Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
Más filtros










Intervalo de año de publicación
1.
Biomed J ; 45(4): 629-641, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-34333108

RESUMEN

BACKGROUND: Progressive renal fibrosis is an underlying pathological process of chronic kidney disease (CKD) evolution. This study aimed to evaluate the roles of bone-marrow-derived mesenchymal stem cells (MSC) in the remodeling of fibrotic kidney parenchyma in the two kidneys-one clip (2K1C) CKD animal model. METHODS: Wistar rats were allocated into three groups: Sham, 2K1C, and 2K1C þ MSC. MSCs (106) were transplanted into the renal subcapsular region two weeks after clipping the left renal artery. Six weeks after clipping, left kidney samples were analyzed using histological and western blotting techniques. ANOVA tests were performed and differences between groups were considered statistically significant if p < 0.05. RESULTS: Clipped kidneys of 2K1C rats displayed renal fibrosis, with excessive collagen deposition, glomerulosclerosis and renal basement membrane disruption. Clipped kidneys of 2K1C þ MSC rats showed preserved Bowman's capsule and tubular basement membranes, medullary tubules morphological reconstitution and reduced collagen deposits. Expression levels of matrix metalloproteinase (MMP)-2 and MMP-9 were elevated, whereas tissue inhibitor of MMPs (TIMP)-1 and TIMP-2 levels were decreased in clipped kidneys of 2K1C rats. MSCs transplantation restored these expression levels. Moreover, MSCs suppressed macrophages and myofibroblasts accumulation, as well as TNF-a expression in clipped kidneys of 2K1C animals. MSCs transplantation significantly increased IL-10 expression. CONCLUSIONS: Transplanted MSCs orchestrate anti-fibrotic and anti-inflammatory events, which reverse renal fibrosis and promote renal morphological restoration. This study supports the notion that only one MSCs delivery into the renal subcapsular region represents a possible therapeutic strategy against renal fibrosis for CKD treatment.


Asunto(s)
Hipertensión Renovascular , Células Madre Mesenquimatosas , Insuficiencia Renal Crónica , Animales , Médula Ósea , Colágeno/metabolismo , Fibrosis , Hipertensión Renovascular/metabolismo , Hipertensión Renovascular/patología , Interleucina-10/metabolismo , Riñón/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratas , Ratas Wistar , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Insuficiencia Renal Crónica/terapia , Inhibidor Tisular de Metaloproteinasa-2/metabolismo
2.
Life Sci ; 281: 119768, 2021 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-34186042

RESUMEN

AIMS: The purpose of this work was to study the effects of mesenchymal stem cells conditioned medium (MSC CM) treatment in animals with cholestatic liver fibrosis. MATERIALS AND METHODS: We induced cholestatic liver fibrosis by bile duct ligation in C57Bl/6 mice. In the 5th and 6th days after bile duct ligation proceeding, conditioned medium obtained of cultures of mesenchymal stem cells derived from adipose tissue was injected in the animals. Blood levels of hepatic transaminases, alkaline phosphatase and albumin were measured in each group. Analysis of collagen deposition was realized by Picro Sirius red staining and cytokine profiling was performed by cytometric bead array (CBA). KEY FINDINGS: Our results showed that MSC CM treatment decreased levels of hepatic enzymes and collagen deposition in the liver. After MSC CM treatment, profibrotic IL-17A was decreased andIL-6 and IL-4 were increased. SIGNIFICANCE: In summary, MSC CM treatment demonstrated therapeutic potential to cholestatic liver fibrosis, favoring matrix remodeling and cytokine profile towards liver regeneration.


Asunto(s)
Colestasis/patología , Cirrosis Hepática/patología , Células Madre Mesenquimatosas/citología , Tejido Adiposo/citología , Animales , Colestasis/metabolismo , Colágeno/metabolismo , Medios de Cultivo Condicionados , Citocinas/metabolismo , Citometría de Flujo , Cirrosis Hepática/metabolismo , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL
3.
Cell Tissue Res ; 384(3): 721-734, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33977324

RESUMEN

Bone marrow cells (BMCs) from obese Swiss mice fed with Western diet show mitochondrial dysfunction. Obesity interferes with BMCs disrupting energetic metabolism, stimulating apoptosis, and reducing cell proliferation since adipose tissue releases inflammatory adipokines into the medullar microenvironment. These changes lead to reduction of BMC differentiation capacity and hematopoiesis impairment, a process responsible for blood cell continuous production through hematopoietic stem cells (HSCs). This work aimed to analyze the effects of IGF-1 therapy on BMC viability in Western diet-induced obesity, in vivo. We observed that after only 1 week of treatment, obese Swiss mice presented reduced body weight and visceral fat and increased mitochondrial oxidative capacity and coupling, indicating mitochondrial function improvement. In addition, IGF-1 was able to reduce apoptosis of total BMCs, stem cell subpopulations (hematopoietic and mesenchymal), and leukocytes, restoring all progenitor hematopoietic lineages. The treatment also contributed to increase proliferative capacity of hematopoietic stem cells and leukocytes, keeping the hematopoietic and immune systems balanced. Therefore, we conclude that IGF-1 short period therapy improved BMC survival, proliferation, and differentiation capacity in obese Swiss mice.


Asunto(s)
Células de la Médula Ósea , Factor I del Crecimiento Similar a la Insulina/farmacología , Obesidad , Animales , Apoptosis/efectos de los fármacos , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Masculino , Ratones , Ratones Obesos , Mitocondrias/efectos de los fármacos , Obesidad/tratamiento farmacológico , Obesidad/patología
4.
Life Sci ; 278: 119510, 2021 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-33865879

RESUMEN

Currently, the world has been devastated by an unprecedented pandemic in this century. The Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the agent of coronavirus disease 2019 (COVID-19), has been causing disorders, dysfunction and morphophysiological alterations in multiple organs as the disease evolves. There is a great scientific community effort to obtain a therapy capable of reaching the multiple affected organs in order to contribute for tissue repair and regeneration. In this regard, mesenchymal stem cells (MSCs) have emerged as potential candidates concerning the promotion of beneficial actions at different stages of COVID-19. MSCs are promising due to the observed therapeutic effects in respiratory preclinical models, as well as in cardiac, vascular, renal and nervous system models. Their immunomodulatory properties and secretion of paracrine mediators, such as cytokines, chemokines, growth factors and extracellular vesicles allow for long range tissue modulation and, particularly, blood-brain barrier crossing. This review focuses on SARS-CoV-2 impact to lungs, kidneys, heart, vasculature and central nervous system while discussing promising MSC's therapeutic mechanisms in each tissue. In addition, MSC's therapeutic effects in high-risk groups for COVID-19, such as obese, diabetic and hypertensive patients are also explored.


Asunto(s)
COVID-19/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , COVID-19/inmunología , COVID-19/patología , Humanos , Inmunomodulación , Células Madre Mesenquimatosas/inmunología , SARS-CoV-2/inmunología , SARS-CoV-2/aislamiento & purificación
5.
Artículo en Inglés | MEDLINE | ID: mdl-34988430

RESUMEN

AIMS: To analyze therapeutic potential of the conditioned medium from adipose tissue-derived stem cells (ASC) cultivated in 2D (CM-2D) and 3D (CM-3D) models, in mice with Type 1 diabetes (T1D) induced by streptozotocin. MAIN METHODS: Viability andCD105 expression of 2D and 3D ASC were analyzed by flow cytometry. T1D was induced in mice by multiple injections of streptozocin. On the 28th and 29th days after the first injection of streptozocin, diabetic animals received CM-2D or CM-3D. Pancreatic, CM-2D, and CM-3D cytokines were analyzed by cytometric bead array (CBA) and insulin and PDX-1 were observed and quantified by immunohistochemistry. Apoptosis-related proteins were quantified by Western Blotting. KEY FINDINGS: ASC in three-dimensional culture released increased levels of IL-6 and IL-2, while IL-4 was decreased. CM-2D induced pancreatic PDX-1 expression and was able to reduce glycemia in diabetic mice one week after injections but not CM-3D. On the other hand, CM-2D and CM-3D were not able to reverse apoptosis of pancreatic cells in diabetic mice nor to increase insulin expression. SIGNIFICANCE: Together, these results demonstrate that the 3D cell culture secretome was not able to improve diabetes type 1 symptoms at the times observed, while 2D cell secretome improved glycemic levels in T1D mice.

6.
Nutr Metab Cardiovasc Dis ; 30(1): 151-161, 2020 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-31753790

RESUMEN

BACKGROUND AND AIMS: Cardiovascular diseases are the main cause of mortality in obesity. Despite advanced understanding, the mechanisms that regulate cardiac progenitor cells (CPC) survival in pathological conditions are not clear. Low IGF-1 plasma levels are correlated to obesity, cardiomyopathy and CPC death, so this work aimed to investigate IGF-1 therapeutic potential on cardiomyopathy and its relationship with the survival, proliferation and differentiation of CPC in Western diet-induced obesity. METHODS AND RESULTS: Male Swiss mice were divided into control group (CG, n = 8), fed with standard diet; and obese group (OG, n = 16), fed with Western diet, for 12 weeks. At 11th week, OG was subdivided to receive a daily subcutaneous injection of human recombinant IGF-1 (100 µg.Kg-1) for seven consecutive days (OG + IGF1, n = 8). Results showed that IGF-1 therapy improved the metabolic parameters negatively impacted by western diet in OG, reaching levels similar to CG. OG + IGF-1 also demonstrated restored heart energetic metabolism, fibrosis resolution, decreased apoptosis level, restored cardiac gap junctions and intracellular calcium balance. Cardiomyopathy improvement was accompanied by increased CPC survival, proliferation and newly cardiomyocytes formation related to increased pAkt/Akt ratio. CONCLUSION: These results suggest that only one week of IGF-1 therapy has cardioprotective effects through Akt pathway upregulation, ensuring CPC survival and differentiation, contributing to heart failure rescue.


Asunto(s)
Cardiomiopatías/prevención & control , Factor I del Crecimiento Similar a la Insulina/administración & dosificación , Miocitos Cardíacos/efectos de los fármacos , Obesidad/tratamiento farmacológico , Células Madre/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Señalización del Calcio , Cardiomiopatías/etiología , Cardiomiopatías/metabolismo , Cardiomiopatías/patología , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Esquema de Medicación , Uniones Comunicantes/efectos de los fármacos , Uniones Comunicantes/metabolismo , Uniones Comunicantes/patología , Inyecciones Subcutáneas , Masculino , Ratones , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Obesidad/complicaciones , Fosforilación , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Recombinantes/administración & dosificación , Células Madre/metabolismo , Células Madre/patología , Factores de Tiempo , Remodelación Ventricular/efectos de los fármacos
7.
Cells ; 8(11)2019 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-31671842

RESUMEN

Fibrosis is a common feature in most pathogenetic processes in the liver, and usually results from a chronic insult that depletes the regenerative capacity of hepatocytes and activates multiple inflammatory pathways, recruiting resident and circulating immune cells, endothelial cells, non-parenchymal hepatic stellate cells, and fibroblasts, which become activated and lead to excessive extracellular matrix accumulation. The ongoing development of liver fibrosis results in a clinically silent and progressive loss of hepatocyte function, demanding the constant need for liver transplantation in clinical practice, and motivating the search for other treatments as the chances of obtaining compatible viable livers become scarcer. Although initially cell therapy has emerged as a plausible alternative to organ transplantation, many factors still challenge the establishment of this technique as a main or even additional therapeutic tool. Herein, the authors discuss the most recent advances and point out the corners and some controversies over several protocols and models that have shown promising results as potential candidates for cell therapy for liver fibrosis, presenting the respective mechanisms proposed for liver regeneration in each case.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Cirrosis Hepática/terapia , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Células Endoteliales/patología , Células Endoteliales/fisiología , Hepatocitos/fisiología , Humanos , Cirrosis Hepática/patología , Cirrosis Hepática/fisiopatología , Fallo Hepático/patología , Fallo Hepático/fisiopatología , Fallo Hepático/terapia , Regeneración Hepática/fisiología , Células Madre/fisiología
8.
Exp Physiol ; 104(5): 740-754, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30790372

RESUMEN

NEW FINDINGS: What is the central question of this study? Can a single bone marrow mononuclear cell (BMMC) transplant into the subcapsular region of kidney improve cellular communication and adhesion, while restoring renal tissue cytoarchitecture and function during renovascular hypertension? What is the main finding and its importance? The BMMC transplantation restored connexin 40 expression and led to recovery of N- and E-cadherin levels within 15 days. It was observed, for the first time, that BMMC transplantation restores expression of nephrin, a component of the glomerular filtration barrier related to podocytes and the glomerular basal membrane. ABSTRACT: Stem cell therapy has emerged as a potential treatment for renal diseases owing to the regenerative potential of stem cells. However, a better understanding of the morphological and functional changes of damaged renal cells in the presence of transplanted stem cells is needed. The aim of this study was to investigate cell-cell communication and adhesion in renal parenchyma, with analysis of fibrosis, to evaluate renal morphology and function after bone marrow mononuclear cell (BMMC) transplantation in two-kidney-one-clip rats. The BMMC therapy significantly decreased blood pressure and renin expression, improved renal morphology and restored the glomerular filtration barrier, with remodelling of podocytes. In addition, there was a reduction in fibrosis, and connexin 40 and nephrin expression were significantly increased after 7 and 15 days of transplantation. Plasma creatinine, urea and total protein levels were restored, and proteinuria was reduced. Furthermore, N- and E-cadherin expression was increased soon after BMMC therapy. Green fluorescent protein-positive BMMCs were found in the renal cortex 24 and 48 h after transplantation into the renal subcapsule, and at 7 and 15 days after transplantation, these cells were observed throughout the renal medulla, indicating cellular migration. Therefore, these data suggest that transplanted BMMCs improve cell-cell communication and adhesion between damaged cells, which is accompanied by a recovery of renal morphology and function.


Asunto(s)
Trasplante de Médula Ósea/métodos , Barrera de Filtración Glomerular/patología , Hipertensión Renovascular/patología , Hipertensión Renovascular/terapia , Uniones Intercelulares/patología , Animales , Presión Sanguínea , Cadherinas/metabolismo , Comunicación Celular , Fibrosis , Riñón/patología , Corteza Renal/patología , Masculino , Monocitos/trasplante , Podocitos/patología , Ratas , Ratas Wistar , Renina/biosíntesis
9.
Artículo en Inglés | MEDLINE | ID: mdl-29853957

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is recognized as the most common cause of liver dysfunction worldwide and is commonly associated with obesity. Evidences suggest that NAFLD might be a mitochondrial disease, which contributes to the hepatic steatosis, oxidative stress, cytokine release, and cell death. Capybara oil (CO) is a rich source of polyunsaturated fatty acids (PUFA), which is known to improve inflammation and oxidative stress. In order to determine the effects of CO on NAFLD, C57Bl/6 mice were divided into 3 groups and fed a high-fat diet (HFD) (NAFLD group and NAFLD + CO group) or a control diet (CG group) during 16 weeks. The CO (1.5 g/kg/daily) was administered by gavage during the last 4 weeks of the diet protocol. We evaluated plasma liver enzymes, hepatic steatosis, and cytokine expression in liver as well as hepatocyte ultrastructural morphology and mitochondrial function. CO treatment suppressed hepatic steatosis, attenuated inflammatory response, and decreased plasma alanine aminotransferase (ALT) in mice with NAFLD. CO was also capable of restoring mitochondrial ultrastructure and function as well as balance superoxide dismutase and catalase levels. Our findings indicate that CO treatment has positive effects on NAFLD improving mitochondrial dysfunction, steatosis, acute inflammation, and oxidative stress.

10.
Stem Cell Rev Rep ; 14(4): 535-545, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-29667027

RESUMEN

Nutritional changes in the development (intrauterine life and postnatal period) may trigger long-term pathophysiological complications such as obesity and cardiovascular disease. Metabolic programming leads to organs and tissues modifications, including adipose tissue, with increased lipogenesis, production of inflammatory cytokines, and decreased glucose uptake. However, stem cells participation in adipose tissue dysfunctions triggered by overfeeding during lactation has not been elucidated. Therefore, this study was the first to evaluate the effect of metabolic programming on adipose mesenchymal stem cells (ASC) from mice submitted to overfeeding during lactation, using the litter reduction model. Cells were evaluated for proliferation capacity, viability, immunophenotyping, and reactive oxygen species (ROS) production. The content of UCP-2 and PGC1-α was determined by Western Blot. ASC differentiation potential in adipogenic and osteogenic environments was also evaluated, as well the markers of adipogenic differentiation (PPAR-γ and FAB4) and osteogenic differentiation (osteocalcin) by RT-qPCR. Results indicated that neonatal overfeeding does not affect ASC proliferation, ROS production, and viability. However, differentiation potential and proteins related to metabolism were altered. ASC from overfed group presented increased adipogenic differentiation, decreased osteogenic differentiation, and also showed increased PGC1-α protein content and reduced UCP-2 expression. Thus, ASC may be involved with the increased adiposity observed in neonatal overfeeding, and its therapeutic potential may be affected.


Asunto(s)
Diferenciación Celular , Lactancia/fisiología , Células Madre Mesenquimatosas/citología , Grasa Subcutánea/citología , Animales , Animales Recién Nacidos , Células Cultivadas , Conducta Alimentaria , Femenino , Expresión Génica , Masculino , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteocalcina/genética , PPAR gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Grasa Subcutánea/metabolismo
11.
Placenta ; 64: 7-16, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29626983

RESUMEN

INTRODUCTION: Bone marrow cells (BMC) from obese adult mice display an increased apoptosis rate over proliferation. Hematopoietic stem cells (HSC) form all blood cells and are important BMC used in cell therapy. Because it is known that prenatal development can be affected by adverse metabolic epigenetic programming from the maternal organism, this work aimed to investigate the effects of maternal overweight on placenta and fetal liver hematopoietic niches. METHODS: Overweight was induced in female mice by overfeeding during lactation. After Swiss females were mated with healthy males, fetuses at 19 dpc (day post conception) and placentas were analyzed. Maternal biometric parameters were compared, and hematopoiesis in the dissociated placenta and fetal liver cells was analyzed by flow cytometry. Placenta morphology and protein content were also studied. RESULTS: The model induced accumulation of adipose tissue, weight gain, and maternal hyperglycemia. Placentas from the overfed group (OG) displayed altered morphology, higher carbohydrate and lipid deposition, and increased protein content of fibronectin and PGC-1α. Cytometric analysis showed that placentas from OG presented a higher percentage of circulating macrophages, endothelial progenitor cells, HSC, and progenitor cells. No difference was detected in the percentage of neutrophil granulocytes and total leukocytes or in the proliferation of total cells, HSC, or total leukocytes. With regard to liver analysis of the OG group, there was a significant increase in circulating macrophages, primitive HSC, and oval cells but no difference in hematopoietic progenitor cells, total leukocytes, or leukocyte or total cell proliferation. CONCLUSION: Unregulated maternal metabolism can affect hematopoietic populations within the placenta and fetal liver.


Asunto(s)
Hematopoyesis , Sobrepeso/fisiopatología , Placenta/fisiopatología , Complicaciones del Embarazo/fisiopatología , Animales , Animales Recién Nacidos , Biometría , Femenino , Feto/patología , Hígado/patología , Masculino , Ratones , Sobrepeso/metabolismo , Sobrepeso/patología , Placenta/metabolismo , Placenta/patología , Embarazo , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/patología
12.
PLoS One ; 12(11): e0187970, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29176797

RESUMEN

Bone marrow cells (BMC) migrate to the injured liver after transplantation, contributing to regeneration through multiple pathways, but mechanisms involved are unclear. This work aimed to study BMC migration, characterize cytokine profile, cell populations and proliferation in mice with liver fibrosis transplanted with GFP+ BMC. Confocal microscopy analysis showed GFP+ BMC near regions expressing HGF and SDF-1 in the fibrotic liver. Impaired liver cell proliferation in fibrotic groups was restored after BMC transplantation. Regarding total cell populations, there was a significant reduction in CD68+ cells and increased Ly6G+ cells in transplanted fibrotic group. BMC contributed to the total populations of CD144, CD11b and Ly6G cells in the fibrotic liver, related to an increment of anti-fibrotic cytokines (IL-10, IL-13, IFN-γ and HGF) and reduction of pro-inflammatory cytokines (IL-17A and IL-6). Therefore, HGF and SDF-1 may represent important chemoattractants for transplanted BMC in the injured liver, where these cells can give rise to populations of extrahepatic macrophages, neutrophils and endothelial progenitor cells that can interact synergistically with other liver cells towards the modulation of an anti-fibrotic cytokine profile promoting the onset of liver regeneration.


Asunto(s)
Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Comunicación Celular , Colestasis/terapia , Citocinas/metabolismo , Hepatocitos/metabolismo , Cirrosis Hepática/terapia , Animales , Movimiento Celular , Proliferación Celular , Quimiocina CXCL12/metabolismo , Colestasis/complicaciones , Colestasis/genética , Colestasis/patología , Colágeno/metabolismo , Citocinas/genética , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Proteínas Fluorescentes Verdes/metabolismo , Factor de Crecimiento de Hepatocito/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/complicaciones , Cirrosis Hepática/genética , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa
13.
Cell Tissue Res ; 369(2): 287-301, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28374150

RESUMEN

Renovascular hypertension (RVH) is a progressive disease, leading to chronic kidney disease when untreated and no specific treatment is available. Therefore, development of new therapeutic modalities is imperative. RVH is triggered by renal artery stenosis and subsequent renin-angiotensin-aldosterone system activation; it can be experimentally induced by the 2 Kidneys-1 Clip (2K1C) model. This study investigates the therapeutic potential of renal subcapsular mesenchymal stem cell (MSC) infusion in 2K1C rats. Renal morphological and functional changes were analyzed, including Na++K+-ATPase activity and expression, renin angiotensin-converting enzyme (ACE) and angiotensin-II type 1 (AT1R) and type 2 (AT2R) receptors expression. 2K1C rats developed hypertension accompanied by renin upregulation (clipped kidney) and renal Na++K+-ATPase activity and expression reduction. MSC therapy decreased systolic blood pressure, renin, ACE, and AT1R, upregulated AT2R and podocin expression and restored renal Na++K+-ATPase activity and expression. In addition, MSC improved renal morphology, reduced fibrosis and TGF-ß expression in the clipped kidney, decreased proteinuria and restored protein plasma levels. In conclusion, transplantation into a renal subcapsule is an efficient route and MSC is a good candidate for cell therapy, which may represent an interesting approach for chronic kidney disease treatment.


Asunto(s)
Células de la Médula Ósea/citología , Hipertensión Renovascular/enzimología , Hipertensión Renovascular/fisiopatología , Riñón/enzimología , Riñón/fisiopatología , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas/citología , ATPasa Intercambiadora de Sodio-Potasio/metabolismo , Animales , Apoptosis , Presión Sanguínea , Proliferación Celular , Rastreo Celular , Colágeno/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Hipertensión Renovascular/patología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Riñón/metabolismo , Riñón/patología , Pruebas de Función Renal , Masculino , Proteínas de la Membrana/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Ratas Wistar , Receptor de Angiotensina Tipo 1 , Receptor de Angiotensina Tipo 2 , Renina , Sístole , Regulación hacia Arriba , Proteína X Asociada a bcl-2/metabolismo
14.
Lasers Med Sci ; 31(5): 841-8, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26886589

RESUMEN

Low-level infrared laser is considered safe and effective for treatment of muscle injuries. However, the mechanism involved on beneficial effects of laser therapy are not understood. The aim was to evaluate cell viability, reactive oxygen species, apoptosis, and necrosis in myoblast cultures exposed to low-level infrared laser at therapeutic fluences. C2C12 myoblast cultures at different (2 and 10 %) fetal bovine serum (FBS) concentrations were exposed to low-level infrared laser (808 nm, 100 mW) at different fluences (10, 35, and 70 J/cm(2)) and evaluated after 24, 48, and 72 h. Cell viability was evaluated by WST-1 assay; reactive oxygen species (ROS), apoptosis, and necrosis were evaluated by flow cytometry. Cell viability was decreased atthe lowest FBS concentration. Laser exposure increased the cell viability in myoblast cultures at 2 % FBS after 48 and 72 h, but no significant increase in ROS was observed. Apoptosis was decreased at the higher fluence and necrosis was increased at lower fluence in myoblast cultures after 24 h of laser exposure at 2 % FBS. No laser-induced alterations were obtained at 10 % FBS. Results show that level of reactive oxygen species is not altered, at least to those evaluated in this study, but low-level infrared laser exposure affects cell viability, apoptosis, and necrosis in myoblast cultures depending on laser fluence and physiologic conditions of cells.


Asunto(s)
Apoptosis/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Terapia por Luz de Baja Intensidad/métodos , Necrosis/radioterapia , Especies Reactivas de Oxígeno/efectos de la radiación , Animales , Apoptosis/efectos de los fármacos , Bovinos , Mioblastos
15.
J Nutr Biochem ; 29: 124-32, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26608021

RESUMEN

Several studies have demonstrated that overnutrition during early postnatal period can increase the long-term risk of developing obesity and cardiac disorders, yet the short-term effects of postnatal overfeeding in cardiac metabolism remains unknown. The aim of our study was to investigate the cardiac metabolism of weaned mice submitted to overnutrition during lactation, particularly as to mitochondrial function, substrate preference and insulin signaling. Postnatal overfeeding was induced by litter size reduction in mice at postnatal day 3. At 21 days of age (weaning), mice in the overfed group (OG) presented biometric and biochemical parameters of obesity, including increased body weight, visceral fat, liver weight and increased left ventricle weight/tibia length ratio; indicating cardiac hypertrophy, hyperglycemia, hyperinsulinemia and increased liver glycogen content compared to control group. In the heart, we detected impaired insulin signaling, mainly due to decreased IRß, pTyr-IRS1, PI3K, GLUT4 and pAkt/Akt and increased PTP1B, GLUT1 and pAMPKα/AMPKα content. Activities of lactate dehydrogenase and citrate synthase were increased, accompanied by enhanced carbohydrate oxidation, as observed by high-resolution respirometry. Moreover, OG hearts had lower CPT1, PPARα and increased UCP2 mRNA expression, associated with increased oxidative stress (4-HNE content), BAX/BCL2 ratio and cardiac fibrosis. Ultrastructural analysis of OG hearts demonstrated mild mitochondrial damage without alterations in OXPHOS complexes. In conclusion, overnutrition during early life induces short-term metabolic disturbances, impairment in heart insulin signaling, up-regulates GLUT-1 and switch cardiac fuel preference in juvenile mice.


Asunto(s)
Metabolismo de los Hidratos de Carbono , Transportador de Glucosa de Tipo 1/metabolismo , Insulina/metabolismo , Lactancia , Mitocondrias Cardíacas/metabolismo , Hipernutrición , Transducción de Señal , Regulación hacia Arriba , Animales , Ratones , Oxidación-Reducción
16.
Acta Cir Bras ; 30(7): 452-60, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-26270136

RESUMEN

PURPOSE: To study the effect of remote ischemic preconditioning (RIPC) in ischemia-reperfusion (I/R) liver injury and in the expression of IL-6 and IL-10 in a rat model. METHODS: Thirty-six male rats were divided in three groups: Sham; I/R injury, a 45 minutes lobar liver ischemia and reperfusion; and RIPC, six cycles of four minutes of ischemia and four minutes of reperfusion on the right hindlimb followed by a 45 minutes lobar liver ischemia and reperfusion. Tissue and blood samples were collected after 1h and 3h of reperfusion for histopathological study, plasma cytokines and alanine aminotransferase (ALT) measurement. RESULTS: The histopathological study demonstrated a significant reduction in liver necrosis in the RIPC group (p<0,001). The ALT levels were also significant lower in the RIPC group (p<0.01). The cytokines assessment showed that IL-6 levels were increased in the RIPC group after 1h of reperfusion, in comparison to the I/R group (p<0.05). Interleukin-10 levels in RIPC groups did not differ significantly from I/R group. CONCLUSIONS: Remote ischemic preconditioning is effective in decreasing liver necrosis in a rat model of ischemia-reperfusion. The IL-6 expression is up-regulated and peaked at 60 min of reperfusion. There was no difference in IL-10 expression between the groups.


Asunto(s)
Modelos Animales de Enfermedad , Interleucina-10/sangre , Interleucina-6/sangre , Precondicionamiento Isquémico/métodos , Hígado/irrigación sanguínea , Daño por Reperfusión/sangre , Alanina Transaminasa/sangre , Animales , Ensayo de Inmunoadsorción Enzimática , Hígado/patología , Masculino , Necrosis/patología , Necrosis/prevención & control , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Factores de Tiempo
17.
Acta cir. bras ; 30(7): 452-460, 07/2015. graf
Artículo en Inglés | LILACS | ID: lil-754977

RESUMEN

PURPOSE: To study the effect of remote ischemic preconditioning (RIPC) in ischemia-reperfusion (I/R) liver injury and in the expression of IL-6 and IL-10 in a rat model. METHODS: Thirty-six male rats were divided in three groups: Sham; I/R injury, a 45 minutes lobar liver ischemia and reperfusion; and RIPC, six cycles of four minutes of ischemia and four minutes of reperfusion on the right hindlimb followed by a 45 minutes lobar liver ischemia and reperfusion. Tissue and blood samples were collected after 1h and 3h of reperfusion for histopathological study, plasma cytokines and alanine aminotransferase (ALT) measurement. RESULTS: The histopathological study demonstrated a significant reduction in liver necrosis in the RIPC group (p<0,001). The ALT levels were also significant lower in the RIPC group (p<0.01). The cytokines assessment showed that IL-6 levels were increased in the RIPC group after 1h of reperfusion, in comparison to the I/R group (p<0.05). Interleukin-10 levels in RIPC groups did not differ significantly from I/R group. CONCLUSIONS: Remote ischemic preconditioning is effective in decreasing liver necrosis in a rat model of ischemia-reperfusion. The IL-6 expression is up-regulated and peaked at 60 min of reperfusion. There was no difference in IL-10 expression between the groups. .


Asunto(s)
Animales , Masculino , Modelos Animales de Enfermedad , /sangre , /sangre , Precondicionamiento Isquémico/métodos , Hígado/irrigación sanguínea , Daño por Reperfusión/sangre , Alanina Transaminasa/sangre , Ensayo de Inmunoadsorción Enzimática , Hígado/patología , Necrosis/patología , Necrosis/prevención & control , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Factores de Tiempo
18.
Exp Cell Res ; 336(1): 15-22, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-25978973

RESUMEN

Mitochondrial dysfunction has been associated with liver cholestatis. Toxic bile salt accumulation leads to chronic injury with mitochondrial damage, ROS increase and apoptosis, resulting in liver dysfunction. This study aimed to analyze mitochondrial bioenergetics in rats with hepatic fibrosis induced by bile duct ligation (BDL) after BMMNC transplantation. Livers were collected from normal rats, fibrotic rats after 14 and 21 days of BDL (F14d and F21d) and rats that received BMMNC at 14 days of BDL, analyzed after 7 days. F21d demonstrated increased collagen I content and consequently decrease after BMMNC transplantation. Both F14d and F21d had significantly reduced mitochondrial oxidation capacity and increased mitochondrial uncoupling, which were restored to levels similar to those of normal group after BMMNC transplantation. In addition, F21d had a significantly increase of UCP2, and reduced PGC-1α content. However, after BMMNC transplantation both proteins returned to levels similar to normal group. Moreover, F14d had a significantly increase in 4-HNE content compared to normal group, but after BMMNC transplantation 4-HNE content significantly reduced, suggesting oxidative stress reduction. Therefore, BMMNC transplantation has a positive effect on hepatic mitochondrial bioenergetics of cholestatic rats, increasing oxidative capacity and reducing oxidative stress, which, in turn, contribute to liver function recover.


Asunto(s)
Trasplante de Médula Ósea , Colestasis/prevención & control , Metabolismo Energético , Cirrosis Hepática/prevención & control , Hígado/fisiopatología , Mitocondrias/metabolismo , Estrés Oxidativo , Animales , Western Blotting , Células Cultivadas , Colestasis/metabolismo , Colestasis/patología , Peroxidación de Lípido , Cirrosis Hepática/metabolismo , Cirrosis Hepática/patología , Masculino , Mitocondrias/patología , Oxidación-Reducción , Ratas , Ratas Wistar
19.
Cell Tissue Res ; 357(1): 185-94, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24744266

RESUMEN

Bone marrow cells (BMCs) are the main type of cells used for transplantation therapies. Obesity, a major world health problem, has been demonstrated to affect various tissues, including bone marrow. This could compromise the success of such therapies. One of the main mechanisms underlying the pathogenesis of obesity is mitochondrial dysfunction, and recent data have suggested an important role for mitochondrial metabolism in the regulation of stem cell proliferation and differentiation. Since the potential use of BMCs for clinical therapies depends on their viability and capacity to proliferate and/or differentiate properly, the analysis of mitochondrial function and cell viability could be important approaches for evaluating BMC quality in the context of obesity. We therefore compared BMCs from a control group (CG) and an obese group (OG) of mice and evaluated their mitochondrial function, proliferation capacity, apoptosis, and levels of proteins involved in energy metabolism. BMCs from OG had increased apoptosis and decreased proliferation rates compared with CG. Mitochondrial respiratory capacity, biogenesis, and the coupling between oxidative phosphorylation and ATP synthesis were significantly decreased in OG compared with CG, in correlation with increased levels of uncoupling protein 2 and reduced peroxisome proliferator-activated receptor-coactivator 1α content. OG also had decreased amounts of the glucose transporter GLUT-1 and insulin receptor (IRß). Thus, Western-diet-induced obesity leads to mitochondrial dysfunction and reduced proliferative capacity in BMCs, changes that, in turn, might compromise the success of therapies utilizing these cells.


Asunto(s)
Células de la Médula Ósea/citología , Mitocondrias/fisiología , Obesidad/patología , Animales , Células de la Médula Ósea/metabolismo , Supervivencia Celular/fisiología , Masculino , Ratones , Ratones Obesos , Obesidad/metabolismo , Fosforilación Oxidativa , Transducción de Señal
20.
J Nutr Biochem ; 25(1): 50-7, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24314865

RESUMEN

Nutritional transition has contributed to growing obesity, mainly by changing eating habits of the population. The mechanisms by which diet-induced obesity leads to cardiac injury are not completely understood, but it is known that obesity is associated to impaired cardiac function and energy metabolism, increasing morbidity and mortality. Therefore, our study aimed to investigate the mechanisms underlying cardiac metabolism impairment related to Western diet-induced obesity. After weaning, male Swiss mice were fed a Western diet for 16 weeks in order to induce obesity. After this period, the content of proteins involved in heart energy metabolism GLUT1, cytosolic lysate and plasma membrane GLUT4, AMPK, pAMPK, IRß, IRS-1, PGC-1α, CPT1 and UCP2 was evaluated. Also, the oxidative phosphorylation of myocardial fibers was measured by high-resolution respirometry. Mice in the Western diet group (WG) presented altered biometric parameters compared to those in control group, including higher body weight, increased myocardial lipid deposition and glucose intolerance, which demonstrate the obesogenic role of Western diet. WG presented increased CPT1 and UCP2 contents and decreased IRS-1, plasma membrane GLUT4 and PGC-1α contents. In addition, WG presented cardiac mitochondrial dysfunction and reduced biogenesis, demonstrating a lower capacity of carbohydrates and fatty acid oxidation and also decreased coupling between oxidative phosphorylation and adenosine triphosphate synthesis. Cardiac metabolism impairment related to Western diet-induced obesity is probably due to damaged myocardial oxidative capacity, reduced mitochondrial biogenesis and mitochondria uncoupling, which compromise the bioenergetic metabolism of heart.


Asunto(s)
Dieta/efectos adversos , Metabolismo Energético , Corazón/fisiopatología , Adenosina Trifosfato/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/metabolismo , Prueba de Tolerancia a la Glucosa , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 1/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Proteínas Sustrato del Receptor de Insulina/genética , Proteínas Sustrato del Receptor de Insulina/metabolismo , Canales Iónicos/genética , Canales Iónicos/metabolismo , Metabolismo de los Lípidos , Masculino , Ratones , Ratones Obesos , Mitocondrias Cardíacas/efectos de los fármacos , Mitocondrias Cardíacas/metabolismo , Mitocondrias Cardíacas/patología , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Recambio Mitocondrial , Miocardio , Fosforilación Oxidativa , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína Desacopladora 2 , Aumento de Peso
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...