Carine Machado Azevedo
Rafael Advanced Defense Systems
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Carine Machado Azevedo.
Neurobiology of Disease | 2012
Zaquer Suzana Munhoz Costa-Ferro; Bruno Solano de Freitas Souza; Marcos Maurício Tosta Leal; Carla Martins Kaneto; Carine Machado Azevedo; Igor Campos da Silva; Milena Botelho Pereira Soares; Ricardo Ribeiro-dos-Santos; Jaderson Costa DaCosta
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-1β 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.
Scientific Reports | 2016
Bruno Solano de Freitas Souza; Gabriela Louise de Almeida Sampaio; Ciro Silveira E. Pereira; Gubio Soares Campos; Silvia Ines Sardi; Luiz Antonio Rodrigues de Freitas; Cláudio Pereira Figueira; Bruno Diaz Paredes; Carolina Kymie Vasques Nonaka; Carine Machado Azevedo; Vinícius Pinto Costa Rocha; Antonio Carlos Bandeira; Rosalia Mendez-Otero; Ricardo Ribeiro dos Santos; Milena Botelho Pereira Soares
Zika virus (ZIKV) infection has been associated with severe complications both in the developing and adult nervous system. To investigate the deleterious effects of ZIKV infection, we used human neural progenitor cells (NPC), derived from induced pluripotent stem cells (iPSC). We found that NPC are highly susceptible to ZIKV and the infection results in cell death. ZIKV infection led to a marked reduction in cell proliferation, ultrastructural alterations and induction of autophagy. Induction of apoptosis of Sox2+ cells was demonstrated by activation of caspases 3/7, 8 and 9, and by ultrastructural and flow cytometry analyses. ZIKV-induced death of Sox2+ cells was prevented by incubation with the pan-caspase inhibitor, Z-VAD-FMK. By confocal microscopy analysis we found an increased number of cells with supernumerary centrosomes. Live imaging showed a significant increase in mitosis abnormalities, including multipolar spindle, chromosome laggards, micronuclei and death of progeny after cell division. FISH analysis for chromosomes 12 and 17 showed increased frequency of aneuploidy, such as monosomy, trisomy and polyploidy. Our study reinforces the link between ZIKV and abnormalities in the developing human brain, including microcephaly.
Cytotherapy | 2012
Sheilla Andrade de Oliveira; Bruno Solano de Freitas Souza; Elton Sá Barreto; Carla Martins Kaneto; Hélio Almeida Neto; Carine Machado Azevedo; Elisalva Teixeira Guimarães; Luiz Antonio Rodrigues de Freitas; Ricardo Ribeiro-dos-Santos; Milena Botelho Pereira Soares
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)-β]. 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-β, 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.
Stem Cell Research & Therapy | 2014
Daniela Nascimento Silva; Bruno Solano de Freitas Souza; Carine Machado Azevedo; Juliana Fraga Vasconcelos; Rejane Hughes Carvalho; Milena Botelho Pereira Soares; Ricardo Ribeiro dos Santos
IntroductionNew 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.MethodsCMSCs 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.ResultsCMSCs 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.ConclusionsWe conclude that the CMSCs exert a protective effect in chronic chagasic cardiomyopathy primarily through immunomodulation.
Arquivos Brasileiros De Cardiologia | 2013
Ticiana Ferreira Larocca; Bruno Solano de Freitas Souza; Cristina Aragão Silva; Carla Martins Kaneto; Adriano Costa de Alcantara; Carine Machado Azevedo; Murilo Fagundes Castro; Simone Garcia Macambira; Milena Botelho Pereira Soares; Ricardo Ribeiro-dos-Santos
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 Eagles 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.
International Journal of Experimental Pathology | 2014
Bruno Solano de Freitas Souza; Carine Machado Azevedo; Ricardo Santana de Lima; Carla Martins Kaneto; Juliana Fraga Vasconcelos; Elisalva Teixeira Guimarães; Ricardo Ribeiro dos Santos; Milena Botelho Pereira Soares
Infection by Trypanosoma cruzi, the aetiological agent of Chagas disease, causes an intense inflammatory reaction in several tissues, including the myocardium. We have previously shown that transplantation of bone marrow cells (BMC) ameliorates the myocarditis in a mouse model of chronic Chagas disease. We investigated the participation of BMC in lesion repair in the heart and skeletal muscle, caused by T. cruzi infection in mice. Infection with a myotropic T. cruzi strain induced an increase in the percentage of stem cells and monocytes in the peripheral blood, as well as in gene expression of chemokines SDF‐1, MCP1, 2, and 3 in the heart and skeletal muscle. To investigate the fate of BMC within the damaged tissue, chimeric mice were generated by syngeneic transplantation of green fluorescent protein (GFP+) BMC into lethally irradiated mice and infected with Trypanosoma cruzi. Migration of GFP+ BMC to the heart and skeletal muscle was observed during and after the acute phase of infection. GFP+ cardiomyocytes and endothelial cells were present in heart sections of chimeric chagasic mice. GFP+ myofibres were observed in the skeletal muscle of chimeric mice at different time points following infection. In conclusion, BMC migrate and contribute to the formation of new resident cells in the heart and skeletal muscle, which can be detected both during the acute and the chronic phase of infection. These findings reinforce the role of BMC in tissue regeneration.
Stem Cells International | 2017
Bruno Solano de Freitas Souza; Kátia Nunes da Silva; Daniela Nascimento Silva; Vinícius Pinto Costa Rocha; Bruno Diaz Paredes; Carine Machado Azevedo; Carolina Kymie Vasques Nonaka; Gisele B. Carvalho; Juliana Fraga Vasconcelos; Ricardo Ribeiro dos Santos; Milena Botelho Pereira Soares
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-1β, 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.
Surgical and Experimental Pathology | 2018
Ticiana Ferreira Larocca; Bruno Solano de Freitas Souza; Carine Machado Azevedo; Juliana Fraga Vasconcelos; Daniela Nascimento Silva; Diogo Crispim Nascimento Portella; Washington Luis Conrado dos Santos; Fabio Tavora; João David de Souza Neto; Ricardo Ribeiro dos Santos; Milena Botelho Pereira Soares
BackgroundChronic Chagas cardiomyopathy (CCC) is characterized by the presence of a multifocal inflammatory response and myocardial damage, leading to fibrosis, arrhythmias and ventricular dysfunction. The expression of syndecan-4, a transmembrane proteoglycan, was previously found to be increased in the hearts of mice chronically infected with Trypanosoma cruzi. The possible involvement of syndecan-4 in the disease pathogenesis, however, remains unknown. Here we evaluated the pattern of expression of syndecan-4 in the heart tissue of T. cruzi infected mice and subjects with Chagas cardiomyopathy, correlating with the degree of inflammation and fibrosis.MethodsThe expression of syndecan-4 was evaluated by immunofluorescence and RT-qPCR in the hearts of C57Bl/6 mice at different time points after infection with the Colombian strain of T. cruzi. Immunostainings for syndecan-4 were performed in heart samples obtained from CCC patients and other etiologies of heart failure. The number of infiltrating inflammatory cells and area of fibrosis were also evaluated and quantified.ResultsIn the experimental model, the number of infiltrating inflammatory cells and fibrosis area in the hearts progressively increased after the acute phase of infection, while syndecan-4 expression remained elevated in similar levels in both the acute and chronic phases. Confocal microscopy analysis demonstrated the localization of syndecan-4 expression in blood vessels, co-localized with α-SMA, a marker for vascular smooth muscle cells (VSMCs). Confocal microscopy analysis of human hearts samples showed a similar pattern of syndecan-4 expression in blood vessels. No correlation between syndecan-4 expression and inflammation or fibrosis was found in the hearts from subjects with CCC. We also compared the expression of syndecan-4 evaluated in subjects with CCC, idiopathic dilated cardiomyopathy and ischemic cardiomyopathy. No differences in the number of syndecan-4 positive vessels/mm2 were found comparing the three groups (P = 0.466), whereas CCC patients presented a higher number of infiltrating inflammatory cells, compared to the other etiologies of heart failure. Additionally, no correlation between syndecan-4 and fibrosis or numbers of inflammatory cells was found.ConclusionsSyndecan-4 is expressed in the heart during the acute and chronic phases of Chagas disease, in association with VSMCs, independently of the degree of myocardial fibrosis or the number of infiltrating inflammatory cells.
Stem Cells International | 2018
Daniela Nascimento Silva; Bruno S. Souza; Carine Machado Azevedo; Juliana Fraga Vasconcelos; Paloma G. de Jesus; Malena S. Feitoza; Cássio Santana Meira; Gisele B. Carvalho; Bruno Raphael Ribeiro Cavalcante; Ricardo Ribeiro-dos-Santos; Milena Botelho Pereira Soares
Mesenchymal stem/stromal cells (MSCs) have been investigated for the treatment of diseases that affect the cardiovascular system, including Chagas disease. MSCs are able to promote their beneficial actions through the secretion of proregenerative and immunomodulatory factors, including insulin-like growth factor-1 (IGF-1), which has proregenerative actions in the heart and skeletal muscle. Here, we evaluated the therapeutic potential of IGF-1-overexpressing MSCs (MSC_IGF-1) in a mouse model of chronic Chagas disease. C57BL/6 mice were infected with Colombian strain Trypanosoma cruzi and treated with MSCs, MSC_IGF-1, or vehicle (saline) six months after infection. RT-qPCR analysis confirmed the presence of transplanted cells in both the heart and skeletal muscle tissues. Transplantation of either MSCs or MSC_IGF-1 reduced the number of inflammatory cells in the heart when compared to saline controls. Moreover, treatment with MSCs or MSC_IGF-1 significantly reduced TNF-α, but only MSC treatment reduced IFN-γ production compared to the saline group. Skeletal muscle sections of both MSC- and MSC_IGF-1-treated mice showed a reduction in fibrosis compared to saline controls. Importantly, the myofiber area was reduced in T. cruzi-infected mice, and this was recovered after treatment with MSC_IGF-1. Gene expression analysis in the skeletal muscle showed a higher expression of pro- and anti-inflammatory molecules in MSC_IGF-1-treated mice compared to MSCs alone, which significantly reduced the expression of TNF-α and IL-1β. In conclusion, our results indicate the therapeutic potential of MSC_IGF-1, with combined immunomodulatory and proregenerative actions to the cardiac and skeletal muscles.
Frontiers in Immunology | 2018
Daniela Nascimento Silva; Bruno S. Souza; Juliana Fraga Vasconcelos; Carine Machado Azevedo; Clarissa Valim; Bruno Diaz Paredes; Vinicius P. C. Rocha; Gisele B. Carvalho; Pamela S. Daltro; Simone Garcia Macambira; Carolina Kymie Vasques Nonaka; Ricardo Ribeiro-dos-Santos; Milena Botelho Pereira Soares
Genetic modification of mesenchymal stem cells (MSCs) is a promising strategy to improve their therapeutic effects. Granulocyte-colony stimulating factor (G-CSF) is a growth factor widely used in the clinical practice with known regenerative and immunomodulatory actions, including the mobilization of regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs). Here we evaluated the therapeutic potential of MSCs overexpressing G-CSF (MSC_G-CSF) in a model of inflammatory cardiomyopathy due to chronic Chagas disease. C57BL/6 mice were treated with wild-type MSCs, MSC_G-CSF, or vehicle (saline) 6 months after infection with Trypanosoma cruzi. Transplantation of MSC_G-CSF caused an increase in the number of circulating leukocytes compared to wild-type MSCs. Moreover, G-CSF overexpression caused an increase in migration capacity of MSCs to the hearts of infected mice. Transplantation of either MSCs or MSC_G-CSF improved exercise capacity, when compared to saline-treated chagasic mice. MSC_G-CSF mice, however, were more potent than MSCs in reducing the number of infiltrating leukocytes and fibrosis in the heart. Similarly, MSC_G-CSF-treated mice presented significantly lower levels of inflammatory mediators, such as IFNγ, TNFα, and Tbet, with increased IL-10 production. A marked increase in the percentage of Tregs and MDSCs in the hearts of infected mice was seen after administration of MSC_G-CSF, but not MSCs. Moreover, Tregs were positive for IL-10 in the hearts of T. cruzi-infected mice. In vitro analysis showed that recombinant hG-CSF and conditioned medium of MSC_G-CSF, but not wild-type MSCs, induce chemoattraction of MDSCs in a transwell assay. Finally, MDSCs purified from hearts of MSC_G-CSF transplanted mice inhibited the proliferation of activated splenocytes in a co-culture assay. Our results demonstrate that G-CSF overexpression by MSCs potentiates their immunomodulatory effects in our model of Chagas disease and suggest that mobilization of suppressor cell populations such as Tregs and MDSCs as a promising strategy for the treatment of chronic Chagas disease. Finally, our results reinforce the therapeutic potential of genetic modification of MSCs, aiming at increasing their paracrine actions.