications , Adult , Aortic Dissection/blood , Aortic Dissection/surgery , Aortic Aneurysm/blood , Aortic Aneurysm/surgery , Female , Hospital Mortality , Humans , Hypertension/complications , Kidney Failure, Chronic/blood , Length of Stay , Leukocyte Count , Male , Middle Aged , Platelet Count , Retrospective Studies , Risk Factors
6.
Cell Death Dis ; 12(7): 665, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34215721

ABSTRACT

It has been reported that growth differentiation factor 11 (GDF11) protects against myocardial ischemia/reperfusion (IR) injury, but the underlying mechanisms have not been fully clarified. Considering that GDF11 plays a role in the aging/rejuvenation process and that aging is associated with telomere shortening and cardiac dysfunction, we hypothesized that GDF11 might protect against IR injury by activating telomerase. Human plasma GDF11 levels were significantly lower in acute coronary syndrome patients than in chronic coronary syndrome patients. IR mice with myocardial overexpression GDF11 (oe-GDF11) exhibited a significantly smaller myocardial infarct size, less cardiac remodeling and dysfunction, fewer apoptotic cardiomyocytes, higher telomerase activity, longer telomeres, and higher ATP generation than IR mice treated with an adenovirus carrying a negative control plasmid. Furthermore, mitochondrial biogenesis-related proteins and some antiapoptotic proteins were significantly upregulated by oe-GDF11. These cardioprotective effects of oe-GDF11 were significantly antagonized by BIBR1532, a specific telomerase inhibitor. Similar effects of oe-GDF11 on apoptosis and mitochondrial energy biogenesis were observed in cultured neonatal rat cardiomyocytes, whereas GDF11 silencing elicited the opposite effects to oe-GDF11 in mice. We concluded that telomerase activation by GDF11 contributes to the alleviation of myocardial IR injury through enhancing mitochondrial biogenesis and suppressing cardiomyocyte apoptosis.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Growth Differentiation Factors/metabolism , Mitochondria, Heart/enzymology , Myocardial Infarction/enzymology , Myocardial Reperfusion Injury/enzymology , Myocytes, Cardiac/enzymology , Organelle Biogenesis , Telomerase/metabolism , Aminobenzoates/pharmacology , Animals , Apoptosis , Bone Morphogenetic Proteins/genetics , Case-Control Studies , Cells, Cultured , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Growth Differentiation Factors/genetics , Humans , Male , Mice, Inbred C57BL , Mitochondria, Heart/drug effects , Mitochondria, Heart/genetics , Mitochondria, Heart/pathology , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/prevention & control , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/pathology , Naphthalenes/pharmacology , Rats , Signal Transduction , Telomerase/antagonists & inhibitors
7.
Cell Death Dis ; 12(4): 378, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33828087

ABSTRACT

Vascular smooth muscle cell (VSMC) phenotypic switching plays a critical role in the formation of abdominal aortic aneurysms (AAAs). FoxO3a is a key suppressor of VSMC homeostasis. We found that in human and animal AAA tissues, FoxO3a was upregulated, SM22α and α-smooth muscle actin (α-SMA) proteins were downregulated and synthetic phenotypic markers were upregulated, indicating that VSMC phenotypic switching occurred in these diseased tissues. In addition, in cultured VSMCs, significant enhancement of FoxO3a expression was found during angiotensin II (Ang II)-induced VSMC phenotypic switching. In vivo, FoxO3a overexpression in C57BL/6J mice treated with Ang II increased the formation of AAAs, whereas FoxO3a knockdown exerted an inhibitory effect on AAA formation in ApoE-/- mice infused with Ang II. Mechanistically, FoxO3a overexpression significantly inhibited the expression of differentiated smooth muscle cell (SMC) markers, activated autophagy, the essential repressor of VSMC homeostasis, and promoted AAA formation. Our study revealed that FoxO3a promotes VSMC phenotypic switching to accelerate AAA formation through the P62/LC3BII autophagy signaling pathway and that therapeutic approaches that decrease FoxO3a expression may prevent AAA formation.


Subject(s)
Aortic Aneurysm/physiopathology , Forkhead Box Protein O3/metabolism , Muscle, Smooth, Vascular/metabolism , Animals , Homeostasis , Humans , Male , Mice , Transfection
8.
Circulation ; 143(23): 2277-2292, 2021 06 08.
Article in English | MEDLINE | ID: mdl-33757294

ABSTRACT

BACKGROUND: Exercise can induce physiological myocardial hypertrophy (PMH), and former athletes can live 5 to 6 years longer than nonathletic controls, suggesting a benefit after regression of PMH. We previously reported that regression of pathological myocardial hypertrophy has antihypertrophic effects. Accordingly, we hypothesized that antihypertrophic memory exists even after PMH has regressed, increasing myocardial resistance to subsequent pathological hypertrophic stress. METHODS: C57BL/6 mice were submitted to 21 days of swimming training to develop PMH. After termination of exercise, PMH regressed within 1 week. PMH regression mice (exercise hypertrophic preconditioning [EHP] group) and sedentary mice (control group) then underwent transverse aortic constriction or a sham operation for 4 weeks. Cardiac remodeling and function were evaluated with echocardiography, invasive left ventricular hemodynamic measurement, and histological analysis. LncRNA sequencing, chromatin immunoprecipitation assay, and comprehensive identification of RNA-binding proteins by mass spectrometry and Western blot were used to investigate the role of Mhrt779 involved in the antihypertrophic effect induced by EHP. RESULTS: At 1 and 4 weeks after transverse aortic constriction, the EHP group showed less increase in myocardial hypertrophy and lower expression of the Nppa and Myh7 genes than the sedentary group. At 4 weeks after transverse aortic constriction, EHP mice had less pulmonary congestion, smaller left ventricular dimensions and end-diastolic pressure, and a larger left ventricular ejection fraction and maximum pressure change rate than sedentary mice. Quantitative polymerase chain reaction revealed that the long noncoding myosin heavy chain-associated RNA transcript Mhrt779 was one of the markedly upregulated lncRNAs in the EHP group. Silencing of Mhrt779 attenuated the antihypertrophic effect of EHP in mice with transverse aortic constriction and in cultured cardiomyocytes treated with angiotensin II, and overexpression enhanced the antihypertrophic effect. Using chromatin immunoprecipitation assay and quantitative polymerase chain reaction, we found that EHP increased histone 3 trimethylation (H3K4me3 and H3K36me3) at the a4 promoter of Mhrt779. Comprehensive identification of RNA-binding proteins by mass spectrometry and Western blot showed that Mhrt779 can bind SWI/SNF related, matrix associated, actin dependent regulator of chromatin, subfamily a, member 4 (Brg1) to inhibit the activation of the histone deacetylase 2 (Hdac2)/phosphorylated serine/threonine kinase (Akt)/phosphorylated glycogen synthase kinase 3ß(p-GSK3ß) pathway induced by pressure overload. CONCLUSIONS: Myocardial hypertrophy preconditioning evoked by exercise increases resistance to pathological stress via an antihypertrophic effect mediated by a signal pathway of Mhrt779/Brg1/Hdac2/p-Akt/p-GSK3ß.


Subject(s)
Cardiomegaly/therapy , Physical Conditioning, Animal , RNA, Long Noncoding/metabolism , Animals , Atrial Natriuretic Factor/genetics , Atrial Natriuretic Factor/metabolism , Cardiomegaly/genetics , Disease Models, Animal , Echocardiography , Glycogen Synthase Kinase 3 beta/metabolism , Hemodynamics , Histone Deacetylase 2/metabolism , Mice , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Up-Regulation , Ventricular Function, Left/physiology , Ventricular Remodeling
9.
Aging (Albany NY) ; 13(2): 2982-3009, 2021 01 15.
Article in English | MEDLINE | ID: mdl-33460402

ABSTRACT

Cardiorenal syndrome (CRS) has a high mortality, but its pathogenesis remains elusive. Fibroblast growth factor 23 (FGF23) is increased in both renal dysfunction and cardiac dysfunction, and FGF receptor 4 (FGFR4) has been identified as a receptor for FGF23. Deficiency of FGF23 causes growth retardation and shortens the lifespan, but it is unclear whether excess FGF23 is detrimental in CRS. This study sought to investigate whether FGF23 plays an important role in CRS-induced renal fibrosis. A mouse model of CRS was created by surgical myocardial infarction for 12 weeks. CRS mice showed a significant increase of circulatory and renal FGF23 protein levels, as well as an upregulation of p-GSK, active-ß-catenin, TGF-ß, collagen I and vimentin, a downregulation of renal Klotho expression and induction of cardiorenal dysfunction and cardiorenal fibrosis. These changes were enhanced by cardiac overexpression of FGF23 and attenuated by FGF receptor blocker PD173074 or ß-catenin blocker IGC001. In fibroblasts (NRK-49F), expression of FGFR4 rather than Klotho was detected. Recombinant FGF23 upregulated the expression of p-GSK, active-ß-catenin, TGF-ß, collagen I and vimentin proteins. These changes were attenuated by FGFR4 blockade with BLU9931 or ß-catenin blockade with IGC001. We concluded that FGF23 promotes CRS-induced renal fibrosis mediated by partly activating FGFR4/ß-catenin signaling pathway.


Subject(s)
Cardio-Renal Syndrome/metabolism , Fibroblast Growth Factors/metabolism , Fibrosis/metabolism , Kidney/pathology , Myocardium/metabolism , Animals , Cardio-Renal Syndrome/genetics , Cardio-Renal Syndrome/pathology , Cell Line , Disease Models, Animal , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Fibrosis/genetics , Fibrosis/pathology , Glucuronidase/genetics , Glucuronidase/metabolism , Kidney/metabolism , Klotho Proteins , Mice , Myocardium/pathology , Rats , Up-Regulation
10.
BMC Cardiovasc Disord ; 20(1): 32, 2020 Jan 28.
Article in English | MEDLINE | ID: mdl-31992206

ABSTRACT

BACKGROUND: The gut microbiome plays an important role in various cardiovascular diseases, such as atherosclerosis and hypertension, which are associated with abdominal aortic aneurysms (AAAs). METHODS: Here, we used 16S rRNA sequencing to explore gut microbiota in C57BL ApoE-/- mice with AAAs. A mouse model of abdominal aortic aneurysms was induced with angiotensin II (Ang II) (1000 ng/min per kg). On day 28 after the operation, fecal samples were collected and stored at - 80 °C until DNA extraction. We determined the relative abundances of bacterial taxonomic groups using 16S rRNA amplicon metabarcoding, and sequences were analyzed using a combination of mother software and UPARSE. RESULTS: We found that the gut microbiome was different between control and AAA mice. The results of correlation analysis between AAA diameter and the gut microbiome as well as LEfSe of the genera Akkermansia, Odoribacter, Helicobacter and Ruminococcus might be important in the progression of AAAs. CONCLUSIONS: AAA mice is subjected to gut microbial dysbiosis, and gut microbiota might be a potential target for further investigation.


Subject(s)
Aortic Aneurysm, Abdominal/microbiology , Bacteria/growth & development , Gastrointestinal Microbiome , Intestines/microbiology , Angiotensin II , Animals , Aortic Aneurysm, Abdominal/chemically induced , Bacteria/genetics , Bacteria/isolation & purification , Disease Models, Animal , Dysbiosis , Feces/microbiology , Male , Mice, Inbred C57BL , Mice, Knockout, ApoE , Ribotyping
11.
Cardiovasc Res ; 113(6): 620-632, 2017 May 01.
Article in English | MEDLINE | ID: mdl-28453729

ABSTRACT

AIMS: To resolve the controversy as to whether periostin plays a role in myocardial regeneration after myocardial infarction (MI), we created a neonatal mouse model of MI to investigate the influence of periostin ablation on myocardial regeneration and clarify the underlying mechanisms. METHODS AND RESULTS: Neonatal periostin-knockout mice and their wildtype littermates were subjected to MI or sham surgery. In the wildtype mice after MI, fibrosis was detectable at 3 days and fibrotic tissue was completely replaced by regenerated myocardium at 21 days. In contrast, in the knockout mice, significant fibrosis in the infarcted area was present at even 3 weeks after MI. Levels of phosphorylated-histone 3 and aurora B in the myocardium, detected by immunofluorescence and western blotting, were significantly lower in knockout than in wildtype mice at 7 days after MI. Similarly, angiogenesis was decreased in the knockout mice after MI. Expression of both the endothelial marker CD-31 and α-smooth muscle actin was markedly lower in the knockout than in wildtype mice at 7 days after MI. The knockout MI group had elevated levels of glycogen synthase kinase (GSK) 3ß and decreased phosphatidylinositol 3-kinase (PI3K), phosphorylated serine/threonine protein kinase B (p-Akt), and cyclin D1, compared with the wildtype MI group. Similar effects were observed in experiments using cultured cardiomyocytes from neonatal wildtype or periostin knockout mice. Administration of SB216763, a GSK3ß inhibitor, to knockout neonatal mice decreased myocardial fibrosis and increased angiogenesis in the infarcted area after MI. CONCLUSION: Ablation of periostin suppresses post-infarction myocardial regeneration by inhibiting the PI3K/GSK3ß/cyclin D1 signalling pathway, indicating that periostin is essential for myocardial regeneration.


Subject(s)
Cell Adhesion Molecules/deficiency , Cyclin D1/metabolism , Myocardial Infarction/enzymology , Myocardium/enzymology , Phosphatidylinositol 3-Kinase/metabolism , Regeneration , Repressor Proteins/metabolism , Signal Transduction , Animals , Animals, Newborn , Cell Adhesion Molecules/genetics , Cells, Cultured , Disease Models, Animal , Fibrosis , Mice, Knockout , Myocardial Infarction/genetics , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Neovascularization, Physiologic , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Regeneration/drug effects , Repressor Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Time Factors
12.
Cardiovasc Diabetol ; 15(1): 136, 2016 Sep 22.
Article in English | MEDLINE | ID: mdl-27659110

ABSTRACT

BACKGROUND: Left ventricular (LV) dysfunction is closely associated with LV hypertrophy or diabetes, as well as insufficient autophagic flux. Acute or chronic hyperglycemia is a prognostic factor for patients with myocardial infarction. However, the effect of acute hyperglycemia on LV dysfunction of the hypertrophic heart and the mechanisms involved are still unclear. This study aimed to confirm our hypothesis that either acute or chronic hyperglycemia suppresses LV diastolic function and autophagic flux. METHODS: The transverse aortic constriction (TAC) model and streptozocin-induced type 1 diabetic mellitus mice were used. LV function was evaluated with a Millar catheter. Autophagic levels and autophagic flux in the whole heart and cultured neonatal rat cardiomyocytes in response to hyperglycemia were examined by using western blotting of LC3B-II and P62. We also examined the effect of an autophagic inhibitor on LC3B-II and P62 protein expression and LC3 puncta. RESULTS: In mice with TAC, we detected diastolic dysfunction as early as 30 min after TAC. This dysfunction was indicated by a greater LV end-diastolic pressure and the exponential time constant of LV relaxation, as well as a smaller maximum descending rate of LV pressure in comparison with sham group. Similar results were also obtained in mice with TAC for 2 weeks, in addition to increased insulin resistance. Acute hyperglycemic stress suppressed diastolic function in mice with myocardial hypertrophy, as evaluated by invasive LV hemodynamic monitoring. Mice with chronic hyperglycemia induced by streptozocin showed myocardial fibrosis and diastolic dysfunction. In high glucose-treated cardiomyocytes and streptozocin-treated mice, peroxisome proliferator-activated receptor-γ coactivator 1α was downregulated, while P62 was upregulated. Autophagic flux was also significantly inhibited in response to high glucose exposure in angiotensin-II treated cardiomyocytes. CONCLUSIONS: Acute hyperglycemia suppresses diastolic function, damages mitochondrial energy signaling, and inhibits autophagic flux in prohypertrophic factor-stimulated cardiomyocytes.

13.
Oncotarget ; 7(40): 64649-64664, 2016 Oct 04.
Article in English | MEDLINE | ID: mdl-27579618

ABSTRACT

Fibroblast growth factor 23 (FGF23) has been reported to induce left ventricular hypertrophy, but it remains unclear whether FGF23 plays a role in cardiac fibrosis. This study is attempted to investigate the role of FGF23 in post-infarct myocardial fibrosis in mice. We noted that myocardial and plasma FGF23 and FGF receptor 4 were increased in mice with heart failure as well as in cultured adult mouse cardiac fibroblasts (AMCFs) exposed to angiotensin II, phenylephrine, soluble fractalkine. Recombinant FGF23 protein increased active ß-catenin , procollagen I and procollagen III expression in cultured AMCFs. Furthermore, intra-myocardial injection of adeno-associated virus-FGF23 in mice significantly increased left ventricular end-diastolic pressure and myocardial fibrosis, and markedly upregulated active ß-catenin, transforming growth factor ß (TGF-ß), procollagen I and procollagen III in both myocardial infarction (MI) and ischemia/reperfusion (IR) mice, while ß-catenin inhibitor or silencing of ß-catenin antagonized the FGF23-promoted myocardial fibrosis in vitro and in vivo. These findings indicate that FGF23 promotes myocardial fibrosis and exacerbates diastolic dysfunction induced by MI or IR, which is associated with the upregulation of active ß-catenin and TGF-ß.


Subject(s)
Fibroblast Growth Factors/metabolism , Fibroblasts/physiology , Hypertrophy, Left Ventricular/genetics , Myocardial Infarction/metabolism , Myocardium/pathology , Reperfusion Injury/metabolism , beta Catenin/metabolism , Angiotensin II/metabolism , Animals , Cells, Cultured , Collagen/metabolism , Dependovirus/genetics , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/genetics , Fibrosis , Humans , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/genetics , Phenylephrine/metabolism , RNA, Small Interfering/genetics , Rats , Rats, Sprague-Dawley , Receptor, Fibroblast Growth Factor, Type 4/blood , Reperfusion Injury/genetics , Transforming Growth Factor beta/metabolism , Up-Regulation , beta Catenin/genetics
14.
PLoS One ; 11(6): e0158514, 2016.
Article in English | MEDLINE | ID: mdl-27355349

ABSTRACT

High-mobility group box1 (HMGB1) exerts effects on inflammation by binding to receptor for advanced glycation end products (RAGE) or Toll-like receptor 4. Considering that inflammation is involved in pressure overload-induced cardiac hypertrophy, we herein attempted to investigate whether HMGB1 plays a role in myocardial hypertrophy in RAGE knockout mice as well as in the growth and apoptosis of cardiomyocytes. The myocardial expression of RAGE was not significantly changed while TLR4 mRNA was upregulated in response to transverse aortic constriction (TAC) for 1 week. The myocardial expression of HMGB1 protein was markedly increased in TAC group when compared to the sham group. Heart weight to body weight ratio (HW/BW) and lung weight to body weight ratio (LW/BW) were evaluated in RAGE knockout (KO) and wild-type (WT) mice 1 week after TAC. Significant larger HW/BW and LW/BW ratios were found in TAC groups than the corresponding sham groups, but no significant difference was found between KO and WT TAC mice. Similar results were also found when TAC duration was extended to 4 weeks. Cultured neonatal rat cardiomyocytes were treated with different concentrations of recombinant HMGB1, then cell viability was determined using MTT and CCK8 assays and cell apoptosis was determined by Hoechst staining and TUNEL assay. The results came out that HMGB1 exerted no influence on viability or apoptosis of cardiomyocytes. Besides, the protein expression levels of Bax and Bcl2 in response to different concentrations of HMGB1 were similar. These findings indicate that HMGB1 neither exerts influence on cardiac remodeling by binding to RAGE nor induces apoptosis of cardiomyocytes under physiological condition.


Subject(s)
Cardiomegaly , HMGB1 Protein/metabolism , Myocardium/metabolism , Receptor for Advanced Glycation End Products/metabolism , Animals , Aorta/pathology , Apoptosis , Cell Survival , Cells, Cultured , DNA Damage , Homozygote , Male , Mice , Mice, Knockout , Myocytes, Cardiac/cytology , Organ Size , Rats , Toll-Like Receptor 4/metabolism
15.
Oncotarget ; 6(22): 18829-44, 2015 Aug 07.
Article in English | MEDLINE | ID: mdl-26299920

ABSTRACT

MiR-497 is predicted to target anti-apoptosis gene Bcl2 and autophagy gene microtubule-associated protein 1 light chain 3 B (LC3B), but the functional consequence of miR-497 in response to anoxia/reoxygenation (AR) or ischemia/reperfusion (IR) remains unknown. This study was designed to investigate the influences of miR-497 on myocardial AR or IR injury. We noted that miR-497 was enriched in cardiac tissues, while its expression was dynamically changed in murine hearts subjected to myocardial infarction and in neonatal rat cardiomyocytes (NRCs) subjected to AR. Forced expression of miR-497 (miR-497 mimic) induced apoptosis in NRCs as determined by Hoechst staining and TUNEL assay. In response to AR, silencing of miR-497 using a miR-497 sponge significantly reduced cell apoptosis and enhanced autophagic flux. Furthermore, the infarct size induced by IR in adenovirus (Ad)-miR-497 sponge infected mice was significantly smaller than in mice receiving Ad-vector or vehicle treatment, while Ad-miR-497 increased infarct size. The expression of Bcl-2 and LC3B-II in NRCs or in murine heart was significantly decreased by miR-497 mimic and enhanced by miR-497 sponge. These findings demonstrate that inhibition of miR-497 holds promise for limiting myocardial IR injury.


Subject(s)
Hypoxia/metabolism , MicroRNAs/antagonists & inhibitors , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Animals , Apoptosis/physiology , Autophagy/physiology , Gene Knockdown Techniques , Hypoxia/genetics , Male , Mice , Mice, Inbred C57BL , MicroRNAs/biosynthesis , MicroRNAs/genetics , MicroRNAs/metabolism , Oxygen/administration & dosage , Oxygen/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/genetics , Reperfusion Injury/metabolism
16.
Biochem Biophys Res Commun ; 464(1): 201-7, 2015 Aug 14.
Article in English | MEDLINE | ID: mdl-26111448

ABSTRACT

Endoplasmic reticulum stress is a proapoptotic and profibrotic stimulus. Ablation of C/EBP homologous protein (CHOP) is reported to reverse cardiac dysfunction by attenuating cardiac endoplasmic reticulum stress in mice with pressure overload or ischemia/reperfusion, but it is unclear whether loss of CHOP also inhibits cardiac remodeling induced by permanent-infarction. In mice with permanent ligation of left coronary artery, we found that ablation of CHOP increased the acute phase mortality. For the mice survived to 4 weeks, left ventricular anterior (LV) wall thickness was larger in CHOP knockout mice than in the wildtype littermates, while no difference was noted on posterior wall thickness, LV dimensions, LV fractional shortening and ejection fraction. Similarly, invasive assessment of LV hemodynamics, morphological analysis of heart and lung weight indexes, myocardial fibrosis and TUNEL-assessed apoptosis showed no significant differences between CHOP knockout mice and their wildtype ones, while in mice with ischemia for 45 min and reperfusion for 1 week, myocardial fibrosis and apoptosis in the infarct area were significantly attenuated in CHOP knockout mice. These findings indicate that ablation of CHOP doesn't ameliorate cardiac remodeling induced by permanent-myocardial infarction, which implicates that early reperfusion is a prerequisite for ischemic myocardium to benefit from CHOP inhibition.


Subject(s)
Myocardial Infarction/genetics , Myocardial Infarction/mortality , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Transcription Factor CHOP/genetics , Ventricular Remodeling , Animals , Apoptosis , Disease Models, Animal , Gene Deletion , Gene Expression , Hemodynamics , Mice , Mice, Knockout , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardium/pathology , Myocytes, Cardiac/pathology , Survival Analysis , Transcription Factor CHOP/deficiency , Ultrasonography
SELECTION OF CITATIONS
SEARCH DETAIL
...