Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Molecules ; 28(19)2023 Sep 26.
Artigo em Inglês | MEDLINE | ID: mdl-37836650

RESUMO

It is estimated that 250 million people worldwide are affected by schistosomiasis. Disease transmission is related to the poor sanitation and hygiene habits that affect residents of impoverished regions in tropical and subtropical countries. The main species responsible for causing disease in humans are Schistosoma Mansoni, S. japonicum, and S. haematobium, each with different geographic distributions. Praziquantel is the drug predominantly used to treat this disease, which offers low effectiveness against immature and juvenile parasite forms. In addition, reports of drug resistance prompt the development of novel therapeutic approaches. Natural products represent an important source of new compounds, especially those obtained from plant sources. This review compiles data from several in vitro and in vivo studies evaluating various compounds and essential oils derived from plants with cercaricidal and molluscicidal activities against both juvenile and adult forms of the parasite. Finally, this review provides an important discussion on recent advances in molecular and computational tools deemed fundamental for more rapid and effective screening of new compounds, allowing for the optimization of time and resources.


Assuntos
Anti-Helmínticos , Produtos Biológicos , Esquistossomose , Humanos , Animais , Anti-Helmínticos/farmacologia , Anti-Helmínticos/uso terapêutico , Schistosoma haematobium , Produtos Biológicos/farmacologia , Produtos Biológicos/uso terapêutico , Esquistossomose/tratamento farmacológico , Esquistossomose/parasitologia , Praziquantel/farmacologia , Schistosoma mansoni
2.
Am J Pathol ; 187(5): 1134-1146, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28322201

RESUMO

Chronic Chagas disease cardiomyopathy, caused by Trypanosoma cruzi infection, is a major cause of heart failure in Latin America. Galectin-3 (Gal-3) has been linked to cardiac remodeling and poor prognosis in heart failure of different etiologies. Herein, we investigated the involvement of Gal-3 in the disease pathogenesis and its role as a target for disease intervention. Gal-3 expression in mouse hearts was evaluated during T. cruzi infection by confocal microscopy and flow cytometry analysis, showing a high expression in macrophages, T cells, and fibroblasts. In vitro studies using Gal-3 knockdown in cardiac fibroblasts demonstrated that Gal-3 regulates cell survival, proliferation, and type I collagen synthesis. In vivo blockade of Gal-3 with N-acetyl-d-lactosamine in T. cruzi-infected mice led to a significant reduction of cardiac fibrosis and inflammation in the heart. Moreover, a modulation in the expression of proinflammatory genes in the heart was observed. Finally, histological analysis in human heart samples obtained from subjects with Chagas disease who underwent heart transplantation showed the expression of Gal-3 in areas of inflammation, similar to the mouse model. Our results indicate that Gal-3 plays a role in the pathogenesis of experimental chronic Chagas disease, favoring inflammation and fibrogenesis. Moreover, by demonstrating Gal-3 expression in human hearts, our finding reinforces that this protein could be a novel target for drug development for Chagas cardiomyopathy.


Assuntos
Cardiomiopatia Chagásica/metabolismo , Galectina 3/metabolismo , Miocardite/metabolismo , Miocárdio/patologia , Acetilgalactosamina/farmacologia , Animais , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Doença Crônica , Colágeno Tipo I/biossíntese , Fibrose/etiologia , Fibrose/metabolismo , Galectina 3/antagonistas & inibidores , Transplante de Coração , Humanos , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Miocardite/etiologia , Miocárdio/metabolismo , Miofibroblastos/metabolismo , Linfócitos T/metabolismo
3.
Exp Parasitol ; 159: 29-36, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26297681

RESUMO

The main pathogenic event caused by Schistosoma mansoni infection is characterized by a granulomatous inflammatory reaction around parasite eggs and fibrosis in the liver. We have previously shown that transplantation of bone marrow cells (BMC) promotes a reduction in liver fibrosis in chronically S. mansoni-infected mice. Here we investigated the presence and phenotype of bone marrow-derived cells in livers of S. mansoni-infected mice. During the chronic phase of infection, C57BL/6 mice had an increased number of circulating mesenchymal stem cells and endothelial progenitor cells in the peripheral blood when compared to uninfected controls. In order to investigate the fate of BMC in the liver, we generated bone marrow chimeric mice by transplanting BMC from transgenic green fluorescent protein (GFP) mice into lethally irradiated wild-type C57BL/6 mice. S. mansoni-infected chimeric mice did not demonstrate increased mortality and developed similar liver histopathological features, when compared to wild-type S. mansoni-infected mice. GFP(+) bone marrow-derived cells were found in the liver parenchyma, particularly in periportal regions. CD45(+)GFP(+) cells were found in the granulomas. Flow cytometry analysis of digested liver tissue characterized GFP(+) cells as lymphocytes, myeloid cells and stem cells. GFP(+) cells were also found in areas of collagen deposition, although rare GFP(+) cells expressed the myofibroblast cell marker α-SMA. Additionally GFP(+) endothelial cells (co-stained with von Willebrand factor) were frequently observed, while BMC-derived hepatocytes (GFP(+) albumin(+) cells) were sparsely found in the liver of chimeric mice chronically infected with S. mansoni. In conclusion, BMC are recruited to the liver during chronic experimental infection with S. mansoni and contribute to the generation of different cell types involved, not only in disease pathogenesis, but possibly in liver regeneration and repair.


Assuntos
Células da Medula Óssea/fisiologia , Fígado/patologia , Esquistossomose mansoni/patologia , Animais , Movimento Celular , Quimera , Doença Crônica , Feminino , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/patologia , Fígado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
4.
Neurochem Res ; 39(2): 259-68, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24343530

RESUMO

Status epilepticus (SE) is a severe clinical manifestation of epilepsy associated with intense neuronal loss and inflammation, two key factors involved in the pathophysiology of temporal lobe epilepsy. Bone marrow mononuclear cells (BMMC) attenuated the consequences of pilocarpine-induced SE, including neuronal loss, in addition to frequency and duration of seizures. Here we investigated the effects of BMMC transplanted early after the onset of SE in mice, as well as the involvement of soluble factors produced by BMMC in the effects of the cell therapy. Mice were injected with pilocarpine for SE induction and randomized into three groups: transplanted intravenously with 1 × 10(7) BMMC isolated from GFP transgenic mice, injected with BMMC lysate, and saline-treated controls. Cell tracking, neuronal counting in hippocampal subfields and cytokine analysis in the serum and brain were performed. BMMC were found in the brain 4 h following transplantation and their numbers progressively decreased until 24 h following transplantation. A reduction in hippocampal neuronal loss after SE was found in mice treated with live BMMC and BMMC lysate when compared to saline-treated, SE-induced mice. Moreover, the expression of inflammatory cytokines IL-1ß, TNF-α, IL-6 was decreased after injection of live BMMC and to a lesser extent, of BMMC lysate, when compared to SE-induced controls. In contrast, IL-10 expression was increased. Analysis of markers for microglia activation demonstrated a reduction of the expression of genes related to type 1-activation. BMMC transplantation promotes neuroprotection and mediates anti-inflammatory effects following SE in mice, possibly through the secretion of soluble factors.


Assuntos
Transplante de Medula Óssea , Fármacos Neuroprotetores , Pilocarpina/administração & dosagem , Estado Epiléptico/induzido quimicamente , Animais , Sequência de Bases , Citocinas/biossíntese , Primers do DNA , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Microglia/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estado Epiléptico/cirurgia
5.
Neurobiol Dis ; 46(2): 302-13, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22198377

RESUMO

Approximately 30% of patients with mesial temporal lobe epilepsy do not respond to treatment with antiepileptic drugs. We have previously shown that transplantation of mononuclear bone marrow cells (BMC) has an anticonvulsant effect in acute epilepsy. Here, we used pilocarpine to induce epilepsy in rats and studied the effects of BMC injected intravenously either at the onset of seizures or after 10 months of recurrent seizures. BMC effectively decreased seizure frequency and duration. In addition, decreased levels of proinflammatory cytokines (TNF-α, IL-1ß and IL-6) and increased levels of anti-inflammatory cytokine (IL-10) were observed in the brain and serum of BMC-treated rats. Transplants performed at seizure-onset protected against pilocarpine-induced neuronal loss and gliosis and stimulated the proliferation of new neurons in epileptic rats. Our data demonstrate that BMC transplantation has potent therapeutic effects and could be a potential therapy for clinically intractable epilepsies.


Assuntos
Transplante de Medula Óssea , Citocinas/biossíntese , Epilepsia/metabolismo , Epilepsia/cirurgia , Leucócitos Mononucleares/transplante , Neurônios/metabolismo , Animais , Transplante de Medula Óssea/métodos , Transplante de Medula Óssea/patologia , Movimento Celular/fisiologia , Epilepsia/patologia , Incidência , Mediadores da Inflamação/metabolismo , Leucócitos Mononucleares/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurônios/patologia , Ratos , Ratos Wistar
6.
Cytotherapy ; 14(3): 339-49, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22149185

RESUMO

BACKGROUND AIMS: Cirrhosis, end-stage liver disease, is caused by different mechanisms of injury, associated with persistent inflammation. Galectin-3 is an important regulator of fibrosis that links chronic inflammation to fibrogenesis. We investigated the role of bone marrow cell (BMC) transplantation in chronic inflammation and hepatic fibrosis. METHODS: Liver cirrhosis was induced by administration of carbon tetrachloride and ethanol to wild-type C57BL/6 or bone marrow chimeric mice. Bone marrow chimeras were generated by lethal irradiation and transplantation with BMC obtained from green fluorescent protein (GFP(+) )donors. Wild-type cirrhotic mice were transplanted with BMC without irradiation. Livers from chimeras and cirrhotic transplanted mice were obtained for evaluation of inflammation, fibrosis and regulatory factors [galectin-3, matrix metallopeptidase (MMP)-9, tissue inhibitor of metalloproteinase (TIMP)-1 and transforming growth factor (TGF)-ß]. RESULTS: The development of cirrhosis was associated with increased expression of galectin-3 by F4/80(+) cells and intense migration of BMC to the liver. Furthermore, when transplanted after the establishment of cirrhosis, BMC also migrated to the liver and localized within the fibrous septa. Two months after BMC therapy, cirrhotic mice had a significant reduction in liver fibrosis and expression of type I collagen. We did not find any difference in levels of TGF-ß, TIMP-1 and MMP-9 between saline and BMC groups. However, the numbers of inflammatory cells, phagocytes and galectin-3(+) cells were markedly lower in the livers of cirrhotic mice treated with BMC. CONCLUSIONS: Our results demonstrate an important role for BMC in the regulation of liver fibrosis and that transplantation of BMC can accelerate fibrosis regression through modulatory mechanisms.


Assuntos
Transplante de Medula Óssea/métodos , Galectina 3/metabolismo , Cirrose Hepática Experimental/terapia , Animais , Células da Medula Óssea/citologia , Células da Medula Óssea/metabolismo , Tetracloreto de Carbono/administração & dosagem , Tetracloreto de Carbono/efeitos adversos , Movimento Celular , Quimera , Colágeno Tipo I/metabolismo , Etanol/administração & dosagem , Etanol/efeitos adversos , Feminino , Proteínas de Fluorescência Verde/metabolismo , Inflamação , Fígado/metabolismo , Fígado/patologia , Masculino , Metaloproteinase 9 da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fagócitos/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Quimeras de Transplante
7.
Biochem Biophys Res Commun ; 405(4): 604-9, 2011 Feb 25.
Artigo em Inglês | MEDLINE | ID: mdl-21272565

RESUMO

Recent studies have demonstrated that communication takes place between the autophagic and phagocytic pathways, indicating that the convergence of these two pathways plays an important role in the innate immune response against intracellular microbes. The present study investigated the effect of autophagic induction on the phagocytic capacity of murine macrophages. Autophagy induced by physiological and pharmacological means was shown to reduce the phagocytic capacity of murine macrophages, regardless of cell origin or the nature of the phagocytosed particles themselves. This autophagic inhibitory effect on phagocytosis was shown to be an early and reversible event that results in no loss of cell viability. Furthermore, the data presented herein demonstrate that the induction of autophagy does not affect a macrophage's capacity to recognize and bind to particles, indicating that autophagy does not inhibit the particle recognition process, even though particle internalization is suppressed. The findings herein support the notion that phagocytosis and autophagy may be interdependent and complementary processes.


Assuntos
Autofagia , Macrófagos/fisiologia , Fagocitose , Animais , Linhagem Celular , Leishmania , Macrófagos/microbiologia , Macrófagos/parasitologia , Camundongos , Camundongos Endogâmicos BALB C , Saccharomyces cerevisiae
8.
Stem Cell Res ; 38: 101454, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31078962

RESUMO

Sickle cell disease (SCD) is one of the most prevalent and severe monogenetic disorders. Previously, we generated iPS cell lines from SCD patients. Here, we generated iPS cell lines from three age-, ethnicity- and gender-matched healthy individuals as control cell lines. Cell reprogramming was performed using erythroblasts expanded from PBMC by a non-integrative method. SCD-iPSC controls expressed pluripotency markers, presented a normal karyotype, were able to differentiate into the three germ layers in embryoid body spontaneous differentiation and confirmed to be integration-free. The cell lines generated here may be used as matched healthy controls for SCD studies.


Assuntos
Anemia Falciforme , Técnicas de Reprogramação Celular , Eritroblastos , Células-Tronco Pluripotentes Induzidas/metabolismo , Anemia Falciforme/genética , Anemia Falciforme/metabolismo , Anemia Falciforme/patologia , Técnicas de Cultura de Células , Linhagem Celular , Eritroblastos/metabolismo , Eritroblastos/patologia , Humanos , Células-Tronco Pluripotentes Induzidas/patologia
9.
Stem Cell Res Ther ; 10(1): 146, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31113444

RESUMO

BACKGROUND: Survival and therapeutic actions of bone marrow-derived mesenchymal stem cells (BMMSCs) can be limited by the hostile microenvironment present during acute spinal cord injury (SCI). Here, we investigated whether BMMSCs overexpressing insulin-like growth factor 1 (IGF-1), a cytokine involved in neural development and injury repair, improved the therapeutic effects of BMMSCs in SCI. METHODS: Using a SCI contusion model in C57Bl/6 mice, we transplanted IGF-1 overexpressing or wild-type BMMSCs into the lesion site following SCI and evaluated cell survival, proliferation, immunomodulation, oxidative stress, myelination, and functional outcomes. RESULTS: BMMSC-IGF1 transplantation was associated with increased cell survival and recruitment of endogenous neural progenitor cells compared to BMMSC- or saline-treated controls. Modulation of gene expression of pro- and anti-inflammatory mediators was observed after BMMSC-IGF1 and compared to saline- and BMMSC-treated mice. Treatment with BMMSC-IGF1 restored spinal cord redox homeostasis by upregulating antioxidant defense genes. BMMSC-IGF1 protected against SCI-induced myelin loss, showing more compact myelin 28 days after SCI. Functional analyses demonstrated significant gains in BMS score and gait analysis in BMMSC-IGF1, compared to BMMSC or saline treatment. CONCLUSIONS: Overexpression of IGF-1 in BMMSC resulted in increased cell survival, immunomodulation, myelination, and functional improvements, suggesting that IGF-1 facilitates the regenerative actions of BMMSC in acute SCI.


Assuntos
Fator de Crescimento Insulin-Like I/genética , Transplante de Células-Tronco Mesenquimais , Células-Tronco Neurais/transplante , Traumatismos da Medula Espinal/terapia , Animais , Células da Medula Óssea/citologia , Diferenciação Celular/genética , Modelos Animais de Doenças , Humanos , Células-Tronco Mesenquimais/citologia , Camundongos , Bainha de Mielina/genética , Bainha de Mielina/patologia , Células-Tronco Neurais/citologia , Recuperação de Função Fisiológica , Regeneração/genética , Traumatismos da Medula Espinal/genética , Traumatismos da Medula Espinal/patologia
10.
Stem Cells Int ; 2017: 3282656, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28769980

RESUMO

Therapies based on transplantation of mesenchymal stromal cells (MSC) hold promise for the management of inflammatory disorders. In chronic Chagas disease cardiomyopathy (CCC), caused by chronic infection with Trypanosoma cruzi, the exacerbated immune response plays a critical pathophysiological role and can be modulated by MSC. Here, we investigated the role of galectin-3 (Gal-3), a beta-galactoside-binding lectin with several actions on immune responses and repair process, on the immunomodulatory potential of MSC. Gal-3 knockdown in MSC did not affect the immunophenotype or differentiation potential. However, Gal-3 knockdown MSC showed decreased proliferation, survival, and migration. Additionally, when injected intraperitoneally into mice with CCC, Gal-3 knockdown MSC showed impaired migration in vivo. Transplantation of control MSC into mice with CCC caused a suppression of cardiac inflammation and fibrosis, reducing expression levels of CD45, TNFα, IL-1ß, IL-6, IFNγ, and type I collagen. In contrast, Gal-3 knockdown MSC were unable to suppress the immune response or collagen synthesis in the hearts of mice with CCC. Finally, infection with T. cruzi demonstrated parasite survival in wild-type but not in Gal-3 knockdown MSC. These findings demonstrate that Gal-3 plays a critical role in MSC survival, proliferation, migration, and therapeutic potential in CCC.

11.
Stem Cell Res Ther ; 5(4): 81, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24984860

RESUMO

INTRODUCTION: New therapeutic options are necessary for patients with chronic Chagas disease, a leading cause of heart failure in Latin American countries. Stem cell therapy focused on improving cardiac function is a promising approach for treating heart disease. Here, we evaluated the therapeutic effects of cardiac mesenchymal stem cells (CMSCs) in a mouse model of chronic Chagas disease. METHODS: CMSCs were isolated from green fluorescent protein (GFP) transgenic C57BL/6 mouse hearts and tested for adipogenic, osteogenic, chondrogenic, endothelial, and cardiogenic differentiation potentials evaluated by histochemical and immunofluorescence techniques. A lymphoproliferation assay was performed to evaluate the immunomodulatory activity of CMSCs. To investigate the therapeutic potential of CMSCs, C57BL/6 mice chronically infected with Trypanosoma cruzi were treated with 106 CMSCs or saline (control) by echocardiography-guided injection into the left ventricle wall. All animals were submitted to cardiac histopathological and immunofluorescence analysis in heart sections from chagasic mice. Analysis by quantitative real-time reverse transcription polymerase chain reaction (qRT-PCR) was performed in the heart to evaluate the expression of cytokines involved in the inflammatory response. RESULTS: CMSCs demonstrated adipogenic, osteogenic, and chondrogenic differentiation potentials. Moreover, these cells expressed endothelial cell and cardiomyocyte features upon defined stimulation culture conditions and displayed immunosuppressive activity in vitro. After intramyocardial injection, GFP+ CMSCs were observed in heart sections of chagasic mice one week later; however, no observed GFP+ cells co-expressed troponin T or connexin-43. Histopathological analysis revealed that CMSC-treated mice had a significantly decreased number of inflammatory cells, but no reduction in fibrotic area, two months after treatment. Analysis by qRT-PCR demonstrated that cell therapy significantly decreased tumor necrosis factor-alpha expression and increased transforming growth factor-beta in heart samples. CONCLUSIONS: We conclude that the CMSCs exert a protective effect in chronic chagasic cardiomyopathy primarily through immunomodulation.


Assuntos
Cardiomiopatia Chagásica/terapia , Transplante de Células-Tronco Mesenquimais , Miocárdio/citologia , Animais , Diferenciação Celular , Conexina 43/metabolismo , Proteínas de Fluorescência Verde/análise , Imunomodulação , Masculino , Células-Tronco Mesenquimais/citologia , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Miocardite , Miocárdio/metabolismo , Troponina T/metabolismo , Trypanosoma cruzi , Fator de Necrose Tumoral alfa
13.
Arq Bras Cardiol ; 100(5): 460-8, 2013 May.
Artigo em Inglês, Português | MEDLINE | ID: mdl-23568098

RESUMO

BACKGROUND: Chagas disease, caused by the protozoan Trypanosoma cruzi, is a major cause of heart failure in Latin America. Tissue therapy has been investigated as a possible therapeutic option for patients with cardiovascular disease. OBJECTIVE: This study evaluated the effects of therapy with mesenchymal stem cells in an experimental model of chronic Chagasic cardiomyopathy. METHODS: C57BL/6 mice were infected with 1000 trypomastigotes from the Colombian strain of T. cruzi and, after six months of infection, were treated with mesenchymal human stem cells from adipose tissue (STAT) or with Dulbecco/Vogt modified Eagle's minimal essential medium - DMEM (control). The treated group received two intraperitoneal injections of STAT (1x10(6) cells/dose), with a month interval between the two doses. Before and after the first and second months of treatment, the chagasic and normal control animals underwent cardiopulmonary exercise testing and electrocardiography. All animals were sacrificed under anesthesia after two months of treatment for histopathological analysis of the heart. RESULTS: No improvement was observed in arrhythmias and cardiovascular function in the group of animals treated with STAT; however, sections of mice hearts in this group revealed a significant reduction in the number of inflammatory cells (p<0.0001) and areas of fibrosis (p<0.01) in comparison with chagasic animals treated with DMEM. CONCLUSION: Thus, it is concluded that administration of intraperitoneal STAT can reduce inflammation and fibrosis in the heart of mice chronically infected with T. cruzi; however, there were no effects on the cardiac function two months after transplantation.


Assuntos
Tecido Adiposo/citologia , Cardiomiopatia Chagásica/cirurgia , Transplante de Células-Tronco Mesenquimais , Trypanosoma cruzi , Análise de Variância , Animais , Arritmias Cardíacas/metabolismo , Cardiomiopatia Chagásica/patologia , Cardiomiopatia Chagásica/fisiopatologia , Modelos Animais de Doenças , Fibrose , Inflamação/metabolismo , Injeções Intraperitoneais , Camundongos , Camundongos Endogâmicos C57BL , Condicionamento Físico Animal/métodos , Distribuição Aleatória
17.
Arq. bras. cardiol ; 100(5): 460-468, maio 2013. ilus, tab
Artigo em Português | LILACS | ID: lil-675608

RESUMO

FUNDAMENTO: A doença de Chagas, causada pelo protozoário Trypanosoma cruzi, é uma das mais importantes causas de insuficiência cardíaca na América Latina. A terapia celular vem sendo investigada como uma possível opção terapêutica para pacientes com doenças cardiovasculares. OBJETIVO: O objetivo deste estudo foi avaliar os efeitos da terapia com células-tronco mesenquimais em um modelo experimental de cardiomiopatia chagásica crônica. MÉTODOS: Camundongos C57BL/6 foram infectados com 1000 tripomastigotas da cepa Colombiana de T. cruzi e, após seis meses de infecção, foram tratados com células-tronco mesenquimais derivadas de tecido adiposo humano (CTTAs) ou com meio DMEM (controle). O grupo tratado recebeu duas injeções intraperitoneais de CTTAs (1x106 células / dose), com um mês de intervalo entre as duas doses. Antes e após 1 e 2 meses de tratamento, os animais chagásicos e controles normais foram submetidos à eletrocardiograma e teste ergoespirométrico. Todos os animais foram sacrificados sob anestesia após 2 meses de tratamento, para análise histopatológica do coração. RESULTADOS: Não foi observada melhora de arritmias e da função cardiovascular no grupo tratado com CTTAs, porém secções de corações de camundongos deste grupo apresentaram uma redução significativa do número de células inflamatórias (p < 0,0001) e da área de fibrose (p < 0,01) em comparação com animais chagásicos tratados com DMEM. CONCLUSÃO: Deste modo, conclui-se que a administração de CTTAs por via intraperitoneal é capaz de reduzir inflamação e fibrose no coração de camundongos cronicamente infectados por T. cruzi, porém não teve efeitos na função cardíaca dois meses após o transplante.


BACKGROUND: Chagas disease, caused by the protozoan Trypanosoma cruzi, is a major cause of heart failure in Latin America. Tissue therapy has been investigated as a possible therapeutic option for patients with cardiovascular disease. OBJECTIVE: This study evaluated the effects of therapy with mesenchymal stem cells in an experimental model of chronic Chagasic cardiomyopathy. METHODS: C57BL/6 mice were infected with 1000 trypomastigotes from the Colombian strain of T. cruzi and, after six months of infection, were treated with mesenchymal human stem cells from adipose tissue (STAT) or with Dulbecco/Vogt modified Eagle's minimal essential medium - DMEM (control). The treated group received two intraperitoneal injections of STAT (1x10(6) cells/dose), with a month interval between the two doses. Before and after the first and second months of treatment, the chagasic and normal control animals underwent cardiopulmonary exercise testing and electrocardiography. All animals were sacrificed under anesthesia after two months of treatment for histopathological analysis of the heart. RESULTS: No improvement was observed in arrhythmias and cardiovascular function in the group of animals treated with STAT; however, sections of mice hearts in this group revealed a significant reduction in the number of inflammatory cells (p<0.0001) and areas of fibrosis (p<0.01) in comparison with chagasic animals treated with DMEM. CONCLUSION: Thus, it is concluded that administration of intraperitoneal STAT can reduce inflammation and fibrosis in the heart of mice chronically infected with T. cruzi; however, there were no effects on the cardiac function two months after transplantation.


Assuntos
Animais , Camundongos , Tecido Adiposo/citologia , Cardiomiopatia Chagásica/cirurgia , Transplante de Células-Tronco Mesenquimais , Trypanosoma cruzi , Análise de Variância , Arritmias Cardíacas/metabolismo , Cardiomiopatia Chagásica/patologia , Cardiomiopatia Chagásica/fisiopatologia , Modelos Animais de Doenças , Fibrose , Injeções Intraperitoneais , Inflamação/metabolismo , Condicionamento Físico Animal/métodos , Distribuição Aleatória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA