Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 77
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Theranostics ; 14(6): 2427-2441, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38646649

RESUMO

Background: MER proto-oncogene tyrosine kinase (MerTK) is a key receptor for efferocytosis, a process for the clearance of apoptotic cells. MerTK is mainly expressed in macrophages and immature dendritic cells. There are very limited reports focused on MerTK biology in aortic endothelial cells (ECs). It remains unclear for the role of blood flow patterns in regulating MerTK-mediated efferocytosis in aortic ECs. This study was designed to investigate whether endothelial MerTK and EC efferocytosis respond to blood flow patterns during atherosclerosis. Methods: Big data analytics, RNA-seq and proteomics combined with our in vitro and in vivo studies were applied to reveal the potential molecular mechanisms. Partial carotid artery ligation combined with AAV-PCSK9 and high fat diet were used to set up acute atherosclerosis in 4 weeks. Results: Our data showed that MerTK is sensitive to blood flow patterns and is inhibited by disturbed flow and oscillatory shear stress in primary human aortic ECs (HAECs). The RNA-seq data in HAECs incubated with apoptotic cells showed that d-flow promotes pro-inflammatory pathway and senescence pathway. Our in vivo data of proteomics and immunostaining showed that, compared with WT group, MerTK-/- aggravates atherosclerosis in d-flow areas through upregulation of endothelial dysfunction markers (e.g. IL-1ß, NF-κB, TLR4, MAPK signaling, vWF, VCAM-1 and p22phox) and mitochondrial dysfunction. Interestingly, MerTK-/-induces obvious abnormal endothelial thickening accompanied with decreased endothelial efferocytosis, promoting the development of atherosclerosis. Conclusions: Our data suggests that blood flow patterns play an important role in regulating MerTK-mediated efferocytosis in aortic ECs, revealing a new promising therapeutic strategy with EC efferocytosis restoration to against atherosclerosis.


Assuntos
Aorta , Aterosclerose , Células Endoteliais , Fagocitose , c-Mer Tirosina Quinase , c-Mer Tirosina Quinase/metabolismo , c-Mer Tirosina Quinase/genética , Aterosclerose/metabolismo , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Humanos , Células Endoteliais/metabolismo , Animais , Aorta/metabolismo , Aorta/patologia , Camundongos , Apoptose , Proto-Oncogene Mas , Masculino , Camundongos Endogâmicos C57BL , Dieta Hiperlipídica , Células Cultivadas , Eferocitose
2.
Redox Biol ; 70: 103061, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38341954

RESUMO

RATIONALE: MER proto-oncogene tyrosine kinase (MerTK) is a key receptor for the clearance of apoptotic cells (efferocytosis) and plays important roles in redox-related human diseases. We will explore MerTK biology in human cells, tissues, and diseases based on big data analytics. METHODS: The human RNA-seq and scRNA-seq data about 42,700 samples were from NCBI Gene Expression Omnibus and analyzed by QIAGEN Ingenuity Pathway Analysis (IPA) with about 170,000 crossover analysis. MerTK expression was quantified as Log2 (FPKM + 0.1). RESULTS: We found that, in human cells, MerTK is highly expressed in macrophages, monocytes, progenitor cells, alpha-beta T cells, plasma B cells, myeloid cells, and endothelial cells (ECs). In human tissues, MerTK has higher expression in plaque, blood vessels, heart, liver, sensory system, artificial tissue, bone, adrenal gland, central nervous system (CNS), and connective tissue. Compared to normal conditions, MerTK expression in related tissues is altered in many human diseases, including cardiovascular diseases, cancer, and brain disorders. Interestingly, MerTK expression also shows sex differences in many tissues, indicating that MerTK may have different impact on male and female. Finally, based on our proteomics from primary human aortic ECs, we validated the functions of MerTK in several human diseases, such as cancer, aging, kidney failure and heart failure. CONCLUSIONS: Our big data analytics suggest that MerTK may be a promising therapeutic target, but how it should be modulated depends on the disease types and sex differences. For example, MerTK inhibition emerges as a new strategy for cancer therapy due to it counteracts effect on anti-tumor immunity, while MerTK restoration represents a promising treatment for atherosclerosis and myocardial infarction as MerTK is cleaved in these disease conditions.


Assuntos
Receptores Proteína Tirosina Quinases , c-Mer Tirosina Quinase , Feminino , Humanos , Masculino , Apoptose/genética , c-Mer Tirosina Quinase/genética , Ciência de Dados , Células Endoteliais/metabolismo , Genômica , Neoplasias/metabolismo , Fagocitose , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo , Encefalopatias/metabolismo
3.
Theranostics ; 13(9): 2914-2929, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37284459

RESUMO

Aims: Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a serine protease that binds to low-density lipoprotein receptors. Efferocytosis is the process by which phagocytes remove apoptotic cells. Both PCSK9 and efferocytosis play important roles in regulating redox biology and inflammation, the key factors contributing to vascular aging. This study was designed to investigate the impact of PCSK9 on efferocytosis in endothelial cells (ECs) and its implications in vascular aging. Methods and Results: Studies were performed in primary human aortic ECs (HAECs) and primary mouse aortic ECs (MAECs) isolated from male wild-type (WT) and PCSK9-/- mice, and in young and aged mice treated with saline or the PCSK9 inhibitor Pep2-8. Our findings include that recombinant PCSK9 protein induces defective efferocytosis and aging marker senescence-associated-ß-galactosidase (SA-ß-gal) expression in ECs, while PCSK9-/- restores efferocytosis and inhibits SA-ß-gal activity. Further studies in aged mice showed that endothelial deficiency of MerTK, a critical receptor for efferocytosis that allows phagocytes to detect the presence of apoptotic cells, may be an indicator of vascular dysfunction in the aortic arch. Pep2-8 treatment markedly restored efferocytosis in endothelium from the aged mice. A proteomics study in the aortic arch from aged mice revealed that Pep2-8 administration significantly downregulates expression of NOX4, MAPK subunits, NF-κB, and secretion of pro-inflammatory cytokines, all known to promote vascular aging. Immunofluorescent staining showed that Pep2-8 administration upregulates expression of eNOS and downregulates expression of pro-IL-1ß, NF-κB and p22phox compared to saline treated group. Conclusions: These findings provide initial evidence for the ability of aortic ECs to accomplish efferocytosis and argue for a role of PCSK9 in attenuating EC efferocytosis, thereby leading to vascular dysfunction and acceleration in vascular aging.


Assuntos
Células Endoteliais , Pró-Proteína Convertase 9 , Masculino , Camundongos , Humanos , Animais , Pró-Proteína Convertase 9/genética , Células Endoteliais/metabolismo , NF-kappa B/metabolismo , Envelhecimento
6.
Theranostics ; 12(1): 418, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34987654

RESUMO

[This corrects the article DOI: 10.7150/thno.45939.].

11.
Front Pharmacol ; 12: 699949, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34512335

RESUMO

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) or COVID-19 infection is the cause of the ongoing global pandemic. Mortality from COVID-19 infection is particularly high in patients with cardiovascular diseases. In addition, COVID-19 patients with preexisting cardiovascular comorbidities have a higher risk of death. Main cardiovascular complications of COVID-19 are myocardial infarction, myocarditis, acute myocardial injury, arrhythmias, heart failure, stroke, and venous thromboembolism. Therapeutic interventions in terms of drugs for COVID-19 have many cardiac adverse effects. Here, we review the relative therapeutic efficacy and adverse effects of anti-COVID-19 drugs.

12.
Am J Physiol Regul Integr Comp Physiol ; 321(5): R639-R654, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34431382

RESUMO

After an ischemic event, there is activation of fibroblasts leading to scar formation. It is critical to limit the profibrotic remodeling and activate the reparative remodeling phase to limit cardiac diastolic dysfunction. Mesenchymal stem cell (MSC) exosomes offer significant protection against ischemia-related systolic dysfunction. Here, we studied if MSC exosomes would offer protection against profibrotic events in mouse hearts subjected to acute ischemia [1 h left coronary artery (LCA) occlusion] or chronic ischemia (7 days LCA occlusion). After acute ischemia, there was activation of inflammatory signals, more in the peri-infarct than in the infarct area, in the saline (vehicle)-treated mice. At the same time, there was expression of cardiac remodeling signals (vimentin, collagens-1 and -3, and fibronectin), more in the infarct area. Treatment with MSC exosomes before LCA ligation suppressed inflammatory signals during acute and chronic ischemia. Furthermore, exosome treatment promoted pro-reparative cardiac extracellular matrix (ECM) remodeling in both infarct and peri-infarct areas by suppressing fibronectin secretion and by modulating collagen secretion to reduce fibrotic scar formation through altered cellular signaling pathways. Proteomics study revealed intense expression of IL-1ß and activation of profibrotic signals in the saline-treated hearts and their suppression in MSC exosome-treated hearts. To our knowledge, this is the first report on the infarct and peri-infarct area proteomics of ischemic mice hearts to explain MSC exosome-mediated suppression of scar formation in the ischemic mouse hearts.


Assuntos
Exossomos/transplante , Fibroblastos/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Isquemia Miocárdica/cirurgia , Miocárdio/metabolismo , Proteoma , Proteômica , Remodelação Ventricular , Animais , Western Blotting , Linhagem Celular , Movimento Celular , Modelos Animais de Doenças , Eletroforese em Gel de Poliacrilamida , Exossomos/metabolismo , Proteínas da Matriz Extracelular/metabolismo , Fibroblastos/patologia , Fibrose , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Masculino , Espectrometria de Massas , Células-Tronco Mesenquimais/metabolismo , Camundongos Endogâmicos C57BL , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patologia , Miocárdio/patologia
13.
Mol Cell Biochem ; 476(4): 1691-1704, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33423165

RESUMO

Mesenchymal stem cell (MSC) exosomes may limit cardiac injury, and even reverse cardiac damage in animal models of ischemia. To understand exosome-mediated improvement in cardiac function we examined the proteomic alternations in the MSC exosome-treated mice hearts subjected to left coronary artery (LCA) ligation, with particular emphasis on peri-infarct areas. At 7 days after LCA ligation, left ventricular end systolic thickness, infarct size and survival of mice were studied. Mass spectrometric analysis of infarct and peri-infarct areas was carried out. Expression of inflammatory markers (LOX-1 and NLRP3) and cell death markers (Bax, Bcl-2, Caspases 1 and 3 and GSDMD) were investigated by Western blots and immunofluorescence. Proteomic analysis of the infarct and peri-infarct areas in saline-treated hearts revealed differentially expressed proteins involved in inflammation and apoptotic cell death, while showing depletion of processes governing cell death. Exosome treatment significantly improved the proteomic profile in both infarct and peri-infarct areas, more so in the peri-infarct areas. The infarct size was smaller (9 ± 1%), and cardiac contractile function (fractional shortening) was preserved in the exosome-treated mice (28 ± 2%). Survival of exosome-treated mice was also better. White blood cell accumulation in and around the infarct area, expression of LOX-1 and NLRP3 inflammasome, and markers of cell death (cleaved Caspase-3, Caspase-1, GSDMD, Bcl-2 and Bax) were dramatically reduced by MSC exosome treatment (all p < 0.01). In cultured primary mouse cardiomyocytes, treatment with MSC exosomes essentially reversed inflammation-induced pro-apoptotic and inflammatory signals (p < 0.01). MSC exosomes exert their cardioprotective effects by suppressing inflammation and pro-apoptotic processes, particularly in the peri-infarct areas, resulting in preservation of cardiac function after LCA ligation.


Assuntos
Exossomos , Células-Tronco Mesenquimais/metabolismo , Infarto do Miocárdio , Animais , Linhagem Celular Transformada , Exossomos/metabolismo , Exossomos/patologia , Exossomos/transplante , Humanos , Masculino , Células-Tronco Mesenquimais/patologia , Camundongos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/prevenção & controle
15.
Basic Res Cardiol ; 115(6): 66, 2020 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-33180196

RESUMO

Proprotein convertase subtilisin/Kexin type 9 (PCSK9) and pyroptosis both play important roles in myocardial infarction. This study was designed to test the hypothesis that PCSK9 regulates pyroptosis in cardiomyocytes during chronic myocardial ischemia. Primary cardiomyocytes were isolated from WT and PCSK9-/- mice. HL-1 cardiomyocytes were used to set up PCSK9-deficient (PCSK9-/-) and PCSK9-upregulated (PCSK9CRISPRa) cardiomyocyte cell line with CRISPR/Cas9 knockout or activation plasmid. Additional studies were performed with chronic myocardial ischemia in WT and PCSK9-/- mice. We observed that PCSK9 initiates mitochondrial DNA (mtDNA) damage, activates NLRP3 inflammasome signaling (NLRP3, ASC, Caspase-1, IL-1ß, and IL-18), and subsequently induces Caspase-1-dependent pyroptosis. There was an intense expression of PCSK9 and pyroptosis marker, GSDMD-NT, in the zone bordering the infarct area. PCSK9-/- significantly suppressed expression of NLRP3 inflammasome signaling, GSDMD-NT, and LDH release. Furthermore, serum levels of PCSK9, NLPR3 inflammasome signaling, and pyroptosis (GSDMD and LDH release) were significantly elevated in patients with chronic myocardial ischemia as compared to those in age-matched healthy subjects. Human hearts with recent infarcts also showed high expression of PCSK9 and GSDMD-NT in the border zone similar to that in the infarcted mouse heart. These observations provide compelling evidence for the role of PCSK9 in regulating Caspase-1-dependent pyroptosis via mtDNA damage and may qualify pro-inflammatory cytokines and pyroptosis as potential targets to treat PCSK9-related cardiovascular diseases.


Assuntos
Dano ao DNA , DNA Mitocondrial/metabolismo , Mitocôndrias Cardíacas/enzimologia , Isquemia Miocárdica/enzimologia , Miócitos Cardíacos/enzimologia , Pró-Proteína Convertase 9/metabolismo , Piroptose , Idoso , Animais , Estudos de Casos e Controles , Caspase 1/metabolismo , Linhagem Celular , Doença Crônica , DNA Mitocondrial/genética , Modelos Animais de Doenças , Feminino , Humanos , Inflamassomos/metabolismo , Mediadores da Inflamação/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/patologia , Isquemia Miocárdica/genética , Isquemia Miocárdica/patologia , Miócitos Cardíacos/patologia , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Proteínas de Ligação a Fosfato/metabolismo , Pró-Proteína Convertase 9/genética , Transdução de Sinais
16.
Theranostics ; 10(16): 7100-7110, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32641981

RESUMO

Background: Both PCSK9 and NLRP3 inflammasome play important roles in atherogenesis. This study was designed to test the hypothesis that NLRP3 inflammasome via IL-1ß induces PCSK9 secretion. The inter-twined relationship between NLRP3 inflammasome, IL-1ß and PCSK9 may be relevant in atherogenesis. Methods: We studied NLRP3 inflammasome-mediated PCSK9 secretion in mouse peritoneal macrophages and in a variety of tissues, such as liver, kidney and small intestine. Macrophages were derived from wild-type (WT) and a variety of gene deletion mice to define the mechanistic basis of NLRP3 inflammasome -mediated PCSK9 secretion. Additional studies were performed in high-fat diet fed mice. Results: We observed that NLRP3 and its downstream signals ASC, Caspase-1, IL-18, and IL-1ß all participate in PCSK9 secretion. IL-1ß seems to be more important than IL-18 in the induction of PCSK9 secretion. Further, there appears to be significant involvement of MAPKs in this process. Lastly, we observed that mice fed high fat diet have high expression of NLRP3 and a greater secretion of PCSK9 than mice fed a standard diet, and this increased secretion of PCSK9 in high fat diet-fed mice was attenuated in IL-1ß-/- mice. Conclusions: This study based on extensive in vitro and in vivo data provides evidence that NLRP3 inflammasome via IL-1ß plays an important role in determining PCSK9 secretion, particularly in the presence of high-fat diet.


Assuntos
Aterosclerose/imunologia , Inflamassomos/imunologia , Interleucina-1beta/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Pró-Proteína Convertase 9/metabolismo , Animais , Aterosclerose/sangue , Células Cultivadas , Dieta Hiperlipídica/efeitos adversos , Modelos Animais de Doenças , Humanos , Inflamassomos/metabolismo , Interleucina-1beta/genética , Sistema de Sinalização das MAP Quinases/imunologia , Macrófagos Peritoneais/imunologia , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Knockout , Cultura Primária de Células , Espécies Reativas de Oxigênio/metabolismo
17.
Cardiovasc Res ; 116(10): 1721-1732, 2020 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-31593224

RESUMO

AIMS: Blood flow patterns play an important role in the localization of atherosclerosis in the sense that low-flow state is pro-atherogenic, and helical flow is protective against atherosclerosis. Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates cholesterol metabolism via low-density lipoprotein receptor (LDLr) degradation and is highly expressed in the atherosclerotic tissues. This study was designed to investigate the role of different blood flow patterns in the regulation of PCSK9 expression. METHODS AND RESULTS: We designed an experimental model guider to generate stable helical flow. Our data showed that compared with normal flow, low-flow state induces whereas helical flow inhibits PCSK9 expression in the rabbit thoracic aorta in an inflammatory state. Our data also identified that TLR4-MyD88-NF-κB signalling plays an important role in PCSK9 expression. On the other hand, TRIF pathway had almost no effect. Further studies showed that the signals downstream of NF-κB, such as pro-inflammatory cytokines (IL-1ß, IL-18, MCP-1, IL-6, TNF-α, IL-12, IFNγ, and GM-CSF) directly influence PCSK9 expression. Interestingly, high fat diet further enhanced PCSK9 expression in an inflammatory milieu. CONCLUSIONS: These observations suggest a link between abnormal flow patterns and PCSK9 expression in inflammatory states, which may qualify helical flow and pro-inflammatory cytokines as potential targets to treat PCSK9-related cardiovascular diseases.


Assuntos
Aorta Torácica/enzimologia , Aortite/enzimologia , Aterosclerose/enzimologia , Citocinas/metabolismo , Mediadores da Inflamação/metabolismo , Fator 88 de Diferenciação Mieloide/metabolismo , Pró-Proteína Convertase 9/metabolismo , Animais , Aorta Torácica/fisiopatologia , Aortite/fisiopatologia , Aterosclerose/fisiopatologia , Simulação por Computador , Modelos Animais de Doenças , Masculino , Mecanotransdução Celular , Modelos Cardiovasculares , NF-kappa B/metabolismo , Coelhos , Fluxo Sanguíneo Regional , Receptor 4 Toll-Like/metabolismo
18.
Cardiovasc Res ; 116(5): 908-915, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-31746997

RESUMO

PCSK9 degrades low-density lipoprotein cholesterol (LDL) receptors and subsequently increases serum LDL cholesterol. Clinical trials show that inhibition of PCSK9 efficiently lowers LDL cholesterol levels and reduces cardiovascular events. PCSK9 inhibitors also reduce the extent of atherosclerosis. Recent studies show that PCSK9 is secreted by vascular endothelial cells, smooth muscle cells, and macrophages. PCSK9 induces secretion of pro-inflammatory cytokines in macrophages, liver cells, and in a variety of tissues. PCSK9 regulates toll-like receptor 4 expression and NF-κB activation as well as development of apoptosis and autophagy. PCSK9 also interacts with oxidized-LDL receptor-1 (LOX-1) in a mutually facilitative fashion. These observations suggest that PCSK9 is inter-twined with inflammation with implications in atherosclerosis and its major consequence-myocardial ischaemia. This relationship provides a basis for the use of PCSK9 inhibitors in prevention of atherosclerosis and related clinical events.


Assuntos
Artérias/enzimologia , Aterosclerose/enzimologia , Citocinas/metabolismo , Dislipidemias/enzimologia , Mediadores da Inflamação/metabolismo , Inflamação/enzimologia , Pró-Proteína Convertase 9/metabolismo , Receptores Depuradores Classe E/metabolismo , Animais , Anti-Infecciosos/uso terapêutico , Artérias/efeitos dos fármacos , Artérias/patologia , Artérias/fisiopatologia , Aterosclerose/tratamento farmacológico , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Dislipidemias/tratamento farmacológico , Dislipidemias/patologia , Dislipidemias/fisiopatologia , Humanos , Hipolipemiantes/uso terapêutico , Inflamação/tratamento farmacológico , Inflamação/patologia , Inflamação/fisiopatologia , Mediadores da Inflamação/antagonistas & inibidores , Mecanotransdução Celular , Inibidores de PCSK9 , Placa Aterosclerótica , Fluxo Sanguíneo Regional , Inibidores de Serina Proteinase/uso terapêutico , Estresse Mecânico
19.
Sci Rep ; 9(1): 19276, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31848380

RESUMO

A host of hormonal-metabolic alterations take place following exposure of cardiomyocytes to hypoxia and other noxious stimuli. Here, we demonstrate that exposure of cultured rat cardiomyocytes to lipopolysaccharide (LPS) resulted in upregulation (~1.5 fold) of oxidized low-density lipoprotein receptor-1 (LOX-1). There was also a marked increase in apoptosis 12 hrs after LPS treatment with caspase-3 levels being significantly elevated (~1.3 fold) and a significant increase in LDH release at 24 hrs. Interestingly, there was a ~1.4-fold upregulation of LC-3 expression post-LPS treatment indicating development of autophagy, which probably is a compensatory response to combat cellular injury induced by LPS. Treatment with LPS also reduced the size and morphology of cardiomyocyte spheroids. In an attempt to limit LPS-induced injury, cardiomyocytes were treated with exosomes derived from mesenchymal stromal cells (MSCs). We noted a significant suppression of LOX-1 expression that in turn suppressed apoptosis as well as autophagic response and restored spheroid morphology. Mass spectrophotometric analysis of MSC exosomes revealed a cargo rich in proteins which are involved in pathways negatively modulating cell death and apoptosis while promoting cell survival. This is first report to our knowledge on the initial molecular events in MSC exosome mediated cytoprotection of stressed cardiomyocytes.


Assuntos
Exossomos/genética , Lipopolissacarídeos/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Receptores Depuradores Classe E/genética , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Autofagia/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Citoproteção/efeitos dos fármacos , Exossomos/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Espectrometria de Massas , Células-Tronco Mesenquimais/efeitos dos fármacos , Ratos , Esferoides Celulares/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...