Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
J Cell Physiol ; 2024 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-38946152

RESUMO

Skeletal muscle injury affects the quality of life in many pathologies, including volumetric muscle loss, contusion injury, and aging. We hypothesized that the nicotinamide phosphoribosyltransferase (Nampt) activator P7C3 improves muscle repair following injury. In the present study, we tested the effect of P7C3 (1-anilino-3-(3,6-dibromocarbazol-9-yl) propan-2-ol) on chemically induced muscle injury. Muscle injury was induced by injecting 50 µL 1.2% barium chloride (BaCl2) into the tibialis anterior (TA) muscle in C57Bl/6J wild-type male mice. Mice were then treated with either 10 mg/kg body weight of P7C3 or Vehicle intraperitoneally for 7 days and assessed for histological, biochemical, and molecular changes. In the present study, we show that the acute BaCl2-induced TA muscle injury was robust and the P7C3-treated mice displayed a significant increase in the total number of myonuclei and blood vessels, and decreased serum CK activity compared with vehicle-treated mice. The specificity of P7C3 was evaluated using Nampt+/- mice, which did not display any significant difference in muscle repair capacity among treated groups. RNA-sequencing analysis of the injured TA muscles displayed 368 and 212 genes to be exclusively expressed in P7C3 and Veh-treated mice, respectively. There was an increase in the expression of genes involved in cellular processes, inflammatory response, angiogenesis, and muscle development in P7C3 versus Veh-treated mice. Conversely, there is a decrease in muscle structure and function, myeloid cell differentiation, glutathione, and oxidation-reduction, drug metabolism, and circadian rhythm signaling pathways. Chromatin immunoprecipitation-quantitative polymerase chain reaction (qPCR) and reverse transcription-qPCR analyses identified increased Pax7, Myf5, MyoD, and Myogenin expression in P7C3-treated mice. Increased histone lysine (H3K) methylation and acetylation were observed in P7C3-treated mice, with significant upregulation in inflammatory markers. Moreover, P7C3 treatment significantly increased the myotube fusion index in the BaCl2-injured human skeletal muscle in vitro. P7C3 also inhibited the lipopolysaccharide-induced inflammatory response and mitochondrial membrane potential of RAW 264.7 macrophage cells. Overall, we demonstrate that P7C3 activates muscle stem cells and enhances muscle injury repair with increased angiogenesis.

2.
J Pharmacol Exp Ther ; 382(2): 233-245, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35680376

RESUMO

Diabetes is associated with increased cardiac injury and sudden death. Nicotinamide phosphoribosyltransferase (Nampt) is an essential enzyme for the NAD+ salvage pathway and is dysregulated in diabetes. Nampt activation results in rescued NADH/NAD+ ratios and provides pharmacological changes necessary for diabetic cardioprotection. Computer docking shows that 1-(3,6-Dibromo-carbazol-9-yl)-3-phenylamino-propan-2-ol (P7C3) allows for enhanced Nampt dimerization and association. To test the pharmacological application, we used male leptin receptor-deficient (db/db) mice and treated them with Nampt activator P7C3. The effects of 4-week P7C3 treatment on cardiac function were evaluated along with molecular signaling changes for phosphorylated protein kinase B (p-AKT), phosphorylated endothelial nitric oxide synthase (p-eNOS), and sirtuin 1 (SIRT1). The cardiac function evaluated by ECG and echocardiography were significantly improved after 4 weeks of P7C3 treatment. Biochemically, higher NADH/NAD+ ratios in diabetic hearts were rescued by P7C3 treatment. Moreover, activities of Nampt and SIRT1 were significantly increased in P7C3-treated diabetic hearts. P7C3 treatment significantly decreased the blood glucose in diabetic mice with 4-week treatment as noted by glucose tolerance test and fasting blood glucose measurements compared with vehicle-treated mice. P7C3 activated Nampt enzymatic activity both in vitro and in the 4-week diabetic mouse hearts, demonstrating the specificity of the small molecule. P7C3 treatment significantly enhanced the expression of cardioprotective signaling of p-AKT, p-eNOS, and Beclin 1 in diabetic hearts. Nampt activator P7C3 allows for decreased infarct size with decreased Troponin I and lactose dehydrogenase (LDH) release, which is beneficial to the heart. Overall, the present study shows that P7C3 activates Nampt and SIRT1 activity and decreases NADH/NAD+ ratio, resulting in improved biochemical signaling providing cardioprotection. SIGNIFICANCE STATEMENT: This study shows that 1-(3,6-Dibromo-carbazol-9-yl)-3-phenylamino-propan-2-ol (P7C3) is effective in treating diabetes and cardiovascular diseases. The novel small molecule is antiarrhythmic and improves the ejection fraction in diabetic hearts. The study successfully demonstrated that P7C3 decreases the infarct size in hearts during myocardial infarction and ischemia-reperfusion injury. Biochemical and cellular signaling show increased NAD+ levels, along with Nampt activity involved in upregulating protective signaling in the diabetic heart. P7C3 has high therapeutic potential for rescuing heart disease.


Assuntos
Diabetes Mellitus Experimental , Infarto do Miocárdio , Animais , Glicemia , Carbazóis , Citocinas/metabolismo , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Diabetes Mellitus Experimental/metabolismo , Masculino , Camundongos , Infarto do Miocárdio/tratamento farmacológico , NAD/metabolismo , Nicotinamida Fosforribosiltransferase , Proteínas Proto-Oncogênicas c-akt , Sirtuína 1/metabolismo
3.
Circulation ; 131(17): 1506-17; discussion 1517, 2015 Apr 28.
Artigo em Inglês | MEDLINE | ID: mdl-25820336

RESUMO

BACKGROUND: Transient preceding brief ischemia provides potent cardioprotection against subsequent long ischemia, termed ischemic preconditioning. Here, we hypothesized that transient short-term hypertrophic stimulation would induce the expression of hypertrophy regression genes and render the heart resistant to subsequent hypertrophic stress, and slow the progression to heart failure, as well. METHODS AND RESULTS: Cardiomyocyte hypertrophy was induced in mice by either transverse aortic constriction or an infusion of phenylephrine, and in neonatal rat ventricular cardiomyocytes by norepinephrine exposures. In the preconditioning groups, hypertrophic stimulation was provided for 1 to 7 days and then withdrawn for several days by either aortic debanding or discontinuing phenylephrine or norepinephrine treatment, followed by subsequent reexposure to the hypertrophic stimulus for the same period as in the control group. One or 6 weeks after transverse aortic constriction, the heart weight/body weight ratio was lower in the preconditioning group than in the control group, whereas the lung weight/body weight ratio was significantly decreased 6 weeks after transverse aortic constriction. Similar results were obtained in mice receiving phenylephrine infusion and neonatal rat ventricular cardiomyocytes stimulated with norepinephrine. Both mRNA and protein expression of S100A8 and S100A9 showed significant upregulation after the removal of hypertrophic stimulation and persisted for 6 weeks in response to reimposition of transverse aortic constriction. The treatment with recombinant S100A8/A9 inhibited norepinephrine-induced myocyte hypertrophy and reduced the expression of calcineurin and NFATc3, but the silencing of S100A8/A9 prevented such changes. CONCLUSIONS: Preconditioning with prohypertrophic factors exerts an antihypertrophic effect and slows the progression of heart failure, indicating the existence of the phenomenon for hypertrophic preconditioning.


Assuntos
Calgranulina A/biossíntese , Calgranulina B/biossíntese , Cardiomiopatia Hipertrófica/patologia , Insuficiência Cardíaca/prevenção & controle , Miócitos Cardíacos/patologia , Animais , Estenose da Valva Aórtica/complicações , Calcineurina/fisiologia , Calgranulina A/genética , Calgranulina B/genética , Calgranulina B/farmacologia , Cardiomiopatia Hipertrófica/complicações , Cardiomiopatia Hipertrófica/genética , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Regulação da Expressão Gênica , Vetores Genéticos , Insuficiência Cardíaca/etiologia , Hipertrofia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/fisiologia , Fenilefrina/farmacologia , Fenilefrina/toxicidade , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Ratos , Proteínas Recombinantes de Fusão/farmacologia
4.
Adv Biol (Weinh) ; : e2400137, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38773896

RESUMO

Aging is associated with a decline in cardiac function. Exercise has been shown to effectively reduce the risks of cardiovascular diseases. Here whether a combination of endurance and resistance exercises can improve cardiac function in aged mice during late life is investigated. Through transcriptome analysis, several signaling pathways activated in the hearts of 22-month-old mice after combined exercise, including cardiac muscle contraction, mitophagy, and longevity regulation are identified. Combined exercise training mitigated age-associated pathological cardiac hypertrophy, reduced oxidative stress, cardiac senescence, and enhanced cardiac function. Upstream stimulatory factor 2 (Usf2) is upregulated in the aged mouse hearts with combined exercise compared to sedentary mice. In the human cardiomyocytes senescent model, overexpression of Usf2 led to anti-senescence effects, while knockdown of Usf2 exacerbated cellular senescence. The results suggest that a combination of endurance and resistance exercises, such as swimming and resistance running, can mitigate age-related pathological cardiac remodeling and cardiac dysfunction in late life. These cardioprotective effects are likely due to the activation of Usf2 and its anti-senescence effect. Therefore, Usf2 can potentially be a novel therapeutic target for mitigating age-related cardiac dysfunction.

5.
Eur Heart J ; 33(24): 3124-33, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21785110

RESUMO

AIMS: We investigated the influence of type one cannabinoid receptor (CB1) deficiency on acute heart failure (AHF) and the underlying mechanism. Acute heart failure syndrome is an important clinical problem because of its high morbidity and mortality rates. Activation of CB1 induces vascular dilation and reinforces the properties of morphine, long-standing therapies for AHF syndrome, but the effect of endogenous CB1 activation on AHF is largely unknown. METHODS AND RESULTS: Acute heart failure mouse model characterized by hypertension and pulmonary oedema was created by using transverse aortic constriction (TAC). Mortality, echocardiography, haemodynamic, morphology, and circulatory catecholamine levels in response to TAC were evaluated in CB1 knockout (KO) and wild-type mice. Type one cannabinoid receptor KO mice had a much higher mortality rate at 1 week after TAC attributable to AHF (65 vs. 11%, P< 0.001). One hour after TAC, CB1 KO mice had significant larger lung weight to body weight ratio (LW/BW, 14.53 + 1.09 mg/g in KO vs. 10.42 + 0.36 mg/g in WT, P < 0.01) and higher plasma epinephrine levels (9720 + 1226 pg/mL vs. 6378 + 832 pg/mL, P < 0.05). Pharmacological activation of CB1 reduced LW/BW in wild-type mice. Administration of epinephrine to wild-type TAC mice significantly increased left ventricular end-diastolic pressure and LW/BW, while CB1 agonists reduced the LW/BW and the plasma levels of catecholamine and increased myocardial activity of AMP-activated protein kinase. CONCLUSION: Endogenous activation of CB1 in mice has cardiac protection in AHF, which is attributable to the inhibition of excessive sympathetic activation.


Assuntos
Insuficiência Cardíaca/etiologia , Hipertensão/complicações , Edema Pulmonar/complicações , Receptor CB1 de Canabinoide/deficiência , Animais , Aorta , Contagem de Células Sanguíneas , Catecolaminas/metabolismo , Células Cultivadas , Epinefrina/farmacologia , Hemodinâmica/efeitos dos fármacos , Ligadura , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Quinases/metabolismo , Vasoconstritores/farmacologia
6.
Front Aging Neurosci ; 15: 1221653, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37577356

RESUMO

Sarcopenia is an age-related, involuntary loss of skeletal muscle mass and strength. Alzheimer's disease (AD) is the most common cause of dementia in elderly adults. To date, no effective cures for sarcopenia and AD are available. Physical and cognitive impairments are two major causes of disability in the elderly population, which severely decrease their quality of life and increase their economic burden. Clinically, sarcopenia is strongly associated with AD. However, the underlying factors for this association remain unknown. Mechanistic studies on muscle-brain crosstalk during cognitive impairment might shed light on new insights and novel therapeutic approaches for combating cognitive decline and AD. In this review, we summarize the latest studies emphasizing the association between sarcopenia and cognitive impairment. The underlying mechanisms involved in muscle-brain crosstalk and the potential implications of such crosstalk are discussed. Finally, future directions for drug development to improve age-related cognitive impairment and AD-related cognitive dysfunction are also explored.

7.
Cells ; 12(14)2023 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-37508502

RESUMO

The failure of muscle to repair after injury during aging may be a major contributor to muscle mass loss. We recently generated muscle progenitor cells (MPCs) from human-induced pluripotent stem-cell (iPSC) cell lines using small molecules, CHIR99021 and Givinostat (Givi-MPCs) sequentially. Here, we test whether the chemokines overexpressed in injured endothelial cells direct MPC migration to the site by binding to their receptor, ITGA4. ITGA4 was heavily expressed in Givi-MPCs. To study the effects on the mobilization of Givi-MPCs, ITGA4 was knocked down by an ITGA4 shRNA lentiviral vector. With and without ITGA4 knocked down, cell migration in vitro and cell mobilization in vivo using aged NOD scid gamma (NSG) mice and mdx/scid mice were analyzed. The migration of shITGA4-Givi-MPCs was significantly impaired, as shown in a wound-healing assay. The knockdown of ITGA4 impaired the migration of Givi-MPCs towards human aortic endothelial cells (HAECs), in which CX3CL1 and VCAM-1 were up-regulated by the treatment of TNF-α compared with scramble ones using a transwell system. MPCs expressing ITGA4 sensed chemokines secreted by endothelial cells at the injury site as a chemoattracting signal to migrate to the injured muscle. The mobilization of Givi-MPCs was mediated by the ligand-receptor interaction, which facilitated their engraftment for repairing the sarcopenic muscle with injury.


Assuntos
Células-Tronco Pluripotentes Induzidas , Músculo Esquelético , Camundongos , Animais , Humanos , Idoso , Músculo Esquelético/metabolismo , Células Endoteliais/metabolismo , Camundongos Endogâmicos mdx , Envelhecimento , Quimiocinas/metabolismo
8.
Crit Care Med ; 40(11): 3026-33, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22926332

RESUMO

OBJECTIVES: To test the hypothesis that resveratrol would improve cardiac remodeling by inhibiting the detrimental effects of fractalkine. We previously reported that fractalkine exacerbates heart failure. Furthermore, this study sought to determine whether resveratrol targets fractalkine to improve myocardial ischemia and cardiac remodeling. DESIGN: Randomized and controlled laboratory investigation. SETTING: Research laboratory. SUBJECTS: Neonatal rat cardiac cells and C57BL/6 mice. INTERVENTIONS: Cardiac cells were treated with recombinant mouse soluble fractalkine for 24 hrs or pretreated with 25 µM resveratrol. Cardiomyocytes were exposed to anoxia/reoxygenation, H2O2, or pretreatment with resveratrol. Ex vivo murine hearts were perfusioned with soluble fractalkine or pretreated with resveratrol after global ischemia. Mice were subjected to the left coronary artery ligation to induce myocardial infarction and randomized to treatment with resveratrol or vehicle alone for 42 days. MEASUREMENTS AND MAIN RESULTS: In a murine myocardial infarction model, we found that resveratrol increased survival and delayed the progression of cardiac remodeling evaluated by serial echocardiography. At 6 wks, the heart weight/body weight ratio, lung weight/body weight ratio, and old infarct size were significantly smaller in resveratrol-treated mice than in untreated myocardial infarction mice. In cultures of neonatal rat cells, exposure to soluble fractalkine increased the atrial natriuretic peptide expression by cardiomyocytes, matrix metalloproteinase-9 and procollagen expression by fibroblasts, and intercellular adhesion molecule-1 expression by microvascular endothelial cells, while it decreased autophagy in cardiomyocytes. All these effects were blocked by coculture with resveratrol. The methyl thiazolyl tetrazolium assay showed that soluble fractalkine reduced the viability of cultured cardiomyocytes during exposure to anoxia/reoxygenation or H2O2, while pretreatment with resveratrol blocked this effect. Perfusion of ex vivo murine hearts with soluble fractalkine after global ischemia led to an increase of infarct size, which was prevented by pretreatment with resveratrol. CONCLUSION: Resveratrol alleviates the deleterious effects of fractalkine on myocardial ischemia and thus reduces subsequent cardiac remodeling.


Assuntos
Quimiocina CX3CL1/efeitos adversos , Insuficiência Cardíaca/tratamento farmacológico , Isquemia Miocárdica/tratamento farmacológico , Inibidores da Agregação Plaquetária/farmacologia , Estilbenos/uso terapêutico , Animais , Animais Recém-Nascidos , Quimiocina CX3CL1/antagonistas & inibidores , Modelos Animais de Doenças , Eletrocardiografia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/fisiopatologia , Distribuição Aleatória , Ratos , Resveratrol , Remodelação Ventricular/efeitos dos fármacos
9.
Front Cell Dev Biol ; 10: 936084, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35813193

RESUMO

Organoid technology has significantly advanced in recent years and revolutionized the field for generation of organs using in vitro systems (a.k.a "organs in a dish"). The use of pluripotent stem cells or tissue derived cells for generating a 3-dimensional culture system to recapitulate the architecture and function of the organ is central in achieving and improving organoid systems. Unlike most organs in the body, very little progress has been made in cardiac organoid due to its structural complexity and vascularization. In this review, we will discuss the current applications of human cardiac organoids for cardiac disease modeling, drug discovery, drug cardiotoxicity testing, and clinical applications.

10.
Front Pharmacol ; 13: 848310, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35370759

RESUMO

The prognosis of cardiorenal dysfunction induced by diabetes mellitus (DM), which belongs to cardiorenal syndrome type 5, is poor and its pathogenesis remains elusive. We have reported that CX3CL1 exacerbated heart failure and direct inhibition of CX3CL1 improved cardiac function. Emerging evidence supports that CX3CL1 is involved in renal impairment. Here we attempt to clarify whether CX3CL1 might be a therapeutic target for cardiorenal dysfunction in diabetes. We found that cardiac and renal CX3CL1 protein levels were significantly increased in both streptozotocin-induced diabetic mice and in non-obese diabetic mice, and that hyperglycemia led to persistent CX3CL1 expression in the heart and kidneys even after it was controlled by insulin. In cultured cardiac and renal cells, soluble CX3CL1 accelerated mitochondrial-dependent apoptosis via activation of the RhoA/ROCK1-Bax signaling pathway and promoted fibrosis through cellular phenotypic trans-differentiation mediated by the TGF-ß/Smad pathway. In the two diabetic mouse models, knockout of CX3CL1 receptor CX3CR1 or treatment with an CX3CL1 neutralizing antibody significantly improved cardiorenal dysfunction by inhibiting apoptosis, mitochondrial dysfunction, and fibrosis. Moreover, sodium glucose cotransporter 2 inhibitor canagliflozin significantly downregulated cardiac and renal CX3CL1 expression and improved cardiorenal dysfunction. These findings indicate that CX3CL1 could be a new therapeutic target for diabetes-induced cardiorenal dysfunction.

11.
Stem Cell Res Ther ; 12(1): 131, 2021 02 12.
Artigo em Inglês | MEDLINE | ID: mdl-33579366

RESUMO

BACKGROUND: Duchenne muscular dystrophy (DMD) is caused by mutations of the gene that encodes the protein dystrophin. A loss of dystrophin leads to severe and progressive muscle wasting in both skeletal and heart muscles. Human induced pluripotent stem cells (hiPSCs) and their derivatives offer important opportunities to treat a number of diseases. Here, we investigated whether givinostat (Givi), a histone deacetylase inhibitor, with muscle differentiation properties could reprogram hiPSCs into muscle progenitor cells (MPC) for DMD treatment. METHODS: MPC were generated from hiPSCs by treatment with CHIR99021 and givinostat called Givi-MPC or with CHIR99021 and fibroblast growth factor as control-MPC. The proliferation and migration capacity were investigated by CCK-8, colony, and migration assays. Engraftment, pathological changes, and restoration of dystrophin were evaluated by in vivo transplantation of MPC. Conditioned medium from cultured MPC was collected and analyzed for extracellular vesicles (EVs). RESULTS: Givi-MPC exhibited superior proliferation and migration capacity compared to control-MPC. Givi-MPC produced less reactive oxygen species (ROS) after oxidative stress and insignificant expression of IL6 after TNF-α stimulation. Upon transplantation in cardiotoxin (CTX)-injured hind limb of Mdx/SCID mice, the Givi-MPC showed robust engraftment and restored dystrophin in the treated muscle than in those treated with control-MPC or human myoblasts. Givi-MPC significantly limited infiltration of inflammatory cells and reduced muscle necrosis and fibrosis. Additionally, Givi-MPC seeded the stem cell pool in the treated muscle. Moreover, EVs released from Givi-MPC were enriched in several miRNAs related to myoangiogenesis including miR-181a, miR-17, miR-210 and miR-107, and miR-19b compared with EVs from human myoblasts. CONCLUSIONS: It is concluded that hiPSCs reprogrammed into MPC by givinostat possessing anti-oxidative, anti-inflammatory, and muscle gene-promoting properties effectively repaired injured muscle and restored dystrophin in the injured muscle.


Assuntos
Células-Tronco Pluripotentes Induzidas , Distrofia Muscular de Duchenne , Animais , Carbamatos , Distrofina/genética , Humanos , Camundongos , Camundongos Endogâmicos mdx , Camundongos SCID , Fibras Musculares Esqueléticas , Músculo Esquelético , Distrofia Muscular de Duchenne/genética , Mioblastos
12.
Quant Imaging Med Surg ; 11(4): 1600-1612, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33816194

RESUMO

BACKGROUND: The segmentation of cardiac medical images is a crucial step for calculating clinical indices such as wall thickness, ventricular volume, and ejection fraction. METHODS: In this study, we introduce a method named LsUnet that combines multi-channel, fully convolutional neural network, and annular shape level-set methods for efficiently segmenting cardiac cine magnetic resonance (MR) images. In this method, the multi-channel deep learning algorithm is applied to train the segmentation task to extract the left ventricle (LV) endocardial and epicardial contours. Next, the segmentation contours from the multi-channel deep learning method are incorporated into a level-set formulation, which is dedicated explicitly to detecting annular shapes to assure the segmentation's accuracy and robustness. RESULTS: The proposed automatic approach was evaluated on 95 volumes (total 1,076 slices, ~80% as for training datasets, ~20% 2D as for testing datasets). This combined multi-channel deep learning and annular shape level-set segmentation method achieved high accuracy with average Dice values reaching 92.15% and 95.42% for LV endocardium and epicardium delineation, respectively, in comparison to the reference standard (the manual segmentation). CONCLUSIONS: A novel method for fully automatic segmentation of the LV endocardium and epicardium from different MRI datasets is presented. The proposed workflow is accurate and robust compared to the reference and other state-of-the-art methods.

13.
Biochem Biophys Res Commun ; 393(3): 519-25, 2010 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-20152812

RESUMO

This study was aimed to investigate whether the antihypertrophic effects of adiponectin in murine hearts are associated with the modulation of HB-EGF signaling. We determined the myocardial expressions of adiponectin and adiponectin receptors, brain natriuretic peptide (BNP), and HB-EGF in normal and hypertrophied hearts of adiponectin knockout mice or wild-type mice with transverse aortic constriction (TAC). Then, we observed the effects of adiponectin on cardiac hypertrophy and HB-EGF signaling in cultured neonatal rat cardiomyocytes and whole hearts of adiponectin-null mice. The myocardial mRNA and protein expressions of adiponectin in the hypertrophied hearts were significantly downregulated, and the mRNA expression of adiponectin was inversely correlated with the heart-to-body weight ratio, BNP, and HB-EGF. The TAC-induced cardiac hypertrophy and EGF receptor (EGFR) activation in the adiponectin knockout mice were significantly greater than those in the wild-type mice. Furthermore, in vitro experiments revealed that adiponectin inhibited HB-EGF-stimulated protein synthesis, HB-EGF shedding, and EGFR phosphorylation. We conclude that the inhibition of HB-EGF mediated EGFR activation is one of the alternative mechanisms for the antihypertrophic action of adiponectin.


Assuntos
Adiponectina/metabolismo , Cardiomegalia/patologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Miocárdio/patologia , Miócitos Cardíacos/patologia , Adiponectina/genética , Adiponectina/farmacologia , Animais , Cardiomegalia/metabolismo , Cardiomegalia/prevenção & controle , Linhagem Celular , Ventrículos do Coração/metabolismo , Ventrículos do Coração/patologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Peptídeo Natriurético Encefálico/metabolismo , Tamanho do Órgão , Ratos , Transdução de Sinais
14.
Front Cell Dev Biol ; 8: 11, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32154243

RESUMO

Cardiac mesenchymal stem cells (C-MSCs) are a novel mesenchymal stem cell (MSC) subpopulation derived from cardiac tissue, which are reported to be responsible for cardiac regeneration. Notch signaling is believed to aid in cardiac repair following myocardial injury. In this study, we have investigated the role of extracellular vesicles (EVs) from Notch1 engineered C-MSCs on angiogenesis and cardiomyocyte (CM) proliferation in ischemic myocardium. C-MSCs were isolated from Notch1flox mice (C-MSCNotch1 FF). Notch1 gene deletion was accomplished by adenoviral vector-mediated Cre recombination, and Notch1 overexpression was achieved by overexpression of Notch1 intracellular domain (N1ICD). EVs were isolated by using the size exclusion column method. Proteomic composition of EV was carried out by mass spectrometry. A mouse myocardial infarction (MI) model was generated by permanent left anterior descending (LAD) coronary artery ligation. Intramyocardial transplantation of Notch1 knockout C-MSCs (C-MSCsNotch1 KO) did not have any effect on cardiac function and scar size. On the other hand, transplantation of N1ICD-overexpressing C-MSCs (C-MSCsN1ICD) showed significant improvement in cardiac function and attenuation of fibrosis as compared to the control (PBS) group and non-modified C-MSC groups. C-MSCsN1ICD differentiated into smooth muscle cells and formed new vessels. Proteomics profiling identified several proteins, such as lysyl oxidase homolog-2 and biglycan, as highly enriched proteins in EV-C-MSCsN1ICD. Go term analysis indicated that EV-C-MSCsN1ICD were enriched with bioactive factors, potent pro-repair proteins responsible for cell migration and proliferation. EV-C-MSCs Notch1FF and EV-C-MSCsN1ICD were strongly proangiogenic under both in vitro and in vivo conditions. EV-C-MSCsN1ICD caused dense tube formation in vitro and increased neovasculogenesis in the peri-infarct area in vivo. Furthermore, EV-C-MSCsN1ICD attenuated endothelial cell (EC) and CM apoptosis under oxidative stress and ischemic injury. Similarly, EV-C-MSCNotch1 FF and EV-C-MSCN1ICD treatment improved cardiac function and decreased fibrosis in mice post-MI. EV-C-MSCsN1ICD were very effective in improving cardiac function and decreasing fibrosis. Notch1 signaling is a strong stimulus for cardiac regeneration by C-MSCs. EVs secreted by Notch1-overexpressing C-MSCs were highly effective in preventing cell death, promoting angiogenesis and CM proliferation, and restoring cardiac function post-MI. Overall, these results suggest that Notch1 overexpression may further enhance the effectiveness of EVs secreted by C-MSCs in cell-free therapy.

15.
Mater Sci Eng C Mater Biol Appl ; 112: 110896, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32409053

RESUMO

Cell therapy is a promising strategy in which living cells or cellular materials are delivered to treat a variety of diseases. Here, we developed an electrospray bioprinting method to rapidly generate cell-laden hydrogel microspheres, which limit the migration of the captured cells and provide an immunologically privileged microenvironment for cell survival in vivo. Currently, therapeutic angiogenesis aims to induce collateral vessel formation after limb ischemia. However, the clinical application of gene and cell therapy has been impeded by concerns regarding its inefficacy, as well as the associated risk of immunogenicity and oncogenicity. In this study, hydrogel microspheres encapsulating VEGF-overexpressing HEK293T cells showed good safety via subcutaneously injecting into male C57BL/6 mice. In addition, these cell-modified microspheres effectively promoted angiogenesis in a mouse hind-limb ischemia model. Therefore, we demonstrated the great therapeutic potential of this approach to induce angiogenesis in limb ischemia, indicating that bioprinting has a bright future in cell therapy.


Assuntos
Materiais Biocompatíveis/química , Microesferas , Neovascularização Fisiológica/fisiologia , Alginatos/química , Animais , Materiais Biocompatíveis/farmacologia , Materiais Biocompatíveis/uso terapêutico , Bioimpressão , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Modelos Animais de Doenças , Células HEK293 , Membro Posterior/patologia , Células Endoteliais da Veia Umbilical Humana , Humanos , Hidrogéis/química , Isquemia/terapia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neovascularização Fisiológica/efeitos dos fármacos , Alicerces Teciduais/química , Fator A de Crescimento do Endotélio Vascular/metabolismo
16.
Stem Cells Int ; 2019: 3726392, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31814833

RESUMO

Cardiac stem cell therapy offers the potential to ameliorate postinfarction remodeling and development of heart failure but requires optimization of cell-based approaches. Cardiac progenitor cells (CPCs) induction by ISX-9, a small molecule possessing antioxidant, prosurvival, and regenerative properties, represents an attractive potential approach for cell-based cardiac regenerative therapy. Here, we report that extracellular vesicles (EV) secreted by ISX-9-induced CPCs (EV-CPCISX-9) faithfully recapitulate the beneficial effects of their parent CPCs with regard to postinfarction remodeling. These EV contain a distinct repertoire of biologically active miRNAs that promoted angiogenesis and proliferation of cardiomyocytes while ameliorating fibrosis in the infarcted heart. Amongst the highly enriched miRNAs, miR-373 was strongly antifibrotic, targeting 2 key fibrogenic genes, GDF-11 and ROCK-2. miR-373 mimic itself was highly efficacious in preventing scar formation in the infarcted myocardium. Together, these novel findings have important implications with regard to prevention of postinfarction remodeling.

17.
Med Phys ; 46(1): 180-189, 2019 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-30352129

RESUMO

PURPOSE: Segmentation of cardiac medical images, an important step in measuring cardiac function, is usually performed either manually or semiautomatically. Fully automatic segmentation of the left ventricle (LV), the right ventricle (RV) as well as the myocardium of three-dimensional (3D) magnetic resonance (MR) images throughout the entire cardiac cycle (four-dimensional, 4D), remains challenging. This study proposes a deformable-based segmentation methodology for efficiently segmenting 4D (3D + t) cardiac MR images. METHODS: The proposed methodology first used the Hough transform and the local Gaussian distribution method (LGD) to segment the LV endocardial contours from cardiac MR images. Following this, a novel level set-based shape prior method was applied to generate the LV epicardial contours and the RV boundary. RESULTS: This automatic image segmentation approach has been applied to studies on 17 subjects. The results demonstrated that the proposed method was efficient compared to manual segmentation, achieving a segmentation accuracy with average Dice values of 88.62 ± 5.47%, 87.35 ± 7.26%, and 82.63 ± 6.22% for the LV endocardial, LV epicardial, and RV contours, respectively. CONCLUSIONS: We have presented a method for accurate LV and RV segmentation. Compared to three existing methods, the proposed method can successfully segment the LV and yield the highest Dice value. This makes it an option for clinical assessment of the volume, size, and thickness of the ventricles.


Assuntos
Coração/diagnóstico por imagem , Coração/fisiologia , Imageamento Tridimensional/métodos , Imageamento por Ressonância Magnética , Automação , Ventrículos do Coração/diagnóstico por imagem , Humanos
18.
Shock ; 50(6): 627-639, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-29485473

RESUMO

Cardiac progenitor cells (CPCs) being multipotent offer a promising source for cardiac repair due to their ability to proliferate and multiply into cardiac lineage cells. Here, we explored a novel strategy for human CPCs generation from human induced pluripotent stem cells (hiPSCs) using a cardiogenic small molecule, isoxazole (ISX-9) and their ability to grow in the scar tissue for functional improvement in the infarcted myocardium. CPCs were induced from hiPSCs with ISX-9. CPCs were characterized by immunocytochemistry and RT-PCR. The CPC survival and differentiation in the infarcted hearts were determined by in vivo transplantation in immunodeficient mice following left anterior descending artery ligation and their effects were determined on fibrosis and functional improvement. ISX-9 simultaneously induced expression of cardiac transcription factors, NK2 homeobox 5, islet-1, GATA binding protein 4, myocyte enhancer factor-2 in hiPSCs within 3 days of treatment and successfully differentiated into three cardiac lineages in vitro. Messenger RNA and microRNA-sequencing results showed that ISX-9 targeted multiple cardiac differentiation, proliferation signaling pathways and upregulated myogenesis and cardiac hypertrophy related-microRNA. ISX-9 activated multiple pathways including transforming growth factor ß induced epithelial-mesenchymal transition signaling, canonical, and non-canonical Wnt signaling at different stages of cardiac differentiation. CPCs transplantation promoted myoangiogenesis, attenuated fibrosis, and led to functional improvement in treated mice.


Assuntos
Fibrose/terapia , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Infarto do Miocárdio/terapia , Miócitos Cardíacos/citologia , Animais , Diferenciação Celular/efeitos dos fármacos , Modelos Animais de Doenças , Fibrose/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Isoxazóis/farmacologia , Camundongos , Infarto do Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transplante de Células-Tronco , Tiofenos/farmacologia
19.
Int J Cardiol ; 243: 367-373, 2017 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-28747035

RESUMO

BACKGROUND: Interleukin-32 (IL-32) is a newly discovered proinflammatory cytokine. However, there are limited data regarding IL-32 as a biomarker for heart failure (HF). In this study, we assessed the prognostic value of IL-32 in patients with chronic HF after myocardial infarction (MI). METHODS AND RESULTS: Over a period of 1.8years, we prospectively enrolled 100 patients with chronic HF after MI. IL-32, NT-proBNP, Matrix metallopeptidase 9 (MMP-9), procollagen type I (PI) and type III (PIII) were measured at baseline. Study endpoint was adverse cardiac events. High IL-32 levels were associated with numerous factors that are related to deteriorate cardiac function and cardiac fibrosis. Strong expression of IL-32 was detected in human cardiomyocytes from HF tissue. ROC curve revealed the area under the curve of IL-32 for predicting negative outcome of HF was 0.72 (95% CI: 0.60-0.83, P<0.01). Kaplan-Meier statistics showed that the risk of adverse cardiac event was 5.75 fold (hazard ratio 5.75, 95% CI 1.53-21.58, P=0.009), which increased in the highest quartile (>296pg/mL). Cox regression analysis revealed IL-32 was an independent predictor for cardiac events (hazard ratio 2.78, 95% CI 1.02-7.57, P=0.046). Recombinant IL-32 significantly exacerbated infarct size in a mouse model of MI. IL-32 upregulated expression of MMP-9, PIII and transforming growth factor beta in rat fibroblasts. CONCLUSION: IL-32 might be a novel predictor of adverse cardiac event in patients with HF after MI. The pro-fibrotic effect of IL-32 may contribute to adverse cardiac remodeling and progression to HF.


Assuntos
Insuficiência Cardíaca/sangue , Insuficiência Cardíaca/diagnóstico , Interleucinas/sangue , Infarto do Miocárdio/sangue , Infarto do Miocárdio/diagnóstico , Idoso , Animais , Biomarcadores/sangue , Células Cultivadas , Feminino , Seguimentos , Insuficiência Cardíaca/epidemiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Infarto do Miocárdio/epidemiologia , Miócitos Cardíacos/metabolismo , Prognóstico , Estudos Prospectivos , Ratos
20.
Oncotarget ; 7(40): 64649-64664, 2016 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-27579618

RESUMO

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.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Fibroblastos/fisiologia , Hipertrofia Ventricular Esquerda/genética , Infarto do Miocárdio/metabolismo , Miocárdio/patologia , Traumatismo por Reperfusão/metabolismo , beta Catenina/metabolismo , Angiotensina II/metabolismo , Animais , Células Cultivadas , Colágeno/metabolismo , Dependovirus/genética , Fator de Crescimento de Fibroblastos 23 , Fatores de Crescimento de Fibroblastos/genética , Fibrose , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/genética , Fenilefrina/metabolismo , RNA Interferente Pequeno/genética , Ratos , Ratos Sprague-Dawley , Receptor Tipo 4 de Fator de Crescimento de Fibroblastos/sangue , Traumatismo por Reperfusão/genética , Fator de Crescimento Transformador beta/metabolismo , Regulação para Cima , beta Catenina/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa