Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 24
1.
J Transl Med ; 21(1): 915, 2023 Dec 16.
Article En | MEDLINE | ID: mdl-38104081

BACKGROUND: SARS-CoV-2, the pathogen of COVID-19, is a worldwide threat to human health and causes a long-term burden on the cardiovascular system. Individuals with pre-existing cardiovascular diseases are at higher risk for SARS-CoV-2 infection and tend to have a worse prognosis. However, the relevance and pathogenic mechanisms between COVID-19 and cardiovascular diseases are not yet completely comprehended. METHODS: Common differentially expressed genes (DEGs) were obtained in datasets of human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) infected with SARS-CoV-2 and myocardial tissues from heart failure patients. Further GO and KEGG pathway analysis, protein-protein interaction (PPI) network construction, hub genes identification, immune microenvironment analysis, and drug candidate predication were performed. Then, an isoproterenol-stimulated myocardial hypertrophy cell model and a transverse aortic constriction-induced mouse heart failure model were employed to validate the expression of hub genes. RESULTS: A total of 315 up-regulated and 78 down-regulated common DEGs were identified. Functional enrichment analysis revealed mitochondrial metabolic disorders and extensive immune inflammation as the most prominent shared features of COVID-19 and cardiovascular diseases. Then, hub DEGs, as well as hub immune-related and mitochondria-related DEGs, were screened. Additionally, nine potential therapeutic agents for COVID-19-related cardiovascular diseases were proposed. Furthermore, the expression patterns of most of the hub genes related to cardiovascular diseases in the validation dataset along with cellular and mouse myocardial damage models, were consistent with the findings of bioinformatics analysis. CONCLUSIONS: The study unveiled the molecular networks and signaling pathways connecting COVID-19 and cardiovascular diseases, which may provide novel targets for intervention of COVID-19-related cardiovascular diseases.


COVID-19 , Cardiovascular Diseases , Heart Failure , Induced Pluripotent Stem Cells , Mitochondrial Diseases , Humans , Animals , Mice , Cardiovascular Diseases/genetics , SARS-CoV-2 , Computational Biology , Disease Models, Animal , Inflammation/genetics
2.
Adv Healthc Mater ; 12(13): e2202959, 2023 05.
Article En | MEDLINE | ID: mdl-36739582

Myocardial infarction (MI) is a cardiovascular disease that poses a serious threat to human health. Uncontrolled and excessive cardiac fibrosis after MI has been recognized as a primary contributor to mortality by heart failure. Thus, prevention of fibrosis or alleviation of fibrosis progression is important for cardiac repair. To this end, a biocompatible microneedle (MN) patch based on gelatin is fabricated to load exosomes containing microRNA-29b (miR-29b) mimics with antifibrotic activity to prevent excessive cardiac fibrosis after MI. Exosomes are isolated from human umbilical cord mesenchymal stem cells and loaded with miR-29b mimics via electroporation, which can be internalized effectively in cardiac fibroblasts to upregulate the expression of miR-29b and downregulate the expression of fibrosis-related proteins. After being implanted in the infarcted heart of a mouse MI model, the MN patch can increase the retention of loaded exosomes in the infarcted myocardium, leading to alleviation of inflammation, reduction of the infarct size, inhibition of fibrosis, and improvement of cardiac function. This design explored the MN patch as a suitable platform to deliver exosomes containing antifibrotic biomolecules locally for the prevention of cardiac fibrosis, showing the potential for MI treatment in clinical applications.


Exosomes , Fibrosis , MicroRNAs , Myocardial Infarction , Fibrosis/prevention & control , Myocardial Infarction/complications , Disease Models, Animal , MicroRNAs/therapeutic use , Humans , Animals , Mice , Electroporation/methods , Mesenchymal Stem Cells , Human Umbilical Vein Endothelial Cells
3.
Front Cell Dev Biol ; 9: 687769, 2021.
Article En | MEDLINE | ID: mdl-34395420

Human pluripotent stem cell-derived cardiomyocytes (hPSC-CMs) represent an infinite cell source for cardiovascular disease modeling, drug screening and cell therapy. Despite extensive efforts, current approaches have failed to generate hPSC-CMs with fully adult-like phenotypes in vitro, and the immature properties of hPSC-CMs in structure, metabolism and electrophysiology have long been impeding their basic and clinical applications. The prenatal-to-postnatal transition, accompanied by severe nutrient starvation and autophagosome formation in the heart, is believed to be a critical window for cardiomyocyte maturation. In this study, we developed a new strategy, mimicking the in vivo starvation event by Earle's balanced salt solution (EBSS) treatment, to promote hPSC-CM maturation in vitro. We found that EBSS-induced starvation obviously activated autophagy and mitophagy in human embryonic stem cell-derived cardiomyocytes (hESC-CMs). Intermittent starvation, via 2-h EBSS treatment per day for 10 days, significantly promoted the structural, metabolic and electrophysiological maturation of hESC-CMs. Structurally, the EBSS-treated hESC-CMs showed a larger cell size, more organized contractile cytoskeleton, higher ratio of multinucleation, and significantly increased expression of structure makers of cardiomyocytes. Metabolically, EBSS-induced starvation increased the mitochondrial content in hESC-CMs and promoted their capability of oxidative phosphorylation. Functionally, EBSS-induced starvation strengthened electrophysiological maturation, as indicated by the increased action potential duration at 90% and 50% repolarization and the calcium handling capacity. In conclusion, our data indicate that EBSS intermittent starvation is a simple and efficient approach to promote hESC-CM maturation in structure, metabolism and electrophysiology at an affordable time and cost.

4.
Cell Death Dis ; 11(5): 349, 2020 05 11.
Article En | MEDLINE | ID: mdl-32393744

Mesenchymal stem cells (MSCs) are derived from a wide range of sources and easily isolated and cultured. MSCs have the capacity for in vitro amplification and self-renewal, low immunogenicity and immunomodulatory properties, and under certain conditions, MSCs can be differentiated into a variety of cells. In the cardiovascular system, MSCs can protect the myocardium by reducing the level of inflammation, promoting the differentiation of myocardial cells around infarct areas and angiogenesis, increasing apoptosis resistance, and inhibiting fibrosis, which are ideal qualities for cardiovascular repair. Preclinical studies have shown that MSCs can be transplanted and improve cardiac repair, but challenges, such as their low rate of migration to the ischemic myocardium, low tissue retention, and low survival rate after transplantation, remain. This article reviews the potential and methods of MSC transplantation in the treatment of cardiovascular diseases (CVDs) and the challenges of the clinical use of MSCs.


Cardiovascular Diseases/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Animals , Cardiovascular Diseases/pathology , Cell Differentiation , Clinical Trials as Topic , Humans , Immunomodulation , Neovascularization, Physiologic
5.
J Mater Chem B ; 8(2): 308-315, 2020 01 14.
Article En | MEDLINE | ID: mdl-31808500

Low circulating levels of insulin-like growth factor 1 (IGF-1) have been correlated with an increased risk for cardiovascular diseases in humans. In this work, an injectable alginate hydrogel containing silk fibroin (SF) microspheres with the capability to sustain the release of IGF-1 was prepared to induce myocardial repair after myocardial infarction (MI). First, IGF-1 was physically adsorbed onto SF microspheres prepared by the coaxial needle system, and these IGF-1-containing microspheres were subsequently encapsulated into sodium alginate solutions at different concentrations (1.0-2.5%). Finally, this solution was crosslinked with 0.68% calcium gluconate solution to prepare the composite injectable hydrogel. The composite hydrogel prepared using a sodium alginate solution at a concentration of 1.5% could promote proliferation of H9C2 cardiomyocytes and reduce the cellular apoptosis rate under hypoxic conditions. The enzyme-linked immunosorbent assay results indicated that SF microspheres as microcarriers could effectively enhance the sustained release of IGF-1 from the hydrogels, causing the composite hydrogel to possess a better sustained release ability than the system without the SF microspheres. Moreover, echocardiography, hematoxylin-eosin staining, and Masson trichrome staining results indicated that an intramyocardial injection of the composite hydrogel into the peripheral region of a MI rat model could reduce the infarct size and improve the cardiac function after 28 days. The applications of such a composite hydrogel may comprise a powerful platform in cardiac tissue engineering.


Cell Proliferation/drug effects , Delayed-Action Preparations/pharmacology , Drug Carriers/chemical synthesis , Heart/drug effects , Insulin-Like Growth Factor I , Myocardial Infarction/drug therapy , Alginates/therapeutic use , Animals , Apoptosis/drug effects , Cell Line , Female , Fibroins/therapeutic use , Hydrogels/therapeutic use , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor I/therapeutic use , Microspheres , Rats , Rats, Sprague-Dawley
6.
Stem Cell Res Ther ; 10(1): 295, 2019 09 23.
Article En | MEDLINE | ID: mdl-31547872

BACKGROUND: Interleukin 33 is known to have an important influence in the process of myocardial infarction, and the immunoregulatory function of MSCs could be influenced by cell factors. In this study, we evaluated the therapeutic efficacy of IL-33-overexpressing bone marrow mesenchymal stem cells (IL33-MSCs) on myocardial infarction (MI) and detected the inflammatory level and cardiac function in rats. METHODS AND RESULTS: First, we evaluated the proliferation of T cells and polarization of macrophages that had been co-cultured with Vector-MSCs or IL33-MSCs. Co-culture experiments indicated that IL33-MSCs reduced T cell proliferation and enhanced CD206+ macrophage polarization. Second, we determined the inflammation level and cardiac function of PBS-, Vector-MSC-, and IL33-MSC-injected rats. Echocardiography indicated that left ventricular ejection fraction (LVEF) was enhanced in IL33-MSC-injected rats compared with Vector-MSC-injected rats. Postmortem analysis of rat heart tissue showed reduced fibrosis and less inflammation in IL33-MSC-injected rats. CONCLUSION: These studies indicated that the IL33-MSC injection improved heart function and reduces inflammation in rats with MI compared with PBS or Vector-MSC injections. IL-33 overexpression enhances the immunomodulatory function and therapeutic effects of MSCs on acute MI via enhancing the polarization of macrophages toward M2, enhancing the differentiation of CD4+ T cells toward CD4+IL4+Th2 cells, and finally, reducing heart inflammation and enhancing heart function.


Interleukin-33/metabolism , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/therapy , Animals , Cells, Cultured , Culture Media, Conditioned/pharmacology , Interleukin-33/genetics , Macrophages/drug effects , Male , Rats , Rats, Sprague-Dawley , T-Lymphocytes/drug effects
7.
Mol Ther Nucleic Acids ; 16: 284-294, 2019 Jun 07.
Article En | MEDLINE | ID: mdl-30951965

Abnormal phenotypic switch, migration, and proliferation of vascular smooth muscle cells (VSMCs) are hallmarks for pathogenesis of thoracic aortic dissection (TAD). In the current study, we identified miR-134-5p as a critical regulator controlling human VSMC phenotypic switch and migration to investigate whether miR-134-5p affects human VSMC functions and development of TAD. Using miRNA microarray of aorta specimens from 12 TAD and 12 controls, we identified miR-134-5p, which was significantly downregulated in TAD tissues. With qPCR detection, we found that miR-134-5p was also evidently decreased in human AoSMCs. Ectopic expression of miR-134-5p obviously promoted VSMC differentiation and expression of contractile markers, such as α-SMA, SM22α, and MYH11. miR-134-5p potently inhibited PDGF-BB-induced VSMC phenotypic switch and migration. We further identified STAT5B and ITGB1 as downstream targets of miR-134-5p in human VSMCs and proved them to be mediators in VSMC phenotypic switch and progression of TAD. Finally, Ad-miR-134-5p obviously suppressed the aorta dilatation and vascular media degeneration by 39% in TAD mice after vascular injury induced by Ang II. Our findings revealed that miR-134-5p was a novel regulator in vascular remodeling and pathological progress of TAD via targeting STAT5B/ITGB1 expression. Targeting miR-134-5p or its downstream molecules in VSMCs might develop new avenues in clinical treatment of TAD.

9.
Aging (Albany NY) ; 10(12): 3683-3701, 2018 12 07.
Article En | MEDLINE | ID: mdl-30530919

Thoracic aortic aneurysm and dissection (TAAD) is the most fatal macro vascular disease. The mortality of 48h after diagnosis of dissection is up to approximately 50-68%. However, the genetic factors and potential mechanism underlying sporadic TAAD remain largely unknown. Our previous study suggested rs12455792 variant of SMAD4 gene significantly contributed to the increased risk and might participated the pathological progression of TAAD. This investigation aims to test (1) the associations between rs12455792 and MØ recruitment, inflammatory response in aggressiveness of TAAD, and (2) the molecular mechanism accounting for their effects. In TGF-ß signaling molecular detection, rs12455792 C>T variant activated the canonical and non-canonical TGF-ß mediators. It also increased the secretion of chemotactic factors of HASMCs. To confirm the impact of this change, we detected MØ recruitment and infiltration in HASMCs and aortic tissues of TAAD patients. We found that MØ recruitment in cells and tissues with rs12455792 variant genotypes was increased than that in wild type groups. Moreover, rs12455792 variant increased M1 type inflammatory response, which might contribute much to TAAD progression. To mimic the SMAD4 suppression effect of rs12455792 in vivo, we constructed the SMAD4 KD mouse. After induction with Ang II for 4w, the thoracic aorta dilatation and vascular remodeling were more serious than that of wild type group. In conclusion, rs12455792 increased MØ recruitment, M1 type inflammatory response via activated TGF-ß signaling, and further promoted vascular remodeling and pathological progress of TAAD.


Aortic Aneurysm, Thoracic/metabolism , Aortic Dissection/metabolism , Inflammation Mediators/metabolism , Macrophages/metabolism , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Aortic Dissection/genetics , Aortic Dissection/pathology , Animals , Aorta, Thoracic/metabolism , Aorta, Thoracic/pathology , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/pathology , Case-Control Studies , Cells, Cultured , Dilatation, Pathologic , Disease Models, Animal , Female , Macrophages/pathology , Male , Mice, Knockout , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/pathology , Polymorphism, Single Nucleotide , Signal Transduction , Smad4 Protein/deficiency , Smad4 Protein/genetics , Vascular Remodeling
10.
J Thorac Dis ; 10(9): 5476-5488, 2018 Sep.
Article En | MEDLINE | ID: mdl-30416797

BACKGROUND: Bone marrow-derived mesenchymal stem cells (BMSCs) have been proved to be capable of differentiating into endothelial cells (ECs), however, the differentiation efficiency is rather low. Sonic hedgehog (Shh), an important factor in vascular development and postnatal angiogenesis, exerted promotional effect on new vessel formation in the ischemic animal models. Therefore, the current study aims to investigate whether Shh could induce the endothelial differentiation of BMSCs both in vitro and in vivo, as well as the mechanism of differentiation induction. METHODS: The current study over-expressed Shh in BMSCs by lentivirus transduction. Reverse-transcription quantitative polymerase chain reaction (RT-qPCR) analysis was performed to determine the angiogenic factors in both control BMSCs and Shh over-expressed BMSCs. Immunocytochemistry was also conducted to examine the EC markers. Angiogenesis was determined by in vitro tube-forming assay on Matrigel and in vivo Matrigel plug in severe combined immunodeficient (SCID) mice. Last, mRNA sequencing analysis was used to elaborate the underlying mechanisms. Loss of function study was performed by vascular endothelial growth factor D (VEGF-D) siRNA. RESULTS: Shh expression was increased by about 3,000-fold and 5,000-fold at 3 days-transfection and 7 days-transfection, respectively. Patched 1 (Ptch1), the receptor for Shh, had a two-fold increase after transduction. The angiogenic factors such as hepatocyte growth factor (HGF), angiopoietin-1 (Ang-1), insulin-like growth factor 1 (IGF1) and vascular endothelial growth factor A (VEGF-A) had at least a 1.5-fold increase after transduction. Expression of EC-lineage markers, CD31 and VE-cadherin, on Shh-overexpressed BMSCs were increasingly detected by immunocytostaining. Angiogenesis of BMSCs could be efficiently induced by Shh overexpression in the in vitro tube-formation assay and in vivo Matrigel plug. Additionally, mRNA sequencing analysis revealed that Shh activation upregulated the expression of several pro-angiogenic factors, like Angptl4, Egfl6, VEGF-D. Loss of function study by VEGF-D siRNA confirmed that Shh enhanced the angiogenic ability of BMSCs via VEGF-D. CONCLUSIONS: This study demonstrated that Shh could promote endothelial differentiation of BMSCs via VEGF-D.

11.
Exp Ther Med ; 16(5): 3785-3792, 2018 Nov.
Article En | MEDLINE | ID: mdl-30344653

Apolipoprotein M (apoM) is a recently identified human apolipoprotein that is associated with the formation of high-density lipoprotein (HDL). Studies have demonstrated that statins may affect the expression of apoM; however, the regulatory effects of statins on apoM are controversial. Furthermore, the underlying mechanisms by which statins regulate apoM remain unclear. In the present study, in vivo and in vitro models were used to investigate whether the anti-atherosclerotic effects of statins are associated with its apoM-regulating effects and the underlying mechanism. Hyperlipidemia was induced by in apolipoprotein E-deficient mice by providing a high-fat diet. Atorvastatin was administered to hyperlipidemic mice and HepG2 cells to investigate its effect on apoM expression. The liver X receptor α (LXRα) agonist T0901317 was also administered together with atorvastatin to hyperlipidemic mice and HepG2 cells. The results revealed that atorvastatin increased apoM expression, which was accompanied with decreased expression of LXRα in the liver of hyperlipidemic apolipoprotein E-deficient mice and HepG2 cells. Additionally, apoM upregulation was inhibited following treatment with T0901317. In summary, atorvastatin exhibited anti-atherosclerotic effects by upregulating apoM expression in hyperlipidemic mice, which may be mediated by the inhibition of LXRα.

12.
Stem Cells Int ; 2018: 3290372, 2018.
Article En | MEDLINE | ID: mdl-30271437

BACKGROUND: To cure ischemic diseases, angiogenesis needs to be improved by various strategies in ischemic area. Considering that microRNA-132 (miR-132) regulates endothelial cell behavior during angiogenesis and the safe and efficacious delivery of microRNAs in vivo is rarely achieved, an ideal vehicle for miR-132 delivery could bring the promise for ischemic diseases. As a natural carrier of biological molecules, exosomes are more and more developed as an ideal vehicle for miRNA transfer. Meanwhile, mesenchymal stem cells could release large amounts of exosomes. Thus, this study aimed to investigate whether MSC-derived exosomes can be used for miR-132 delivery in the treatment of myocardial ischemia. METHODS: MSC-derived exosomes were electroporated with miR-132 mimics and inhibitors. After electroporation, miR-132 exosomes were labelled with DiI and added to HUVECs. Internalization of DiI-labelled exosomes was examined by fluorescent microscopy. Expression levels of miR-132 in exosomes and HUVECs were quantified by real-time PCR. The mRNA levels of miR-132 target gene RASA1 in HUVECs were quantified by real-time PCR. Luciferase reporter assay was performed to examine the targeting relationship between miR-132 and RASA1. The effects of miR-132 exosomes on the angiogenic ability of endothelial cells were evaluated by tube formation assay. Matrigel plug assay and myocardial infarction model were used to determine whether miR-132 exosomes can promote angiogenesis in vivo. RESULTS: miR-132 mimics were effectively electroporated and highly detected in MSC-derived exosomes. The expression level of miR-132 was high in HUVECs preincubated with miR-132 mimic-electroporated exosomes and low in HUVECs preincubated with miR-132 inhibitor-electroporated exosomes. The expression level of RASA1, miR-132 target gene, was reversely correlated with miR-132 expression in HUVECs pretreated with exosomes. Luciferase reporter assay further confirmed that RASA1 was a direct target of miR-132. Exosomes loaded with miR-132, as a vehicle for miRNA transfer, significantly increased tube formation of endothelial cells. Moreover, subcutaneous injection of HUVECs pretreated with miR-132 exosomes in nude mice significantly increased their angiogenesis capacity in vivo. In addition, transplantation of miR-132 exosomes in the ischemic hearts of mice markedly enhanced the neovascularization in the peri-infarct zone and preserved heart functions. CONCLUSIONS: The findings suggest that the export of miR-132 via MSC-derived exosomes represents a novel strategy to enhance angiogenesis in ischemic diseases.

13.
EBioMedicine ; 21: 197-205, 2017 Jul.
Article En | MEDLINE | ID: mdl-28666732

Recent studies indicate important roles for SMAD4 in SMCs proliferation, extracellular matrix maintenance, and blood vessel remodeling. However, the genetic effects of SMAD4 in the pathogenesis of thoracic aortic aneurysm and dissection (TAAD) are still largely unknown. Here we identified a functional variant of SMAD4 which might be involved in the pathological progression of TAAD. Five tagging SNPs of SMAD4 were genotyped in 202 TAAD cases and 400 controls using MALDI-TOF. rs12455792 CT or TT variant genotypes was associated with an significantly elevated TAAD risk (adjusted OR=1.58, 95%CI=1.09-2.30) under a dominant genetic model. It was located in the 5'UTR and predicted to influence transcription activity and RNA folding of SMAD4. In luciferase reporter assay, rs12455792 T allele markedly decreased luciferase activities. Accordingly, SMAD4 expression in tissues was lower in patients with CT or TT genotypes, compared with CC. Movat's pentachrome showed that rs12455792 T allele enhanced SMCs loss and fibers accumulation. With angiotensin II induction, rate of Apoptotic SMCs was significantly higher while SMAD4 silenced. Moreover, rs12455792 T allele also increased Versican degradation via ADAMTS-4. In conclusion, this variant might promote SMCs apoptosis and proteoglycans degradation, and further facilitate the progress of TAAD. Our findings identified rs12455792 as a predictor for progression of vascular media pathological changes related thoracic aortic disorders.


Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Aortic Dissection/genetics , Aortic Dissection/metabolism , Apoptosis/genetics , Myocytes, Smooth Muscle/metabolism , Proteoglycans/metabolism , Smad4 Protein/genetics , Adult , Aged , Alleles , Computational Biology/methods , Disease Susceptibility , Gene Expression , Genetic Linkage , Genotype , Humans , Linkage Disequilibrium , Middle Aged , Models, Biological , Odds Ratio , Polymorphism, Single Nucleotide , Smad4 Protein/chemistry , Smad4 Protein/metabolism
14.
Heart Surg Forum ; 20(2): E066-E068, 2017 Apr 30.
Article En | MEDLINE | ID: mdl-28481746

Myxoma is the most commonly found cardiac primary tumor. The left atrium is the most common localization of myxoma, followed by the right atrium. However, it is rare in the left and right ventricles. Myxoma originating from cardiac valves is extremely rare. This article presents a case of a 17-year-old male who was admitted due to heart murmur for one year. Transthoracic echocardiography indicated a 1.9 cm round solid mass in the left ventricular outflow tract. Excision surgery and aortic valve replacement were performed in this patient. Histopathology revealed the mass as a myxoma. The aortic valve remains a very rare myxoma localization position. Echocardiography can provide a precise method for myxoma diagnosis. Early excision associated with valve replacement can provide good curative effects.


Heart Neoplasms/diagnosis , Myxoma/diagnosis , Adolescent , Aortic Valve , Cardiac Surgical Procedures , Echocardiography, Transesophageal , Heart Neoplasms/surgery , Humans , Male , Myxoma/surgery
15.
J Exp Clin Cancer Res ; 34: 151, 2015 Dec 21.
Article En | MEDLINE | ID: mdl-26689197

BACKGROUND: Metastasis-associated gene 1(MTA1) has been identified as an oncogene in many tumors, and aberrant MTA1 expression has been linked to carcinogenesis and metastasis. We aim to investigate the mechanism of MTA1 and metastasis in malignant pleural mesothelioma (MPM). METHODS: Real-time polymerase chain reaction (PCR) and immunohistochemical staining were employed to detect MTA1 and E-cadherin expression in MPM tissues and corresponding adjacent tissues. Stable clone with knock-down of MTA1 was generated with shRNA via lentivirus technology in MPM cell lines. Wound-healing assay, transwell assay and PCR array were carried out for detecting invasion and migration of MPM cells. Luciferase reporter assay was performed to validate the effect of MTA1 on E-cadherin. RESULTS: MTA1 expression is up-regulated in MPM and shown a negative correlation with E-cadherin expression. MTA1 could enhance the invasion and migration of MPM cells via suppressing the expression of E-cadherin. MTA1 overexpression is associated with pathology, metastasis and survival rate of MPM patients. CONCLUSIONS: MTA1 plays an important role in Epithelial-to-mesenchymal transition (EMT) to promote metastasis via suppressing E-cadherin expression, resulting in a poor prognosis in MPM. MTA1 is a novel biomarker and indicative of a poor prognosis in MPM patients.


Biomarkers, Tumor/biosynthesis , Cadherins/biosynthesis , Epithelial-Mesenchymal Transition/genetics , Histone Deacetylases/biosynthesis , Lung Neoplasms/genetics , Mesothelioma/genetics , Repressor Proteins/biosynthesis , Adult , Biomarkers, Tumor/genetics , Cadherins/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Histone Deacetylases/genetics , Humans , Male , Mesothelioma, Malignant , Middle Aged , Neoplasm Invasiveness/genetics , Prognosis , RNA, Small Interfering/genetics , Repressor Proteins/genetics , Survival Analysis , Trans-Activators
16.
J Surg Res ; 196(1): 49-55, 2015 Jun 01.
Article En | MEDLINE | ID: mdl-25813142

BACKGROUND: In patients with chronic severe aortic regurgitation (AR), aortic valve replacement (AVR) has been proved to promote left ventricular (LV) remodeling, especially LV end-diastolic dimension (LVEDD) reduction. However, there is little research whether postoperative LVEDD could return to normal parameter after AVR. The objective of this study was to determine predictors for the recovery of dilated LVEDD early after AVR. METHODS: The echocardiographic data of 105 patients, who underwent AVR for chronic pure AR between January 2005 and December 2011, were analyzed at the preoperative (3-7 d), early (6-8 mo), and late (2-y) postoperative stages, retrospectively. According to the baseline level, LVEDD >70 mm or LV end-systolic dimension (LVESD) >50 mm or LVESD index >25 mm/m(2) were defined as severe LV dilation. Patients were then categorized into two groups (group 1: severe LV dilation; group 2: nonsevere LV dilation). RESULTS: In all patients, four-fifth of the reduction in LV dimension occurred at early (6-8 mo) postoperative stage. The patients in both groups had significant decreases in the LVEDD and LVESD early after AVR, with an additional but insignificant reduction at late postoperative stage. The ejection fraction (EF) in both groups significantly increased at either early or late stage. However, the LVEDD and LVESD in group 1 were larger than those in group 2, and the EF in group 1 was lower than that in group 2 at early postoperative stage. By multivariate analysis, we found that the preoperative EF was a good predictor for the recovery of dilated LVEDD early after AVR (P = 0.009). Receiver-operating characteristics analysis showed that EF >52% was the best cut-off value for the recovery of LVEDD. CONCLUSIONS: In patients with chronic pure AR, preoperative EF may be a good predictor for successful recovery of dilated LVEDD early after AVR.


Aortic Valve Insufficiency/surgery , Aortic Valve/surgery , Heart Valve Prosthesis Implantation , Stroke Volume , Ventricular Function, Left , Adult , Aged , Aortic Valve Insufficiency/physiopathology , Chronic Disease , Diastole/physiology , Female , Humans , Male , Middle Aged
17.
Sci China Life Sci ; 57(2): 195-200, 2014 Feb.
Article En | MEDLINE | ID: mdl-24430558

It is still unclear whether the timing of intracoronary stem cell therapy affects the therapeutic response in patients with myocardial infarction. The natural course of healing the infarction and the presence of putative homing signals within the damaged myocardium appear to favor cell engraftment during the transendothelial passage in the early days after reperfusion. However, the adverse inflammatory environment, with its high oxidative stress, might be deleterious if cells are administered too early after reperfusion. Here we highlight several aspects of the timing of intracoronary stem cell therapy. Our results showed that transplantation of bone marrow mesenchymal stem cells at 2-4 weeks after myocardial infarction is more favorable for reduction of the scar area, inhibition of left ventricular remodeling, and recovery of heart function. Coronary injection of autologous bone marrow mesenchymal stem cells at 2-4 weeks after acute myocardial infarction is safe and does not increase the incidence of complications.


Bone Marrow Cells/cytology , Bone Marrow Transplantation , Mesenchymal Stem Cells/cytology , Myocardial Infarction/surgery , Animals , Enzyme-Linked Immunosorbent Assay , Heart Function Tests , Myocardial Infarction/physiopathology , Oxidative Stress , Swine , Transplantation, Autologous
18.
Differentiation ; 86(1-2): 57-64, 2013.
Article En | MEDLINE | ID: mdl-23974360

The objective of this study was to screen mouse bone marrow mesenchymal stromal cells (BMSCs) according to expression of cardiac stem cell (CSC) surface antigens and to assess the effects of resulting BMSC-like subsets on cardiac function after injection in a mouse myocardial infarct model. BMSCs were sorted by magnetic beads according to the expression of differentiation antigens on the surface of mouse CSCs, and four subsets were identified on the basis of CD45 and CD31 expression: stem cell antigen-1+ (Sca-1+)/CD45-/CD31-, Sca-1+/CD45-/CD31+, Sca-1+/CD45+/CD31-, and Sca-1+/CD45+/CD31+. When co-cultured with myocardial stem cells and 5-aza-2'-deoxycytidine for 14 days, each subset showed expression of cardiac markers α-actin, connexin 43, desmin, and cardiac troponin I; however, expression was greatest in Sca-1+/CD45+/CD31+ cells. To assess the ability of these cells to improve cardiac function, each subset was injected separately into mice with myocardial infarct induced by ligation of the left anterior descending coronary artery, and in vivo cardiac dual inversion recovery (DIR) imaging and Doppler echocardiography were performed 48 h, 96 h, and 7 days after injection. Results indicated that Sca-1+/CD45+/CD31+ cells were superior in improving cardiac function compared with the other subsets and with unsorted BMSCs. These results suggest that mouse BMSC cells are polyclonal and that the BMSC-like Sca-1+/CD45+/CD31+ subset was effective in directing cardiac differentiation and improving cardiac function in mice with myocardial infarcts.


Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Myocardial Infarction/surgery , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Antigens, Ly/genetics , Antigens, Ly/metabolism , Leukocyte Common Antigens/genetics , Leukocyte Common Antigens/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mice , Myoblasts, Cardiac/cytology , Myoblasts, Cardiac/metabolism , Myocardial Infarction/metabolism , Myocardial Infarction/physiopathology , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/genetics
19.
Zhonghua Xin Xue Guan Bing Za Zhi ; 41(3): 210-4, 2013 Mar.
Article Zh | MEDLINE | ID: mdl-23879945

OBJECTIVE: To search for the bone mesenchymal stem cell (MSC) subgroup which might be more effective on repairing myocardial damage. METHODS: In this experiment, four MSC subgroups were defined based on the surface differentiation antigen detection of mouse bone mesenchymal stem cells (mBMSCs): SCA-1(+)/CD45(+)/CD31(+), SCA-1(+)/CD45(+)/CD31(-), SCA-1(+)/CD45(-)/CD31(-) and SCA-1(+)/CD45(-)/CD31(+). These subgroup cells and unselected mBMSCs were injected into infarcted mouse via tail vein. Echocardiographic heart function measurement and in vivo DiR-labeled stem cells imaging were performed at 48 h after injection. In situ C-kit (a flag antigen of cardiac stem cells) and cardiac-specific differentiation antigen immunohistochemistry detection was made in the infarcted myocardium. RESULTS: The capacity of the SCA-1(+)/CD45(+)/CD31(+) cells on improving heart function was significantly higher than other cell groups (all P < 0.05). In vivo imaging showed that the mean fluorescence intensity of the SCA-1(+)/CD45(+)/CD31(+) cells was also higher than other cell groups (all P < 0.05). Number of cardiac stem cells in the infracted myocardium was significantly increased after the injection of all subgroup cells and unsorted mBMSCs cells for 48 h compared untreated infracted myocardium. The capacity of mobilizing cardiac stem cells is as follows: SCA-1(+)/CD45(+)/CD31(+) >SCA-1(+)/CD45(-)/CD31(+) >SCA-1(+)/CD45(-)/CD31(-) >SCA-1(+)/CD45(+)/CD31(-). CONCLUSION: The SCA-1(+)/CD45(+)/CD31(+) subgroups of mBMSCs exhibites the highest capacity to improve cardiac function after myocardial infarction and to mobilize autologous cardiac stem cells compared with other mBMSCs subgroups and unsorted mBMSCs cells.


Mesenchymal Stem Cell Transplantation , Myocardial Infarction/surgery , Animals , Cells, Cultured , Disease Models, Animal , Mice , Mice, Inbred C57BL
20.
J Surg Res ; 185(2): 896-903, 2013 Dec.
Article En | MEDLINE | ID: mdl-23870834

BACKGROUND: Mesenchymal stem cells (MSCs) have the potential to improve graft outcomes and promote allograft tolerance. In this study, we examined the effects and mechanism of combined intrathymic (i.t.) and intravenous (i.v.) injection of MSCs on the survival of transplanted hearts in a rat allograft model. METHODS: Recipient Sprague-Dawley rats were transplanted with hearts from Wistar rats. Wistar rat MSCs were infused via i.t. or i.v. or combined i.t. and i.v. (i.t./i.v.) injection at designated intervals. In vitro mixed lymphocyte reaction assays were performed to assess the immunosuppressive capacity of MSCs. Mesenchymal stem cell surface markers and CD4+, CD25+, and Foxp3+ T-cells in the peripheral blood were detected using flow cytometry analysis. The expression of microRNAs and cytokines in graft infiltrating lymphocytes was analyzed by real-time polymerase chain reaction. RESULTS: The MSCs cultured in vitro had multipotential differentiation capacity. Mixed lymphocyte reaction assays showed that donor-derived MSCs could not stimulate a proliferative response of recipient lymphocytes and could markedly suppress T-cell responses. Survival of the allografts was significantly prolonged by administration of i.t./i.v. injection of MSCs compared with controls, with a mean survival of 32.2 versus 6.5 d, respectively. Compared with the syngeneic groups posttransplant, miR-155 expression was significantly increased in the allogeneic group, and could be restored by injection of MSCs, especially i.t./i.v. injection of MSCs. Moreover, i.t./i.v. injection of MSCs decreased the level of interleukin (IL)-2 and interferon-gamma, but increased the levels of IL-4 and IL-10 in the allogeneic group. More important, i.t./i.v. injection of MSCs was the best way to increase the percentage of CD4+, CD25+, and Foxp3+ T-cell peripheral blood. CONCLUSIONS: Our results indicated that i.t./i.v. injection of MSCs can prolong the survival of rat cardiac allograft, which may be associated with down-regulating miR-155 expression, a shift in the Th1/Th2 balance, and up-regulation of Treg cells expression.


Graft Survival/immunology , Mesenchymal Stem Cell Transplantation/methods , MicroRNAs/genetics , Myocardium/cytology , Thymus Gland/cytology , Animals , Cells, Cultured , Coculture Techniques , Injections, Intravenous , Lymphocyte Culture Test, Mixed , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/immunology , Rats , Rats, Sprague-Dawley , Rats, Wistar , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th1 Cells/immunology , Th2 Cells/cytology , Th2 Cells/immunology , Transplantation, Homologous
...