Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oxid Med Cell Longev ; 2019: 8912768, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30774748

RESUMO

Cardiovascular diseases frequently coexist with chronic kidney disease that constitutes a major determinant of outcome in patients with heart failure. Dysfunction of both organs is related to chronic inflammation, endothelial dysfunction, oxidative stress, and fibrosis. Widespread expression of serine protease DPP4 that degrades varieties of substrates suggests its involvement in numerous physiological processes. In this study, we tested the effects of selective DPP4 inhibition on the progression of renal disease in a nondiabetic model of hypertensive heart disease using Dahl salt-sensitive rats. Chronic DPP4 inhibition positively affected renal function with a significant reduction in albuminuria and serum creatinine. DPP4 inhibition attenuated the inflammatory component by reducing the expression of NF-κB, TNFα, IL-1ß, IL-6, and MCP-1. Kidney macrophages expressed GLP-1R, and DPP4 inhibition promoted macrophage polarization toward the anti-inflammatory M2 phenotype. Finally, high degrees of NADPH oxidase 4 expression and oxidation of nucleic acids, lipids, and proteins were reduced upon DPP4 inhibition. Our study provides evidence of renoprotection by DPP4 inhibition in a nondiabetic hypertension-induced model of chronic cardiorenal syndrome, indicating that DPP4 pathway remains a valid object to study in the context of chronic multiorgan diseases.


Assuntos
Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Hipertensão/complicações , Hipertensão/tratamento farmacológico , Insuficiência Renal Crônica/complicações , Insuficiência Renal Crônica/tratamento farmacológico , Cloreto de Sódio na Dieta/efeitos adversos , Animais , Inibidores da Dipeptidil Peptidase IV/farmacologia , Modelos Animais de Doenças , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Fibrose , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Hipertensão/fisiopatologia , Inflamação/patologia , Rim/efeitos dos fármacos , Rim/fisiopatologia , Testes de Função Renal , Modelos Biológicos , Estresse Nitrosativo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Fenótipo , Ratos Endogâmicos Dahl , Insuficiência Renal Crônica/fisiopatologia , Transdução de Sinais/efeitos dos fármacos
2.
Stem Cells Int ; 2018: 9492038, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29731780

RESUMO

Pulmonary emphysema is a respiratory condition characterized by alveolar destruction that leads to airflow limitation and reduced lung function. Although with extensive research, the pathophysiology of emphysema is poorly understood and effective treatments are still missing. Evidence suggests that mesenchymal stem cells (MSCs) possess the ability to engraft the injured tissues and induce repair via a paracrine effect. Thus, the aim of this study was to test the effects of the intratracheal administration of lung-derived mouse MSCs in a model of elastase-induced emphysema. Pulmonary function (static lung compliance) showed an increased stiffness induced by elastase, while morphometric findings (mean linear intercept and tissue/alveolar area) confirmed the severity of alveolar disruption. Contrarily, MSC administration partially restored lung elasticity and alveolar architecture. In the absence of evidence that MSCs acquired epithelial phenotype, we detected an increased proliferative activity of aquaporin 5- and surfactant protein C-positive lung cells, suggesting MSC-driven paracrine mechanisms. The data indicate the mediation of hepatocyte growth factor in amplifying MSC-driven tissue response after injury. Our study shed light on supportive properties of lung-derived MSCs, although the full identification of mechanisms orchestrated by MSCs and responsible for epithelial repair after injury is a critical aspect yet to be achieved.

3.
Sci Rep ; 8(1): 2961, 2018 02 13.
Artigo em Inglês | MEDLINE | ID: mdl-29440646

RESUMO

Spermatogenesis depends on endocrine, autocrine and paracrine communications along the hypothalamus-pituitary-gonad axis. Bisphenol A (BPA), an estrogen-mimic endocrine disrupting chemical, is an environmental contaminant used to manufacture polycarbonate plastics and epoxy resins with toxic effects for male reproduction. Here we investigated whether the chronic exposure to low BPA doses affects spermatogenesis through the modulation of SIRT1, a NAD+-dependent deacetylase involved in the progression of spermatogenesis, with outcomes on apoptosis, oxidative stress, metabolism and energy homeostasis. BPA exposure via placenta first, and lactation and drinking water later, affected the body weight gain in male offspring at 45 postnatal days and the first round of spermatogenesis, with impairment of blood testis barrier, reactive oxygen species production, DNA damage and decreased expression of SIRT1. The analysis of SIRT1 downstream molecular pathways revealed the increase of acetyl-p53Lys370, γH2AX foci, the decrease of oxidative stress defenses and the higher apoptotic rate in the testis of treated animals, with partial rescue at sex maturation. In conclusion, SIRT1 pathways disruption after BPA exposure can have serious consequences on the first round of spermatogenesis.


Assuntos
Compostos Benzidrílicos/toxicidade , Fenóis/toxicidade , Sirtuína 1/metabolismo , Espermatogênese/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Compostos Benzidrílicos/sangue , Peso Corporal/efeitos dos fármacos , Dano ao DNA , Relação Dose-Resposta a Droga , Regulação para Baixo/efeitos dos fármacos , Feminino , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Masculino , Estresse Oxidativo/efeitos dos fármacos , Fenóis/sangue , Ratos , Ratos Wistar , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/fisiologia , Fatores de Tempo
4.
Br J Pharmacol ; 174(21): 3696-3712, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-28320043

RESUMO

BACKGROUND AND PURPOSE: Doxorubicin is a highly effective anticancer drug, but its clinical application is hampered by cardiotoxicity. Asymptomatic diastolic dysfunction can be the earliest manifestation of doxorubicin cardiotoxicity. Therefore, a search for therapeutic intervention that can interfere with early manifestations and possibly prevent later development of cardiotoxicity is warranted. Increased doxorubicin-dependent ROS may explain, in part, Ca2+ and Na+ overload that contributes to diastolic dysfunction and development of heart failure. Therefore, we tested whether the administration of ranolazine, a selective blocker of late Na+ current, immediately after completing doxorubicin therapy, could affect diastolic dysfunction and interfere with the progression of functional decline. EXPERIMENTAL APPROACH: Fischer 344 rats received a cumulative dose of doxorubicin of 15 mg·kg-1 over a period of 2 weeks. After the assessment of diastolic dysfunction, the animals were treated with ranolazine (80 mg·kg-1 , daily) for the following 4 weeks. KEY RESULTS: While diastolic and systolic function progressively deteriorated in doxorubicin-treated animals, treatment with ranolazine relieved diastolic dysfunction and prevented worsening of systolic function, decreasing mortality. Ranolazine lowered myocardial NADPH oxidase 2 expression and oxidative/nitrative stress. Expression of the Na+ /Ca2+ exchanger 1 and Nav 1.5 channels was reduced and of the sarcoplasmic/endoplasmic reticulum Ca2+ -ATPase 2 protein was increased. In addition, ranolazine lowered doxorubicin-induced hyper-phosphorylation and oxidation of Ca2+ /calmodulin-dependent protein kinase II, and decreased myocardial fibrosis. CONCLUSIONS AND IMPLICATIONS: Ranolazine, by the increased Na+ influx, induced by doxorubicin, altered cardiac Ca2+ and Na+ handling and attenuated diastolic dysfunction induced by doxorubicin, thus preventing the progression of cardiomyopathy. LINKED ARTICLES: This article is part of a themed section on New Insights into Cardiotoxicity Caused by Chemotherapeutic Agents. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.21/issuetoc.


Assuntos
Doxorrubicina/toxicidade , Ranolazina/farmacologia , Bloqueadores dos Canais de Sódio/farmacologia , Disfunção Ventricular Esquerda/prevenção & controle , Animais , Antibióticos Antineoplásicos/toxicidade , Cálcio/metabolismo , Cardiotoxicidade/etiologia , Cardiotoxicidade/prevenção & controle , Modelos Animais de Doenças , Progressão da Doença , Feminino , Estresse Nitrosativo/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Ratos Endogâmicos F344 , Sódio/metabolismo , Disfunção Ventricular Esquerda/induzido quimicamente
5.
Int J Cardiol ; 241: 330-343, 2017 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-28343765

RESUMO

AIMS: In the present study, we aimed to develop a percutaneous approach and a reproducible methodology for the isolation and expansion of Cardiac Progenitor Cells (CPCs) from EndoMyocardial Biopsies (EMB) in vivo. Moreover, in an animal model of non-ischemic heart failure (HF), we would like to test whether CPCs obtained by this methodology may engraft the myocardium and differentiate. METHODS AND RESULTS: EMB were obtained using a preformed sheath and a disposable bioptome, advanced via right femoral vein in 12 healthy mini pigs, to the right ventricle. EMB were enzymatically dissociated, cells were expanded and sorted for c-kit. We used 3D-Electro-Anatomic Mapping (3D-EAM) to obtain CPCs from 32 patients affected by non-ischemic cardiomyopathy. The in vivo regenerative potential of CPCs was tested in a rodent model of drug-induced non-ischemic cardiomyopathy. c-kit positive CPCs replicative capacity was assessed in 30 patients. Telomere length averaged 7.4±0.4kbp and telomerase activity was present in all preparations (1.7×105 copies). The in situ hybridization experiments showed that injected human CPCs may acquire a neonatal myocyte phenotype given the expression of the alpha-sarcomeric actin together with the presence of the Alu probe, suggesting a beneficial impact on LV performance. CONCLUSIONS: The success in obtaining CPCs characterized by high regenerative potential, in vitro and in vivo, from EMB indicates that harvesting without thoracotomy in patients affected by either ischemic or non-ischemic cardiomyopathy is feasible. These initial results may potentially expand the future application of CPCs to all patients affected by HF not undergoing surgical procedures.


Assuntos
Ecocardiografia Tridimensional/métodos , Endocárdio/citologia , Endocárdio/diagnóstico por imagem , Imageamento Tridimensional/métodos , Miócitos Cardíacos/fisiologia , Células-Tronco/fisiologia , Adulto , Animais , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Miócitos Cardíacos/transplante , Regeneração/fisiologia , Suínos , Porco Miniatura
6.
Br J Pharmacol ; 174(22): 4070-4086, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27922176

RESUMO

BACKGROUND AND PURPOSE: Heart failure with preserved ejection fraction (HFpEF) is a systemic syndrome driven by co-morbidities, and its pathophysiology is poorly understood. Several studies suggesting that dipeptidyl peptidase 4 (DPP4) might be involved in the pathophysiology of heart failure have prompted experimental and clinical investigations of DPP4 inhibitors in the cardiovascular system. Here we have investigated whether the DPP4 inhibitor sitagliptin affected the progression of HFpEF independently of its effects on glycaemia. EXPERIMENTAL APPROACH: Seven-week-old Dahl salt-sensitive rats were fed a high-salt diet for 5 weeks to induce hypertension. Then the rats continued with the high-salt diet and were treated with either sitagliptin (10 mg·kg-1 ) or vehicle for the following 8 weeks. Blood pressure and cardiac function were measured in vivo. Histochemical and molecular biology analyses of myocardium were used to assay cytokines, fibrotic markers, DPP4 and glucagon-like peptide-1 (GLP-1)/GLP-1 receptor. KEY RESULTS: Treatment with sitagliptin attenuated diastolic dysfunction, reduced mortality and reduced cardiac DPP4 activity, along with increased circulating GLP-1 and myocardial expression of GLP-1 receptors. Myocardial levels of pro-inflammatory cytokines (TNF-α, IL-6 and CCL2) were reduced. Sitagliptin treatment decreased the levels of endothelial NOS monomer, responsible for generation of ROS, while the amount of NO-producing dimeric form increased. Markers of oxidative and nitrosative stress were decreased. Moreover, increased collagen deposition and activation of pro-fibrotic signalling, inducing elevated myocardial stiffness, were attenuated by sitagliptin treatment. CONCLUSIONS AND IMPLICATIONS: Sitagliptin positively modulated active relaxation and passive diastolic compliance by decreasing inflammation-related endothelial dysfunction and fibrosis, associated with HFpEF. LINKED ARTICLES: This article is part of a themed section on Targeting Inflammation to Reduce Cardiovascular Disease Risk. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.22/issuetoc and http://onlinelibrary.wiley.com/doi/10.1111/bcp.v82.4/issuetoc.


Assuntos
Anti-Inflamatórios/uso terapêutico , Inibidores da Dipeptidil Peptidase IV/uso terapêutico , Insuficiência Cardíaca/tratamento farmacológico , Fosfato de Sitagliptina/uso terapêutico , Animais , Anti-Inflamatórios/farmacologia , Pressão Sanguínea/efeitos dos fármacos , Diástole/efeitos dos fármacos , Dipeptidil Peptidase 4/metabolismo , Inibidores da Dipeptidil Peptidase IV/farmacologia , Fibrose , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/metabolismo , Coração/efeitos dos fármacos , Coração/fisiologia , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Masculino , Miocárdio/patologia , Óxido Nítrico/metabolismo , Ratos Endogâmicos Dahl , Fosfato de Sitagliptina/farmacologia , Volume Sistólico
7.
Oncotarget ; 7(38): 62312-62326, 2016 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-27694688

RESUMO

New strategies to prevent and early detect the cardiotoxic effects of the anticancer drug doxorubicin (DOXO) are required. MicroRNAs emerged as potential diagnostic, therapeutic and prognostic approaches in cardiovascular diseases. MiR-34a has a role in cardiac dysfunction and ageing and is involved in several cellular processes associated with DOXO cardiotoxicity. Our in vitro and in vivo results indicated that after DOXO exposure the levels of miR-34a are enhanced in cardiac cells, including Cardiac Progenitor Cells (CPCs). Since one of the determining event responsible for the initiation and evolution of the DOXO toxicity arises at the level of the CPC compartment, we evaluated if miR-34a pharmacological inhibition in these cells ameliorates the detrimental aftermath of the drug. AntimiR-34a has beneficial consequences on vitality, proliferation, apoptosis and senescence of DOXO-treated rat CPC. These effects are mediated by an increase of prosurvival miR-34a targets Bcl-2 and SIRT1, accompanied by a decrease of acetylated-p53 and p16INK4a. Importantly, miR-34a silencing also reduces the release of this miRNA from DOXO-exposed rCPCs, decreasing its negative paracrine effects on other rat cardiac cells. In conclusion, the silencing of miR-34a could represent a future therapeutic option for cardioprotection in DOXO toxicity and at the same time, it could be considered as a circulating biomarker for anthracycline-induced cardiac damage.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Cardiomiopatias/sangue , Doxorrubicina/efeitos adversos , Coração/efeitos dos fármacos , MicroRNAs/metabolismo , Mioblastos Cardíacos/metabolismo , Acetilação , Animais , Apoptose/efeitos dos fármacos , Biomarcadores/análise , Biomarcadores/metabolismo , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/genética , Cardiotoxicidade/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/metabolismo , Modelos Animais de Doenças , Células Endoteliais , Feminino , MicroRNAs/antagonistas & inibidores , MicroRNAs/sangue , Mioblastos Cardíacos/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Endogâmicos F344 , Reação em Cadeia da Polimerase em Tempo Real , Sirtuína 1/genética , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/metabolismo
8.
PLoS One ; 11(7): e0158746, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27434719

RESUMO

BACKGROUND: The need for new options for chronic lung diseases promotes the research on stem cells for lung repair. Bone marrow-derived mesenchymal stem cells (MSCs) can modulate lung inflammation, but the data on cellular processes involved in early airway remodeling and the potential involvement of neuropeptides are scarce. OBJECTIVES: To elucidate the mechanisms by which local administration of MSCs interferes with pathophysiological features of airway hyperresponsiveness in an animal model. METHODS: GFP-tagged mouse MSCs were intratracheally delivered in the ovalbumin mouse model with subsequent functional tests, the analysis of cytokine levels, neuropeptide expression and histological evaluation of MSCs fate and airway pathology. Additionally, MSCs were exposed to pro-inflammatory factors in vitro. RESULTS: Functional improvement was observed after MSC administration. Although MSCs did not adopt lung cell phenotypes, cell therapy positively affected airway remodeling reducing the hyperplastic phase of the gain in bronchial smooth muscle mass, decreasing the proliferation of epithelium in which mucus metaplasia was also lowered. Decrease of interleukin-4, interleukin-5, interleukin-13 and increase of interleukin-10 in bronchoalveolar lavage was also observed. Exposed to pro-inflammatory cytokines, MSCs upregulated indoleamine 2,3-dioxygenase. Moreover, asthma-related in vivo upregulation of pro-inflammatory neurokinin 1 and neurokinin 2 receptors was counteracted by MSCs that also determined a partial restoration of VIP, a neuropeptide with anti-inflammatory properties. CONCLUSION: Intratracheally administered MSCs positively modulate airway remodeling, reduce inflammation and improve function, demonstrating their ability to promote tissue homeostasis in the course of experimental allergic asthma. Because of a limited tissue retention, the functional impact of MSCs may be attributed to their immunomodulatory response combined with the interference of neuropeptide system activation and tissue remodeling.


Assuntos
Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/imunologia , Receptores da Neurocinina-1/imunologia , Receptores da Neurocinina-2/imunologia , Hipersensibilidade Respiratória/terapia , Animais , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/imunologia , Expressão Gênica , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Indolamina-Pirrol 2,3,-Dioxigenase/genética , Indolamina-Pirrol 2,3,-Dioxigenase/imunologia , Interleucina-10/genética , Interleucina-10/imunologia , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-4/genética , Interleucina-4/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Intubação Intratraqueal , Pulmão/imunologia , Pulmão/patologia , Células-Tronco Mesenquimais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Ovalbumina , Receptores da Neurocinina-1/genética , Receptores da Neurocinina-2/genética , Hipersensibilidade Respiratória/induzido quimicamente , Hipersensibilidade Respiratória/imunologia , Hipersensibilidade Respiratória/patologia
9.
Int J Cardiol ; 217: 69-79, 2016 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-27179211

RESUMO

BACKGROUND: To investigate the effects of chronic administration of ranolazine (RAN) on experimental model of heart failure with preserved ejection fraction. METHODS: Seven-weeks old Dahl salt-sensitive rats were fed a high salt diet for 5weeks to induce hypertension. Afterwards, rats continued with a high salt diet and were administered either with vehicle or RAN (20mg/kg/die, ip) for the following 8weeks. Control rats were maintained on a low salt diet. RESULTS: While systolic parameters were not altered, diastolic parameters were changed in high salt animals. Hemodynamic analysis showed a decreased dP/dt min, increased LVEDP, longer time constant and steeper slope of the end-diastolic pressure-volume relationship. Treatment with RAN attenuated these alterations and determined a reduction in mortality. Additionally, the magnitude of myocardial hypertrophy and activation of PI3K/Akt pathway were reduced. Alteration in diastolic compliance as a consequence of elevated myocardial stiffness was confirmed by an increase of collagen deposition and activation of pro-fibrotic TGF-ß/SMAD3/CTGF signaling. These effects were counteracted by RAN. High salt rats had a decrease in SERCA2 and an increase in Na(+)/Ca(2+) exchanger (NCX). Treatment with RAN reduced NCX expression and determined an increment of SERCA2. Moreover, the levels of nitrotyrosine and oxidized dyhydroethidium were higher in high salt rats. RAN induced a decrement of oxidative stress, supporting the concept that reduction in ROS may mediate beneficial effects. CONCLUSIONS: Our findings support the possibility that diastolic dysfunction can be attenuated by RAN, indicating its ability to affect active relaxation and passive diastolic compliance.


Assuntos
Fármacos Cardiovasculares/administração & dosagem , Insuficiência Cardíaca/tratamento farmacológico , Ranolazina/administração & dosagem , Volume Sistólico/efeitos dos fármacos , Remodelação Ventricular/efeitos dos fármacos , Animais , Modelos Animais de Doenças , Esquema de Medicação , Insuficiência Cardíaca/metabolismo , Insuficiência Cardíaca/fisiopatologia , Humanos , Hipertensão/induzido quimicamente , Hipertensão/metabolismo , Hipertensão/fisiopatologia , Masculino , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Endogâmicos Dahl , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento
10.
Int J Cardiol ; 205: 99-110, 2016 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-26730840

RESUMO

BACKGROUND: Doxorubicin (DOXO) is an effective anti-neoplastic drug but its clinical benefits are hampered by cardiotoxicity. Oxidative stress, apoptosis and myocardial fibrosis mediate the anthracycline cardiomyopathy. ROS trigger TGF-ß pathway that activates cardiac fibroblasts promoting fibrosis. Myocardial stiffness contributes to diastolic dysfunction, less studied aspect of anthracycline cardiomyopathy. Considering the role of SIRT1 in the inhibition of the TGF-ß/SMAD3 pathway, resveratrol (RES), a SIRT1 activator, might improve cardiac function by interfering with the development of cardiac fibrosis in a model of DOXO-induced cardiomyopathy. METHODS: F344 rats received a cumulative dose of 15 mg/kg of DOXO in 2 weeks or DOXO+RES (DOXO and RES, 2.5mg/kg/day, concomitantly for 2 weeks and then RES alone for 1 more week). The effects of RES on cardiac fibroblasts were also tested in vitro. RESULTS: Along with systolic dysfunction, DOXO was also responsible of diastolic abnormalities. Myocardial stiffness correlated with fibroblast activation and collagen deposition. DOXO+RES co-treatment significantly improved ± dP/dt and, more interestingly, ameliorated end-diastolic pressure/volume relationship. Treatment with RES resulted in reduced fibrosis and fibroblast activation and, most importantly, the mortality rate was significantly reduced in DOXO+RES group. Fibroblasts isolated from DOXO+RES-treated rats, in which SIRT1 was upregulated, showed decreased levels of TGF-ß and pSMAD3/SMAD3 when compared to cells isolated from DOXO-exposed hearts. CONCLUSIONS: Our findings reveal a key role of SIRT1 in supporting animal survival and functional parameters of the heart. SIRT1 activation by interfering with fibrogenesis can improve relaxation properties of myocardium and attenuate myocardial remodeling related to chemotherapy.


Assuntos
Cardiomiopatias/metabolismo , Cardiomiopatias/prevenção & controle , Diástole/efeitos dos fármacos , Doxorrubicina/toxicidade , Sirtuína 1/metabolismo , Estilbenos/uso terapêutico , Animais , Antraciclinas/toxicidade , Antibióticos Antineoplásicos/toxicidade , Cardiomiopatias/induzido quimicamente , Células Cultivadas , Modelos Animais de Doenças , Feminino , Fibrose , Ratos , Ratos Endogâmicos F344 , Resveratrol , Estilbenos/farmacologia
11.
Mediators Inflamm ; 2016: 3917471, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28090152

RESUMO

Structural changes contribute to airway hyperresponsiveness and airflow obstruction in asthma. Emerging evidence points to the involvement of c-kit+ cells in lung homeostasis, although their potential role in asthma is unknown. Our aim was to isolate c-kit+ cells from normal mouse lungs and to test whether these cells can interfere with hallmarks of asthma in an animal model. Adult mouse GFP-tagged c-kit+ cells, intratracheally delivered in the ovalbumin-induced airway hyperresponsiveness, positively affected airway remodeling and improved airway function. In bronchoalveolar lavage fluid of cell-treated animals, a reduction in the number of inflammatory cells and in IL-4, IL-5, and IL-13 release, along with an increase of IL-10, was observed. In MSC-treated mice, the macrophage polarization to M2-like subset may explain, at least in part, the increment in the level of anti-inflammatory cytokine IL-10. After in vitro stimulation of c-kit+ cells with proinflammatory cytokines, the indoleamine 2,3-dioxygenase and TGFß were upregulated. These data, together with the increased apoptosis of inflammatory cells in vivo, indicate that c-kit+ cells downregulate immune response in asthma by influencing local environment, possibly by cell-to-cell contact combined to paracrine action. In conclusion, intratracheally administered c-kit+ cells reduce inflammation, positively modulate airway remodeling, and improve function. These data document previously unrecognized properties of c-kit+ cells, able to impede pathophysiological features of experimental airway hyperresponsiveness.


Assuntos
Hiper-Reatividade Brônquica/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Remodelação das Vias Aéreas , Animais , Asma/imunologia , Lavagem Broncoalveolar , Líquido da Lavagem Broncoalveolar/química , Modelos Animais de Doenças , Proteínas de Fluorescência Verde/química , Homeostase , Sistema Imunitário , Inflamação , Interleucina-10/uso terapêutico , Pulmão/patologia , Macrófagos/citologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C
12.
Cardiooncology ; 2(1): 2, 2016 Mar 03.
Artigo em Inglês | MEDLINE | ID: mdl-33530140

RESUMO

The cardiotoxicity of doxorubicin is becoming an interdisciplinary point of interest given a growing population of cancer survivors. The complex and not completely understood pathogenesis of this complication makes difficult to design successful preventive or curative measures. Although cardiomyocyte has been considered a classical cellular target, other cells including various types of undifferentiated cells are involved in myocardial homeostasis. Such perspective may shed light on previously unrecognized aspects of cardiotoxicity and promote new experimental and clinical cardioprotective strategies. In this review, different cellular targets of doxorubicin are discussed with the focus on cardiac progenitor cells, oxidative stress, DNA damage, senescence and apoptosis all of which contribute to their compromised functional properties.

13.
Int J Cardiol ; 189: 30-44, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25889431

RESUMO

BACKGROUND: The search for compounds able to counteract chemotherapy-induced heart failure is extremely important at the age of global cancer epidemic. The role of SIRT1 in the maintenance of progenitor cell homeostasis may contribute to its cardioprotective effects. SIRT1 activators, by preserving progenitor cells, could have a clinical relevance for the prevention of doxorubicin (DOXO)-cardiotoxicity. METHODS: To determine whether SIRT1 activator, resveratrol (RES), interferes with adverse effects of DOXO on cardiac progenitor cells (CPCs): 1) human CPCs (hCPCs) were exposed in vitro to DOXO or DOXO+RES and their regenerative potential was tested in vivo in an animal model of DOXO-induced heart failure; 2) the in vivo effects of DOXO+RES co-treatment on CPCs were studied in a rat model. RESULTS: In contrast to healthy cells, DOXO-exposed hCPCs were ineffective in a model of anthracycline cardiomyopathy. The in vitro activation of SIRT1 decreased p53 acetylation, overcame suppression of the IGF-1/Akt pro-survival and anti-apoptotic signaling, enhanced oxidative stress defense and prevented senescence and growth arrest of hCPCs. Priming with RES counterbalanced the onset of dysfunctional phenotype in DOXO-exposed hCPCs, partly restoring their ability to repair the damage with improvement in cardiac function and animal survival. The in vivo co-treatment DOXO+RES prevented the anthracycline-induced alterations in CPCs, partly preserving cardiac function. CONCLUSION: SIRT1 activation protects DOXO-exposed CPCs and re-establishes their proper function. Pharmacological intervention at the level of tissue-specific progenitor cells may provide cardiac benefits for the growing population of long-term cancer survivors that are at risk of chemotherapy-induced cardiovascular toxicity.


Assuntos
Cardiomiopatias/patologia , Doxorrubicina/toxicidade , Sirtuína 1/metabolismo , Células-Tronco/efeitos dos fármacos , Estilbenos/farmacologia , Análise de Variância , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Cardiomiopatias/induzido quimicamente , Cardiomiopatias/tratamento farmacológico , Células Cultivadas , Modelos Animais de Doenças , Doxorrubicina/farmacologia , Feminino , Humanos , Imuno-Histoquímica , Miócitos Cardíacos/efeitos dos fármacos , Distribuição Normal , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Resveratrol , Sirtuína 1/efeitos dos fármacos , Estatísticas não Paramétricas , Células-Tronco/fisiologia
14.
Mol Ther ; 21(10): 1841-51, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23760446

RESUMO

Exogenous high-mobility group box 1 protein (HMGB1) administration to the mouse heart, during acute myocardial infarction (MI), results in cardiac regeneration via resident c-kit(+) cell (CPC) activation. Aim of the present study was to identify the molecular pathways involved in HMGB1-induced heart repair. Gene expression profiling was performed to identify differentially expressed genes in the infarcted and bordering regions of untreated and HMGB1-treated mouse hearts, 3 days after MI. Functional categorization of the transcripts, accomplished using Ingenuity Pathway Analysis software (IPA), revealed that genes involved in tissue regeneration, that is, cardiogenesis, vasculogenesis and angiogenesis, were present both in the infarcted area and in the peri-infarct zone; HMGB1 treatment further increased the expression of these genes. IPA revealed the involvement of Notch signaling pathways in HMGB1-treated hearts. Importantly, HMGB1 determined a 35 and 58% increase in cardiomyocytes and CPCs expressing Notch intracellular cytoplasmic domain, respectively. Further, Notch inhibition by systemic treatment with the γ-secretase inhibitor DAPT, which blocked the proteolytic activation of Notch receptors, reduced the number of CPCs, their proliferative fraction, and cardiomyogenic differentiation in HMGB1-treated infarcted hearts. The present study gives insight into the molecular processes involved in HMGB1-mediated cardiac regeneration and indicates Notch signaling as a key player.


Assuntos
Perfilação da Expressão Gênica , Proteína HMGB1/farmacologia , Infarto do Miocárdio/metabolismo , Miocárdio/metabolismo , Receptores Notch/metabolismo , Regeneração/genética , Transdução de Sinais , Animais , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Feminino , Proteína HMGB1/administração & dosagem , Coração/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/fisiopatologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Regeneração/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
15.
Basic Res Cardiol ; 108(2): 334, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23411815

RESUMO

The increasing population of cancer survivors faces considerable morbidity and mortality due to late effects of the antineoplastic therapy. Cardiotoxicity is a major limiting factor of therapy with doxorubicin (DOXO), the most effective anthracycline, and is characterized by a dilated cardiomyopathy that can develop even years after treatment. Studies in animals have proposed the cardiac progenitor cells (CPCs) as the cellular target responsible for DOXO-induced cardiomyopathy but the relevance of these observations to clinical settings is unknown. In this study, the analysis of the DOXO-induced cardiomyopathic human hearts showed that the majority of human CPCs (hCPCs) was senescent. In isolated hCPCs, DOXO triggered DNA damage response leading to apoptosis early after exposure, and telomere shortening and senescence at later time interval. Functional properties of hCPCs, such as migration and differentiation, were also negatively affected. Importantly, the differentiated progeny of DOXO-treated hCPCs prematurely expressed the senescence marker p16(INK4a). In conclusion, DOXO exposure severely affects the population of hCPCs and permanently impairs their function. Premature senescence of hCPCs and their progeny can be responsible for the decline in the regenerative capacity of the heart and may represent the cellular basis of DOXO-induced cardiomyopathy in humans.


Assuntos
Antibióticos Antineoplásicos/efeitos adversos , Cardiomiopatia Dilatada/induzido quimicamente , Senescência Celular/efeitos dos fármacos , Doxorrubicina/efeitos adversos , Mioblastos Cardíacos/efeitos dos fármacos , Células-Tronco/efeitos dos fármacos , Adulto , Antibióticos Antineoplásicos/uso terapêutico , Western Blotting , Cardiomiopatia Dilatada/metabolismo , Cardiomiopatia Dilatada/patologia , Morte Celular/efeitos dos fármacos , Movimento Celular , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Dano ao DNA/efeitos dos fármacos , Doxorrubicina/uso terapêutico , Feminino , Técnica Indireta de Fluorescência para Anticorpo , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas c-kit/metabolismo , Homeostase do Telômero , beta-Galactosidase/metabolismo
16.
J Heart Valve Dis ; 21(1): 125-34, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22474754

RESUMO

BACKGROUND AND AIM OF THE STUDY: The adult human heart contains a cardiac mesenchymal stromal cell (CStC) population with residual cardiovascular plasticity. The study aim was to investigate the ability of CStCs to populate decellularized aortic homograft leaflets, without mechanical stimulation. METHODS: The ability of CStCs to acquire valve endothelial and interstitial cell phenotypes was tested using in vitro assays. First, trypsin-decellularized aortic leaflets were seeded with CStCs under static conditions; tissue section analyses were then performed before and after decellularization, and at 10, 20, and 30 days after reseeding. RESULTS: Following in vitro treatment, the CStCs differentiated along the endothelial lineage, as shown by their capacity to uptake acetylated low-density lipoprotein and to secrete the pro-angiogenic factor, vascular endothelial growth factor. After seeding, CStCs were able to adhere to the leaflet surface, rescuing up to the 90% of the original cell density and expressing the mature endothelial marker, von Willebrandt factor. The CStC supernatants were also positive for matrix metalloprotease-2 (MMP-2), which confirmed the ability of these cells to penetrate within the leaflet structure; this also suggested that CStCs, once engrafted, would contribute to the extracellular matrix turnover. Accordingly, although at a lower efficiency, CStC repopulation was also evident in the inner portions of the leaflet. CONCLUSION: Seeded CStCs were able to reconstitute, without mechanical stimulation, an endothelial-like layer and to partially infiltrate decellularized homograft leaflets. Hence, CStCs appear to be a potentially useful cell type for engineered heart valves.


Assuntos
Valva Aórtica , Bioprótese , Sobrevivência de Enxerto/imunologia , Transplante de Células-Tronco Mesenquimais , Engenharia Tecidual , Transplante Homólogo/métodos , Adulto , Valva Aórtica/imunologia , Valva Aórtica/cirurgia , Diferenciação Celular , Células Endoteliais/transplante , Feminino , Próteses Valvulares Cardíacas , Implante de Prótese de Valva Cardíaca/instrumentação , Implante de Prótese de Valva Cardíaca/métodos , Humanos , Masculino , Falha de Prótese , Engenharia Tecidual/instrumentação , Engenharia Tecidual/métodos , Transplantes , Tretoquinol
17.
PLoS One ; 6(6): e19845, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21731608

RESUMO

AIMS: HMGB1 injection into the mouse heart, acutely after myocardial infarction (MI), improves left ventricular (LV) function and prevents remodeling. Here, we examined the effect of HMGB1 in chronically failing hearts. METHODS AND RESULTS: Adult C57 BL16 female mice underwent coronary artery ligation; three weeks later 200 ng HMGB1 or denatured HMGB1 (control) were injected in the peri-infarcted region of mouse failing hearts. Four weeks after treatment, both echocardiography and hemodynamics demonstrated a significant improvement in LV function in HMGB1-treated mice. Further, HMGB1-treated mice exhibited a ∼23% reduction in LV volume, a ∼48% increase in infarcted wall thickness and a ∼14% reduction in collagen deposition. HMGB1 induced cardiac regeneration and, within the infarcted region, it was found a ∼2-fold increase in c-kit⁺ cell number, a ∼13-fold increase in newly formed myocytes and a ∼2-fold increase in arteriole length density. HMGB1 also enhanced MMP2 and MMP9 activity and decreased TIMP-3 levels. Importantly, miR-206 expression 3 days after HMGB1 treatment was 4-5-fold higher than in control hearts and 20-25 fold higher that in sham operated hearts. HMGB1 ability to increase miR-206 was confirmed in vitro, in cardiac fibroblasts. TIMP3 was identified as a potential miR-206 target by TargetScan prediction analysis; further, in cultured cardiac fibroblasts, miR-206 gain- and loss-of-function studies and luciferase reporter assays showed that TIMP3 is a direct target of miR-206. CONCLUSIONS: HMGB1 injected into chronically failing hearts enhanced LV function and attenuated LV remodelling; these effects were associated with cardiac regeneration, increased collagenolytic activity, miR-206 overexpression and miR-206 -mediated inhibition of TIMP-3.


Assuntos
Proteína HMGB1/farmacologia , Insuficiência Cardíaca/fisiopatologia , MicroRNAs/metabolismo , Regeneração/efeitos dos fármacos , Inibidor Tecidual de Metaloproteinase-3/antagonistas & inibidores , Remodelação Ventricular/efeitos dos fármacos , Animais , Sequência de Bases , Colágeno/metabolismo , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Fibroblastos/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Coração/fisiopatologia , Insuficiência Cardíaca/complicações , Insuficiência Cardíaca/diagnóstico por imagem , Insuficiência Cardíaca/enzimologia , Testes de Função Cardíaca , Hemodinâmica/efeitos dos fármacos , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , MicroRNAs/genética , Dados de Sequência Molecular , Infarto do Miocárdio/complicações , Infarto do Miocárdio/diagnóstico por imagem , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/enzimologia , Miocárdio/patologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/patologia , Regeneração/genética , Análise de Sobrevida , Inibidor Tecidual de Metaloproteinase-3/metabolismo , Ultrassonografia
18.
Circulation ; 120(10): 876-87, 2009 Sep 08.
Artigo em Inglês | MEDLINE | ID: mdl-19704095

RESUMO

BACKGROUND: Cardiac progenitor cells (CPCs) possess the insulin-like growth factor-1 (IGF-1)-IGF-1 receptor system, and IGF-1 can be tethered to self-assembling peptide nanofibers (NF-IGF-1), leading to prolonged release of this growth factor to the myocardium. Therefore, we tested whether local injection of clonogenic CPCs and NF-IGF-1 potentiates the activation and differentiation of delivered and resident CPCs enhancing cardiac repair after infarction. METHODS AND RESULTS: Myocardial infarction was induced in rats, and untreated infarcts and infarcts treated with CPCs or NF-IGF-1 only and CPCs and NF-IGF-1 together were analyzed. With respect to infarcts exposed to CPCs or NF-IGF-1 alone, combination therapy resulted in a greater increase in the ratio of left ventricular mass to chamber volume and a better preservation of +dP/dt, -dP/dt, ejection fraction, and diastolic wall stress. Myocardial regeneration was detected in all treated infarcts, but the number of newly formed myocytes with combination therapy was 32% and 230% higher than with CPCs and NF-IGF-1, respectively. Corresponding differences in the volume of regenerated myocytes were 48% and 115%. Similarly, the length density of newly formed coronary arterioles with both CPCs and NF-IGF-1 was 73% and 83% greater than with CPCs and NF-IGF-1 alone, respectively. Importantly, activation of resident CPCs by paracrine effects contributed to cardiomyogenesis and vasculogenesis. Collectively, CPCs and NF-IGF-1 therapy reduced infarct size more than CPCs and NF-IGF-1 alone. CONCLUSIONS: The addition of nanofiber-mediated IGF-1 delivery to CPC therapy improved in part the recovery of myocardial structure and function after infarction.


Assuntos
Biotina , Fator de Crescimento Insulin-Like I/administração & dosagem , Infarto do Miocárdio/fisiopatologia , Infarto do Miocárdio/cirurgia , Miócitos Cardíacos , Nanoestruturas , Regeneração , Transplante de Células-Tronco , Células-Tronco , Adaptação Fisiológica , Animais , Apoptose , Fusão Celular , Proliferação de Células , Células Cultivadas , Vasos Coronários/fisiopatologia , Feminino , Infarto do Miocárdio/patologia , Miocárdio/patologia , Miócitos Cardíacos/patologia , Ratos , Ratos Endogâmicos F344 , Regeneração/efeitos dos fármacos , Sobrevivência de Tecidos , Função Ventricular
19.
Circ Res ; 102(5): 597-606, 2008 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-18202313

RESUMO

Heart failure is the leading cause of death in the elderly, but whether this is the result of a primary aging myopathy dictated by depletion of the cardiac progenitor cell (CPC) pool is unknown. Similarly, whether current lifespan reflects the ineluctable genetic clock or heart failure interferes with the genetically determined fate of the organ and organism is an important question. We have identified that chronological age leads to telomeric shortening in CPCs, which by necessity generate a differentiated progeny that rapidly acquires the senescent phenotype conditioning organ aging. CPC aging is mediated by attenuation of the insulin-like growth factor-1/insulin-like growth factor-1 receptor and hepatocyte growth factor/c-Met systems, which do not counteract any longer the CPC renin-angiotensin system, resulting in cellular senescence, growth arrest, and apoptosis. However, pulse-chase 5-bromodeoxyuridine-labeling assay revealed that the senescent heart contains functionally competent CPCs that have the properties of stem cells. This subset of telomerase-competent CPCs have long telomeres and, following activation, migrate to the regions of damage, where they generate a population of young cardiomyocytes, reversing partly the aging myopathy. The senescent heart phenotype and heart failure are corrected to some extent, leading to prolongation of maximum lifespan.


Assuntos
Células-Tronco Adultas/efeitos dos fármacos , Envelhecimento/efeitos dos fármacos , Insuficiência Cardíaca/terapia , Fator de Crescimento de Hepatócito/uso terapêutico , Fator de Crescimento Insulin-Like I/uso terapêutico , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco Adultas/metabolismo , Células-Tronco Adultas/patologia , Envelhecimento/patologia , Animais , Antígenos de Diferenciação/biossíntese , Apoptose/efeitos dos fármacos , Contagem de Células , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Inibidor p16 de Quinase Dependente de Ciclina/biossíntese , Modelos Animais de Doenças , Vias de Administração de Medicamentos , Coração/efeitos dos fármacos , Coração/crescimento & desenvolvimento , Insuficiência Cardíaca/patologia , Insuficiência Cardíaca/fisiopatologia , Masculino , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Fenótipo , Ratos , Ratos Endogâmicos F344 , Receptores de Fatores de Crescimento/metabolismo , Regeneração/efeitos dos fármacos , Taxa de Sobrevida , Telômero/metabolismo
20.
Proc Natl Acad Sci U S A ; 105(5): 1668-73, 2008 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-18216245

RESUMO

Coronary artery disease is the most common cause of cardiac failure in the Western world, and to date there is no alternative to bypass surgery for severe coronary atherosclerosis. We report that c-kit-positive cardiac progenitor cells (CPCs) activated with insulin-like growth factor 1 and hepatocyte growth factor before their injection in proximity of the site of occlusion of the left coronary artery in rats, engrafted within the host myocardium forming temporary niches. Subsequently, CPCs divided and differentiated into endothelial cells and smooth muscle cells and, to a lesser extent, into cardiomyocytes. The acquisition of vascular lineages appeared to be mediated by the up-regulation of hypoxia-inducible factor 1alpha, which promoted the synthesis and secretion of stromal-derived factor 1 from hypoxic coronary vessels. Stromal-derived factor 1 was critical in the conversion of CPCs to the vascular fate. CPCs formed conductive and intermediate-sized coronary arteries together with resistance arterioles and capillaries. The new vessels were connected with the primary coronary circulation, and this increase in vascularization more than doubled myocardial blood flow in the infarcted myocardium. This beneficial effect, together with myocardial regeneration attenuated postinfarction dilated myopathy, reduced infarct size and improved function. In conclusion, locally delivered activated CPCs generate de novo coronary vasculature and may be implemented clinically for restoration of blood supply to the ischemic myocardium.


Assuntos
Vasos Coronários/fisiologia , Mioblastos Cardíacos/fisiologia , Neovascularização Fisiológica , Regeneração , Células-Tronco/fisiologia , Animais , Diferenciação Celular , Quimiocina CXCL12/metabolismo , Vasos Coronários/citologia , Células Endoteliais/citologia , Feminino , Fator de Crescimento de Hepatócito/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator de Crescimento Insulin-Like I/farmacologia , Mioblastos Cardíacos/efeitos dos fármacos , Mioblastos Cardíacos/transplante , Isquemia Miocárdica/metabolismo , Miócitos de Músculo Liso/citologia , Proteínas Proto-Oncogênicas c-kit/análise , Ratos , Ratos Endogâmicos F344 , Transplante de Células-Tronco , Células-Tronco/química , Células-Tronco/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...