Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.798
Filtrar
1.
Int J Mol Sci ; 24(21)2023 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-37958542

RESUMO

One of the largest challenges to the implementation of cardiac cell therapy is identifying selective reparative targets to enhance stem/progenitor cell therapeutic efficacy. In this work, we hypothesized that such a target could be an urokinase-type plasminogen activator receptor (uPAR)-a glycosyl-phosphatidyl-inositol-anchored membrane protein, interacting with urokinase. uPAR is able to form complexes with various transmembrane proteins such as integrins, activating intracellular signaling pathway and thus regulating multiple cell functions. We focused on studying the CD117+ population of cardiac mesenchymal progenitor cells (MPCs), expressing uPAR on their surface. It was found that the number of CD117+ MPCs in the heart of the uPAR-/- mice is lower, as well as their ability to proliferate in vitro compared with cells from wild-type animals. Knockdown of uPAR in CD117+ MPCs of wild-type animals was accompanied by a decrease in survival rate and Akt signaling pathway activity and by an increase in the level of caspase activity in these cells. That suggests the role of uPAR in supporting cell survival. After intramyocardial transplantation of uPAR(-) MPCs, reduced cell retention and angiogenesis stimulation were observed in mice with myocardial infarction model compared to uPAR(+) cells transplantation. Taken together, the present results appear to prove a novel mechanism of uPAR action in maintaining the survival and angiogenic properties of CD117+ MPCs. These results emphasize the importance of the uPAR as a potential pharmacological target for the regulation of reparative properties of myocardial mesenchymal progenitor cells.


Assuntos
Células-Tronco Mesenquimais , Miocárdio , Receptores de Ativador de Plasminogênio Tipo Uroquinase , Animais , Camundongos , Integrinas , Células-Tronco Mesenquimais/metabolismo , Receptores de Ativador de Plasminogênio Tipo Uroquinase/genética , Receptores de Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Transdução de Sinais , Ativador de Plasminogênio Tipo Uroquinase/genética , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Miocárdio/citologia
2.
Signal Transduct Target Ther ; 8(1): 114, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36918543

RESUMO

Cardiac aging is evident by a reduction in function which subsequently contributes to heart failure. The metabolic microenvironment has been identified as a hallmark of malignancy, but recent studies have shed light on its role in cardiovascular diseases (CVDs). Various metabolic pathways in cardiomyocytes and noncardiomyocytes determine cellular senescence in the aging heart. Metabolic alteration is a common process throughout cardiac degeneration. Importantly, the involvement of cellular senescence in cardiac injuries, including heart failure and myocardial ischemia and infarction, has been reported. However, metabolic complexity among human aging hearts hinders the development of strategies that targets metabolic susceptibility. Advances over the past decade have linked cellular senescence and function with their metabolic reprogramming pathway in cardiac aging, including autophagy, oxidative stress, epigenetic modifications, chronic inflammation, and myocyte systolic phenotype regulation. In addition, metabolic status is involved in crucial aspects of myocardial biology, from fibrosis to hypertrophy and chronic inflammation. However, further elucidation of the metabolism involvement in cardiac degeneration is still needed. Thus, deciphering the mechanisms underlying how metabolic reprogramming impacts cardiac aging is thought to contribute to the novel interventions to protect or even restore cardiac function in aging hearts. Here, we summarize emerging concepts about metabolic landscapes of cardiac aging, with specific focuses on why metabolic profile alters during cardiac degeneration and how we could utilize the current knowledge to improve the management of cardiac aging.


Assuntos
Envelhecimento , Senescência Celular , Cardiopatias , Miócitos Cardíacos , Humanos , Envelhecimento/metabolismo , Envelhecimento/patologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Biologia Molecular , Miocárdio/citologia , Miocárdio/metabolismo , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Senescência Celular/fisiologia , Cardiopatias/metabolismo , Cardiopatias/patologia
3.
J Cell Biol ; 221(6)2022 06 06.
Artigo em Inglês | MEDLINE | ID: mdl-35482005

RESUMO

Tissue-resident macrophages play essential functions in the maintenance of tissue homeostasis and repair. Recently, the endocardium has been reported as a de novo hemogenic site for the contribution of hematopoietic cells, including cardiac macrophages, during embryogenesis. These observations challenge the current consensus that hematopoiesis originates from the hemogenic endothelium within the yolk sac and dorsal aorta. Whether the developing endocardium has such a hemogenic potential requires further investigation. Here, we generated new genetic tools to trace endocardial cells and reassessed their potential contribution to hematopoietic cells in the developing heart. Fate-mapping analyses revealed that the endocardium contributed minimally to cardiac macrophages and circulating blood cells. Instead, cardiac macrophages were mainly derived from the endothelium during primitive/transient definitive (yolk sac) and definitive (dorsal aorta) hematopoiesis. Our findings refute the concept of endocardial hematopoiesis, suggesting that the developing endocardium gives rise minimally to hematopoietic cells, including cardiac macrophages.


Assuntos
Linhagem da Célula , Coração , Macrófagos , Miocárdio , Animais , Aorta/citologia , Endocárdio/citologia , Coração/embriologia , Hematopoese/genética , Miocárdio/citologia , Saco Vitelino/citologia
4.
Oxid Med Cell Longev ; 2022: 7664290, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35242277

RESUMO

Human cardiac fibroblasts (HCFs) play key roles in normal physiological functions and pathological processes in the heart. Our recent study has found that, in HCFs, sphingosine 1-phosphate (S1P) can upregulate the expression of cyclooxygenase-2 (COX-2) leading to prostaglandin E2 (PGE2) generation mediated by S1P receptors/PKCα/MAPKs cascade-dependent activation of NF-κB. Alternatively, G protein-coupled receptor- (GPCR-) mediated transactivation of receptor tyrosine kinases (RTKs) has been proved to induce inflammatory responses. However, whether GPCR-mediated transactivation of RTKs participated in the COX-2/PGE2 system induced by S1P is still unclear in HCFs. We hypothesize that GPCR-mediated transactivation of RTKs-dependent signaling cascade is involved in S1P-induced responses. This study is aimed at exploring the comprehensive mechanisms of S1P-promoted COX-2/PGE2 expression and apoptotic effects on HCFs. Here, we used pharmacological inhibitors and transfection with siRNA to evaluate whether matrix metalloprotease (MMP)2/9, heparin-binding- (HB-) epidermal growth factor (EGF), EGF receptor (EGFR), PI3K/Akt, MAPKs, and transcription factor AP-1 participated in the S1P-induced COX-2/PGE2 system determined by Western blotting, real-time polymerase chain reaction (RT-PCR), chromatin immunoprecipitation (ChIP), and promoter-reporter assays in HCFs. Our results showed that S1PR1/3 activated by S1P coupled to Gq- and Gi-mediated MMP9 activity to stimulate EGFR/PI3K/Akt/MAPKs/AP-1-dependent activity of transcription to upregulate COX-2 accompanied with PGE2 production, leading to stimulation of caspase-3 activity and apoptosis. Moreover, S1P-enhanced c-Jun bound to COX-2 promoters on its corresponding binding sites, which was attenuated by these inhibitors of protein kinases, determined by a ChIP assay. These results concluded that transactivation of MMP9/EGFR-mediated PI3K/Akt/MAPKs-dependent AP-1 activity was involved in the upregulation of the COX-2/PGE2 system induced by S1P, in turn leading to apoptosis in HCFs.


Assuntos
Apoptose/efeitos dos fármacos , Ciclo-Oxigenase 2/metabolismo , Dinoprostona/metabolismo , Fibroblastos/metabolismo , Lisofosfolipídeos/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Metaloproteinase 9 da Matriz/metabolismo , Miocárdio/citologia , Esfingosina/análogos & derivados , Ativação Transcricional/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Ciclo-Oxigenase 2/genética , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Sistema de Sinalização das MAP Quinases/genética , Metaloproteinase 9 da Matriz/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Esfingosina/farmacologia , Fator de Transcrição AP-1/metabolismo , Transfecção
5.
Dev Cell ; 57(4): 424-439, 2022 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-35231426

RESUMO

Cardiovascular disease is a leading cause of death worldwide, and thus there remains great interest in regenerative approaches to treat heart failure. In the past 20 years, the field of heart regeneration has entered a renaissance period with remarkable progress in the understanding of endogenous heart regeneration, stem cell differentiation for exogenous cell therapy, and cell-delivery methods. In this review, we highlight how this new understanding can lead to viable strategies for human therapy. For the near term, drugs, electrical and mechanical devices, and heart transplantation will remain mainstays of cardiac therapies, but eventually regenerative therapies based on fundamental regenerative biology may offer more permanent solutions for patients with heart failure.


Assuntos
Coração , Miocárdio/citologia , Miócitos Cardíacos/citologia , Regeneração/fisiologia , Medicina Regenerativa , Animais , Coração/fisiologia , Humanos , Medicina Regenerativa/métodos , Transplante de Células-Tronco/métodos
6.
J Extracell Vesicles ; 11(1): e12178, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-35005847

RESUMO

Extracellular vesicles (EVs) are potent signalling mediators. Although interest in EV translation is ever-increasing, development efforts are hampered by the inability to reliably assess the uptake of EVs and their RNA cargo. Here, we establish a novel qPCR-based method for the detection of unmodified EVS using an RNA Tracer (DUST). In this proof-of-concept study we use a human-specific Y RNA-derived small RNA (YsRNA) we dub "NT4" that is enriched in cardiosphere-derived cell small EVs (CDC-sEVs). The assay is robust, sensitive, and reproducible. Intravenously administered CDC-sEVs accumulated primarily in the heart on a per mg basis. Cardiac injury enhanced EV uptake in the heart, liver, and brain. Inhibition of EV docking by heparin suppressed uptake variably, while inhibition of endocytosis attenuated uptake in all organs. In vitro, EVs were uptaken more efficiently by macrophages, endothelial cells, and cardiac fibroblasts compared to cardiomyocytes. These findings demonstrate the utility of DUST to assess uptake of EVs in vivo and in vitro.


Assuntos
Vesículas Extracelulares/metabolismo , Miocárdio/metabolismo , Pequeno RNA não Traduzido/metabolismo , Animais , Células Endoteliais/metabolismo , Fibroblastos/metabolismo , Traumatismos Cardíacos/metabolismo , Humanos , Macrófagos/metabolismo , Camundongos , Miocárdio/citologia , Miócitos Cardíacos/metabolismo , Pequeno RNA não Traduzido/administração & dosagem , Pequeno RNA não Traduzido/genética , Distribuição Tecidual
7.
Eur J Clin Invest ; 52(4): e13712, 2022 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-34783022

RESUMO

BACKGROUND: Inhibition of histone deacetylases (HDACs) attenuates cardiac fibrosis. In this study, we evaluated whether the inhibition of class I HDACs can attenuate angiotensin II (ANG II)-induced fibrogenesis and mitochondrial malfunction through its effects on reactive oxygen species (ROS) and calcium dysregulation in human cardiac fibroblasts (CFs). METHODS: Seahorse XF24 extracellular flux analyser, fluorescence staining, Western blotting, HDAC activity assays and Transwell migration assay were used to study mitochondrial respiration, adenosine triphosphate (ATP) production, mitochondrial calcium uptake and ROS, HDAC expression and activity and fibroblast activity in CFs without (control) or with ANG II (100 nM) and/or MS-275 (HDAC class 1 inhibitor, 10 µM) for 24 h. RESULTS: ANG II increased HDAC activity without changing protein expression in CFs. Compared with controls, ANG II-treated CFs had greater migration activity, higher ATP production, maximal respiration and spare capacity with higher mitochondrial Ca2+ uptake and ROS generation, which was attenuated by the administration of MS-275. ANG II activated CFs by increasing mitochondrial calcium content and ATP production, which may be caused by increased HDAC activity. Inhibition of HDAC1 attenuated the effects of ANG II by reducing mitochondrial ROS generation and calcium overload. CONCLUSIONS: Modulating mitochondrial function by regulation of HDAC may be a novel strategy for controlling CF activity.


Assuntos
Angiotensina II/fisiologia , Movimento Celular/fisiologia , Fibroblastos/fisiologia , Histona Desacetilases/fisiologia , Mitocôndrias/fisiologia , Miocárdio/citologia , Angiotensina II/efeitos dos fármacos , Cálcio/metabolismo , Células Cultivadas , Inibidores de Histona Desacetilases/farmacologia , Humanos , Mitocôndrias/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo
8.
J Cardiovasc Pharmacol ; 79(3): 296-303, 2022 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-34775426

RESUMO

ABSTRACT: Heart failure is mainly caused by a decline in the systolic function of the heart. Long noncoding RNAs are related to cardiac diseases. This study aimed to explore the effects of long noncoding RNAs testis development related gene 1 (TDRG1) on the fibrogenesis and inflammatory response of transforming growth factor-beta1 (TGF-ß1)-stimulated human cardiac fibroblasts (HCFs). Levels of proinflammatory cytokines were evaluated by enzyme-linked immunosorbent assay. Reverse-transcription quantitative polymerase chain reaction was applied to reveal the expression levels of TDRG1, miR-605-3p, and tumor necrosis factor receptor superfamily (TNFRSF21). Western blot analysis was prepared to detect protein levels of TNFRSF21 and fibrosis-related genes. Luciferase reporter assay was conducted for confirming the interaction between miR-605-3p and TDRG1/TNFRSF21. We found that TGF-ß1-stimulated HCFs showed high concentrations of proinflammatory cytokines and increased protein levels of fibrosis-related genes, suggesting the dysfunctions of TGF-ß1-stimulated HCFs. In addition, TDRG1 was upregulated in TGF-ß1-stimulated HCFs. We found that interfering with TDRG1 alleviated dysfunctions of TGF-ß1-stimulated HCFs. Moreover, TDRG1 bound with miR-605-3p. MiR-605-3p exerted the antifibrogenic and anti-inflammatory effects in TGF-ß1-treated HCFs. As a target gene of miR-605-3p, TNFRSF21 reversed the antifibrogenic and anti-inflammatory effects of TDRG1 knockdown in TGF-ß1-treated HCFs. Overall, our study confirmed that TDRG1 aggravates fibrogenesis and inflammatory response in TGF-ß1-treated HCFs via the miR-605-3p/TNFRSF21 axis.


Assuntos
MicroRNAs , Miocárdio , RNA Longo não Codificante , Receptores do Fator de Necrose Tumoral , Anti-Inflamatórios/farmacologia , Proliferação de Células , Fibroblastos/patologia , Fibrose , Humanos , MicroRNAs/metabolismo , Miocárdio/citologia , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Receptores do Fator de Necrose Tumoral/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
9.
Eur J Pharm Biopharm ; 170: 187-196, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-34968647

RESUMO

Since the discovery of the beneficial therapeutical effects of extracellular vesicles (EVs), these agents have been attracting great interest as next-generation therapies. EVs are nanosized membrane bodies secreted by all types of cells that mediate cell-cell communication. Although the classification of different subpopulations of EVs can be complex, they are broadly divided into microvesicles and exosomes based on their biogenesis and in large and small EVs based on their size. As this is an emerging field, current investigations are focused on basic aspects such as the more convenient method for EV isolation. In the present paper, we used cardiac progenitor cells (CPCs) to study and compare different cell culture conditions for EV isolation as well as two of the most commonly employed purification methods: ultracentrifugation (UC) and size-exclusion chromatography (SEC). Large and small EVs were separately analysed. We found that serum starvation of cells during the EV collecting period led to a dramatic decrease in EV secretion and major cell death. Regarding the isolation method, our findings suggest that UC and SEC gave similar EV recovery rates. Separation of large and small EV-enriched subpopulations was efficiently achieved with both purification protocols although certain difference in sample heterogeneity was observed. Noteworthy, while calnexin was abundant in large EVs, ALIX and CD63 were mainly found in small EVs. Finally, when the functionality of EVs was assessed on primary culture of adult murine cardiac fibroblasts, we found that EVs were taken up by these cells, which resulted in a pronounced reduction in the proliferative and migratory capacity of the cells. Specifically, a tendency towards a larger effect of SEC-related EVs was observed. No differences could be found between large and small EVs. Altogether, these results contribute to establish the basis for the use of EVs as therapeutic platforms, in particular in regenerative fields.


Assuntos
Vesículas Extracelulares , Miocárdio/citologia , Miofibroblastos/metabolismo , Células-Tronco/citologia , Animais , Proteínas de Ligação ao Cálcio/metabolismo , Calnexina/metabolismo , Células Cultivadas , Masculino , Camundongos , Ratos Wistar , Tetraspanina 30/metabolismo
10.
Int J Mol Sci ; 22(22)2021 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-34830141

RESUMO

Fibroblasts contribute to approximately 20% of the non-cardiomyocytic cells in the heart. They play important roles in the myocardial adaption to stretch, inflammation, and other pathophysiological conditions. Fibroblasts are a major source of extracellular matrix (ECM) proteins whose production is regulated by cytokines, such as TNF-α or TGF-ß. The resulting myocardial fibrosis is a hallmark of pathological remodeling in dilated cardiomyopathy (DCM). Therefore, in the present study, the secretome and corresponding transcriptome of human cardiac fibroblasts from patients with DCM was investigated under normal conditions and after TNF-α or TGF-ß stimulation. Secreted proteins were quantified via mass spectrometry and expression of genes coding for secreted proteins was analyzed via Affymetrix Transcriptome Profiling. Thus, we provide comprehensive proteome and transcriptome data on the human cardiac fibroblast's secretome. In the secretome of quiescent fibroblasts, 58% of the protein amount belonged to the ECM fraction. Interestingly, cytokines were responsible for 5% of the total protein amount in the secretome and up to 10% in the corresponding transcriptome. Furthermore, cytokine gene expression and secretion were upregulated upon TNF-α stimulation, while collagen secretion levels were elevated after TGF-ß treatment. These results suggest that myocardial fibroblasts contribute to pro-fibrotic and to inflammatory processes in response to extracellular stimuli.


Assuntos
Citocinas/farmacologia , Fibroblastos/efeitos dos fármacos , Miocárdio/metabolismo , Secretoma/efeitos dos fármacos , Transcriptoma/efeitos dos fármacos , Cardiomiopatia Dilatada/genética , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Citocinas/genética , Citocinas/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Humanos , Microscopia de Fluorescência , Miocárdio/citologia , Análise de Sequência com Séries de Oligonucleotídeos , Secretoma/metabolismo , Espectrometria de Massas em Tandem , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Fator de Crescimento Transformador beta/farmacologia , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
11.
Int J Mol Sci ; 22(22)2021 Nov 22.
Artigo em Inglês | MEDLINE | ID: mdl-34830444

RESUMO

A dome-shaped elastic poly(l-lactide-co-caprolactone) (PLCL) scaffold with a channel and pore structure was fabricated by a combinative method of 3D printing technology and the gel pressing method (13 mm in diameter and 6.5 mm in thickness) for patient-specific regeneration. The PLCL scaffold was combined with adipose decellularized extracellular matrix (adECM) and heart decellularized extracellular matrix (hdECM) hydrogels and human adipose-derived stem cells (hADSCs) to promote adipogenesis and angiogenesis. These scaffolds had mechanical properties similar to those of native adipose tissue for improved tissue regeneration. The results of the in vitro real-time PCR showed that the dECM hydrogel mixture induces adipogenesis. In addition, the in vivo study at 12 weeks demonstrated that the tissue-engineered PLCL scaffolds containing the hydrogel mixture (hdECM/adECM (80:20)) and hADSCs promoted angiogenesis and adipose tissue formation, and suppressed apoptosis. Therefore, we expect that our constructs will be clinically applicable as material for the regeneration of patient-specific large-sized adipose tissue.


Assuntos
Adipogenia/efeitos dos fármacos , Tecido Adiposo/crescimento & desenvolvimento , Neovascularização Fisiológica/efeitos dos fármacos , Regeneração/genética , Tecido Adiposo/transplante , Animais , Apoptose/efeitos dos fármacos , Matriz Extracelular Descelularizada/farmacologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Hidrogéis/química , Hidrogéis/farmacologia , Células-Tronco Mesenquimais/citologia , Camundongos , Miocárdio/citologia , Miocárdio/metabolismo , Neovascularização Fisiológica/genética , Poliésteres/farmacologia , Impressão Tridimensional , Regeneração/efeitos dos fármacos
12.
Bioengineered ; 12(1): 7872-7881, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34612779

RESUMO

Restoration of blood supply through medical or surgical intervention is a commonly adopted method for acute myocardial ischemia, but is also a trigger for cardiac ischemia/reperfusion injury. Studies have shown that remifentanil (REM) displays cardioprotective effects. In this study, the effects of REM on HCMEC viability were examined before and after the induction of H/R using Cell Counting Kit-8 assays. Wound healing and Matrigel angiogenesis assays were performed to assess HCMEC migration and angiogenesis, respectively. Commercial kits and western blotting were used to determine the endothelial barrier function of H/R-stimulated HCMECs with or without REM treatment. The expression of PI3K/Akt/hypoxia-inducible factor-1α (HIF-1α) pathway-related proteins was detected by western blotting. After pre-treatment with PI3K/Akt, the effects of REM on H/R-induced HCMEC injury were examined. We found that pre-treatment with REM displayed no impact on HCMEC viability under normal conditions but noticeably improved cell viability following H/R. The migratory abilities and tube-like structure formations of H/R-stimulated HCMECs were both enhanced by REM in a concentration-dependent manner. REM also decreased the permeability of H/R-stimulated HCMECs and upregulated the expression of tight junction proteins. Furthermore REM increased the expression of PI3K/Akt/HIF-1α signaling-related proteins in HCMECs. Inhibition of PI3K/Akt rescued REM-enhanced HCMEC function under H/R condition. Therefore, the present study demonstrated that REM pretreatment ameliorated H/R-induced HCMEC dysfunction by regulating the PI3K/Akt/HIF-1α signaling pathway.


Assuntos
Cardiotônicos/farmacologia , Hipóxia Celular/efeitos dos fármacos , Células Endoteliais , Miocárdio/citologia , Remifentanil/farmacologia , Células Cultivadas , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos
13.
Int J Mol Sci ; 22(20)2021 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-34681615

RESUMO

BACKGROUND: Carfilzomib is a first-line proteasome inhibitor indicated for relapsed/refractory multiple myeloma (MM), with its clinical use being hampered by cardiotoxic phenomena. We have previously established a translational model of carfilzomib cardiotoxicity in young adult mice, in which metformin emerged as a prophylactic therapy. Considering that MM is an elderly disease and that age is an independent risk factor for cardiotoxicity, herein, we sought to validate carfilzomib's cardiotoxicity in an in vivo model of aging. METHODS: Aged mice underwent the translational two- and four-dose protocols without and with metformin. Mice underwent echocardiography and were subsequently sacrificed for molecular analyses in the blood and cardiac tissue. RESULTS: Carfilzomib decreased proteasomal activity both in PBMCs and myocardium in both protocols. Carfilzomib induced mild cardiotoxicity after two doses and more pronounced cardiomyopathy in the four-dose protocol, while metformin maintained cardiac function. Carfilzomib led to an increased Bip expression and decreased AMPKα phosphorylation, while metformin coadministration partially decreased Bip expression and induced AMPKα phosphorylation, leading to enhanced myocardial LC3B-dependent autophagy. CONCLUSION: Carfilzomib induced cardiotoxicity in aged mice, an effect significantly reversed by metformin. The latter possesses translational importance as it further supports the clinical use of metformin as a potent prophylactic therapy.


Assuntos
Envelhecimento , Coração/efeitos dos fármacos , Metformina/farmacologia , Oligopeptídeos/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Autofagia/efeitos dos fármacos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Associadas aos Microtúbulos/metabolismo , Miocárdio/citologia , Miocárdio/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteína Fosfatase 2/metabolismo , Regulação para Cima/efeitos dos fármacos
14.
Eur J Pharmacol ; 912: 174566, 2021 Dec 05.
Artigo em Inglês | MEDLINE | ID: mdl-34653380

RESUMO

Since chemerin is an adipocytokine whose concentration in blood increases in the subjects with various cardiac diseases, chemerin may be involved in pathogenesis of cardiac diseases. In the present study, we examined the effects of chemerin-9, an active fragment of chemerin, on functions of cardiac fibroblasts, which are involved in pathophysiology of cardiac diseases. Primary cardiac fibroblasts were enzymatically isolated from adult male Wistar rats. Migration of cardiac fibroblasts was measured by a Boyden chamber assay and a scratch assay. Phosphorylation of Akt and extracellular signal-regulated kinase (ERK) was measured by Western blotting. Reactive oxygen species (ROS) production was measured by 2',7'-dichlorodihydrofluoresein staining. Chemerin-9 significantly stimulated migration in cardiac fibroblasts. Chemerin-9 significantly stimulated phosphorylation of Akt and ERK as well as ROS production. An Akt pathway inhibitor, LY294002, an ERK pathway inhibitor, PD98059, an antagonist of chemokine-like receptor 1 (CMKLR1), 2-(α-Napththoyl) ethyltrimethylammonium iodide, or an antioxidant, N-acetyl-L-cysteine prevented the migration induced by chemerin-9. In summary, we for the first time revealed that chemerin-9 stimulates migration perhaps through the ROS-dependent activation of Akt and ERK via CMKLR1 in cardiac fibroblasts. It is proposed that chemerin plays a role in the pathogenesis of cardiac diseases.


Assuntos
Movimento Celular/efeitos dos fármacos , Quimiocinas/metabolismo , Quimiocinas/farmacologia , Fibroblastos/efeitos dos fármacos , Acetilcisteína/farmacologia , Animais , Antioxidantes/farmacologia , Cromonas/farmacologia , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Flavonoides/farmacologia , Masculino , Morfolinas/farmacologia , Miocárdio/citologia , Naftalenos/farmacologia , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Compostos de Amônio Quaternário/farmacologia , Ratos Wistar , Espécies Reativas de Oxigênio/metabolismo , Receptores de Quimiocinas/antagonistas & inibidores , Receptores de Quimiocinas/metabolismo
15.
Cells ; 10(10)2021 09 25.
Artigo em Inglês | MEDLINE | ID: mdl-34685518

RESUMO

Myocardium Infarction (MI) is one of the foremost cardiovascular diseases (CVDs) causing death worldwide, and its case numbers are expected to continuously increase in the coming years. Pharmacological interventions have not been at the forefront in ameliorating MI-related morbidity and mortality. Stem cell-based tissue engineering approaches have been extensively explored for their regenerative potential in the infarcted myocardium. Recent studies on microfluidic devices employing stem cells under laboratory set-up have revealed meticulous events pertaining to the pathophysiology of MI occurring at the infarcted site. This discovery also underpins the appropriate conditions in the niche for differentiating stem cells into mature cardiomyocyte-like cells and leads to engineering of the scaffold via mimicking of native cardiac physiological conditions. However, the mode of stem cell-loaded engineered scaffolds delivered to the site of infarction is still a challenging mission, and yet to be translated to the clinical setting. In this review, we have elucidated the various strategies developed using a hydrogel-based system both as encapsulated stem cells and as biocompatible patches loaded with cells and applied at the site of infarction.


Assuntos
Infarto do Miocárdio/patologia , Miocárdio/citologia , Miócitos Cardíacos/citologia , Regeneração/fisiologia , Células-Tronco/citologia , Diferenciação Celular/fisiologia , Humanos , Infarto do Miocárdio/fisiopatologia , Transplante de Células-Tronco/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais
16.
Biochem Biophys Res Commun ; 577: 6-11, 2021 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-34487961

RESUMO

Premature ovarian failure (POF) is defined by amenorrhea, hypoestrogenism, elevated gonadotropin levels, and infertility. Chemotherapeutic agents are the most gonadotoxic agents that lead to POF. Although some previous studies have presented that mesenchymal stem cells (MSCs) transplantation could rescue the ovary function of POF animal models through the paracrine pathway, these mechanisms require further investigation. However, mechanisms of embryonic stem cell-derived MSCs (ES-MSCs) therapeutic effects on POF animal models have not been fully investigated yet. This study aimed to evaluate the migration and distribution of ES-MSCs in a model of chemotherapy-induced POF. Female mice received intraperitoneal injections of cyclophosphamide (Cy) to induce POF. Then, MSCs were labeled with green fluorescent protein (GFP) in vitro and injected intravenously into POF mice, and the distribution of MSCs was dynamically monitored at 1 week after transplantation. We harvested the lungs, liver, spleen, ovaries, heart, and kidneys 1 week after transplantation. The sections of these tissues were observed under the fluorescent microscope. More than 70% MSCs were successfully labeled with GFP at 72 h after labeling. MSCs were uniformly distributed in multiple organs and tissues including lungs, liver, spleen, ovaries, heart, and kidneys of POF mice. In mice, at 1week after intravenous transplantation, GFP labeled ES-MSCs were observed in the lungs, liver, spleen, ovaries, heart, and kidneys of POF mice, and the number of GFP labeled ES-MSCs in lungs, ovaries, and heart were higher than that in the spleen, kidneys, and liver. Our results revealed intravenously implanted ES-MSCs could migrate into the various tissues in chemotherapy-induced damaged POF mice.


Assuntos
Modelos Animais de Doenças , Células-Tronco Embrionárias Humanas/metabolismo , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/metabolismo , Insuficiência Ovariana Primária/terapia , Animais , Diferenciação Celular , Movimento Celular , Rastreamento de Células/métodos , Células Cultivadas , Ciclofosfamida , Feminino , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células-Tronco Embrionárias Humanas/citologia , Humanos , Pulmão/citologia , Pulmão/metabolismo , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Microscopia de Fluorescência/métodos , Miocárdio/citologia , Miocárdio/metabolismo , Ovário/citologia , Ovário/metabolismo , Insuficiência Ovariana Primária/induzido quimicamente , Insuficiência Ovariana Primária/metabolismo
17.
Bioengineered ; 12(1): 6869-6877, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34521301

RESUMO

The differentiation of cardiac fibroblast to myofibroblast is the key process of cardiac fibrosis. In the study, we aimed to determine the function of E2F Transcription Factor 1 (E2F1) in human cardiac fibroblasts (HCFs) differentiation, search for its downstream genes and elucidate the function of them in HCFs differentiation. As a result, we found that E2F1 was up-regulated in TGF-ß1-induced HCFs differentiation. Silencing the expression of E2F1 by siRNA in HCFs, we found that the expression of differentiation-related genes (Collagen-1, α-Smooth muscle actin, and Fibronectin-1) was significantly suppressed, combining with proliferation and migration assay, we determined that HCFs differentiation was decreased. Luciferase report assay and immunoprecipitation proved that the oncogene CCNE2 was a direct target gene of E2F1, overexpression of CCNE2 was found in differentiated HCFs, silencing the expression of CCNE2 by siRNA decreased HCFs differentiation. Our research suggested that E2F1 and its downstream target gene CCNE2 play a vital role in TGF-ß1-induced HCFs differentiation, thus E2F1 and CCNE2 may be a potential therapeutic target for cardiac fibrosis.


Assuntos
Diferenciação Celular/genética , Ciclinas/genética , Fator de Transcrição E2F1/genética , Miocárdio/citologia , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Ciclinas/metabolismo , Fator de Transcrição E2F1/metabolismo , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Transcrição Gênica/genética , Fator de Crescimento Transformador beta1/farmacologia
18.
Int J Mol Sci ; 22(18)2021 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-34576032

RESUMO

The coronavirus disease 2019 (COVID-19) pandemic with high infectivity and mortality has caused severe social and economic impacts worldwide. Growing reports of COVID-19 patients with multi-organ damage indicated that severe acute respiratory syndrome coronavirus 2 (SARS-CoV2) may also disturb the cardiovascular system. Herein, we used human induced pluripotent stem cell (iPSC)-derived cardiomyocytes (iCMs) as the in vitro platform to examine the consequence of SARS-CoV2 infection on iCMs. Differentiated iCMs expressed the primary SARS-CoV2 receptor angiotensin-converting enzyme-II (ACE2) and the transmembrane protease serine type 2 (TMPRSS2) receptor suggesting the susceptibility of iCMs to SARS-CoV2. Following the infection of iCMs with SARS-CoV2, the viral nucleocapsid (N) protein was detected in the host cells, demonstrating the successful infection. Bioinformatics analysis revealed that the SARS-CoV2 infection upregulates several inflammation-related genes, including the proinflammatory cytokine tumor necrosis factor-α (TNF-α). The pretreatment of iCMs with TNF-α for 24 h, significantly increased the expression of ACE2 and TMPRSS2, SASR-CoV2 entry receptors. The TNF-α pretreatment enhanced the entry of GFP-expressing SARS-CoV2 pseudovirus into iCMs, and the neutralization of TNF-α ameliorated the TNF-α-enhanced viral entry. Collectively, SARS-CoV2 elevated TNF-α expression, which in turn enhanced the SARS-CoV2 viral entry. Our findings suggest that, TNF-α may participate in the cytokine storm and aggravate the myocardial damage in COVID-19 patients.


Assuntos
COVID-19/complicações , Doenças Cardiovasculares/imunologia , Síndrome da Liberação de Citocina/imunologia , SARS-CoV-2/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Enzima de Conversão de Angiotensina 2/metabolismo , COVID-19/imunologia , COVID-19/patologia , COVID-19/virologia , Doenças Cardiovasculares/virologia , Diferenciação Celular , Linhagem Celular , Biologia Computacional , Proteínas do Nucleocapsídeo de Coronavírus/metabolismo , Síndrome da Liberação de Citocina/patologia , Síndrome da Liberação de Citocina/virologia , Humanos , Células-Tronco Pluripotentes Induzidas , Miocárdio/citologia , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/virologia , Fosfoproteínas/metabolismo , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidade , Serina Endopeptidases/metabolismo , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Regulação para Cima/imunologia , Internalização do Vírus/efeitos dos fármacos
19.
J Pharm Pharmacol ; 73(11): 1470-1479, 2021 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-34383044

RESUMO

INTRODUCTION: Sevoflurane (Sevo) prevents hypoxia/reoxygenation (H/R)-induced cardiomyocytes apoptosis. MiR-27a-3p expression is up-regulated in Sevo-treated hippocampal neurons. OBJECTIVE: This study explored whether the effect of Sevo on cardiomyocytes was mediated by miR-27a-3p. METHODS: The cardiomyocytes were cultured under H/R condition or pre-treated with Sevo, and further transfected with miR-27a-3p inhibitor or treated with an autophagy inhibitor 3-methyladenine (3-MA). Then the cell morphology was observed under an optical microscope. The cell viability and apoptosis were measured by MTT and flow cytometry. Expressions of miR-27a-3p, apoptosis-related, and autophagy-related factors were determined by western blot or RT-qPCR. KEY FINDINGS: Sevo improved the abnormal morphology, promoted the cell viability and the expressions of Bcl-2 and miR-27a-3p, but reduced the apoptosis and Bax and C-caspase-3 levels of H/R-induced cardiomyocytes. MiR-27a-3p inhibitor had an effect opposite to Sevo on the cardiomyocytes and further counteracted the effect of Sevo on the H/R-induced cardiomyocytes. Downregulation miR-27a-3p increased the expression of Beclin 1 and the ratio of LC3B-II to LC3B-I in H/R-induced cardiomyocytes. Furthermore, 3-MA had an opposite effect to miR-27a-3p inhibitor and further counteracted the effect of the miR-27a-3p inhibitor on H/R-induced cardiomyocytes. CONCLUSION: Sevo inhibited the apoptosis of H/R-induced cardiomyocytes via regulating miR-27a-3p-mediated autophagy.


Assuntos
MicroRNAs/metabolismo , Miocárdio/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Traumatismo por Reperfusão/metabolismo , Sevoflurano/farmacologia , Anestésicos Inalatórios/farmacologia , Animais , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Regulação para Baixo , Hipocampo , Hipóxia , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/citologia , Miocárdio/patologia , Miócitos Cardíacos/metabolismo , Neurônios , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Traumatismo por Reperfusão/patologia , Regulação para Cima
20.
Cells ; 10(7)2021 06 22.
Artigo em Inglês | MEDLINE | ID: mdl-34206684

RESUMO

Direct cardiac reprogramming of fibroblasts into induced cardiomyocytes (iCMs) is a promising approach but remains a challenge in heart regeneration. Efforts have focused on improving the efficiency by understanding fundamental mechanisms. One major challenge is that the plasticity of cultured fibroblast varies batch to batch with unknown mechanisms. Here, we noticed a portion of in vitro cultured fibroblasts have been activated to differentiate into myofibroblasts, marked by the expression of αSMA, even in primary cell cultures. Both forskolin, which increases cAMP levels, and TGFß inhibitor SB431542 can efficiently suppress myofibroblast differentiation of cultured fibroblasts. However, SB431542 improved but forskolin blocked iCM reprogramming of fibroblasts that were infected with retroviruses of Gata4, Mef2c, and Tbx5 (GMT). Moreover, inhibitors of cAMP downstream signaling pathways, PKA or CREB-CBP, significantly improved the efficiency of reprogramming. Consistently, inhibition of p38/MAPK, another upstream regulator of CREB-CBP, also improved reprogramming efficiency. We then investigated if inhibition of these signaling pathways in primary cultured fibroblasts could improve their plasticity for reprogramming and found that preconditioning of cultured fibroblasts with CREB-CBP inhibitor significantly improved the cellular plasticity of fibroblasts to be reprogrammed, yielding ~2-fold more iCMs than untreated control cells. In conclusion, suppression of CREB-CBP signaling improves fibroblast plasticity for direct cardiac reprogramming.


Assuntos
Plasticidade Celular , Reprogramação Celular , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Fibroblastos/citologia , Fibroblastos/metabolismo , Proteínas de Membrana/metabolismo , Miocárdio/citologia , Fosfoproteínas/metabolismo , Transdução de Sinais , Animais , Benzamidas/farmacologia , Diferenciação Celular/efeitos dos fármacos , Plasticidade Celular/efeitos dos fármacos , Células Cultivadas , Reprogramação Celular/efeitos dos fármacos , Colforsina/farmacologia , AMP Cíclico/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Dioxóis/farmacologia , Fibroblastos/efeitos dos fármacos , Camundongos Transgênicos , Miofibroblastos/citologia , Miofibroblastos/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA