Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(22)2021 Nov 18.
Artículo en Inglés | MEDLINE | ID: mdl-34830314

RESUMEN

The kidney is one of the most susceptible organs to age-related impairments. Generally, renal aging is accompanied by renal fibrosis, which is the final common pathway of chronic kidney diseases. Aristolochic acid (AA), a nephrotoxic agent, causes AA nephropathy (AAN), which is characterized by progressive renal fibrosis and functional decline. Although renal fibrosis is associated with renal aging, whether AA induces renal aging remains unclear. The aim of the present study is to investigate the potential use of AAN as a model of renal aging. Here, we examined senescence-related factors in AAN models by chronically administering AA to C57BL/6 mice. Compared with controls, the AA group demonstrated aging kidney phenotypes, such as renal atrophy, renal functional decline, and tubulointerstitial fibrosis. Additionally, AA promoted cellular senescence specifically in the kidneys, and increased renal p16 mRNA expression and senescence-associated ß-galactosidase activity. Furthermore, AA-treated mice exhibited proximal tubular mitochondrial abnormalities, as well as reactive oxygen species accumulation. Klotho, an antiaging gene, was also significantly decreased in the kidneys of AA-treated mice. Collectively, the results of the present study indicate that AA alters senescence-related factors, and that renal fibrosis is closely related to renal aging.


Asunto(s)
Envejecimiento/efectos de los fármacos , Ácidos Aristolóquicos/farmacología , Colágeno/genética , Riñón/efectos de los fármacos , Nefritis Intersticial/inducido químicamente , Insuficiencia Renal Crónica/inducido químicamente , Envejecimiento/genética , Animales , Colágeno/agonistas , Colágeno/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Modelos Animales de Enfermedad , Fibrosis , Regulación de la Expresión Génica , Humanos , Riñón/metabolismo , Riñón/patología , Proteínas Klotho/genética , Proteínas Klotho/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/patología , Nefritis Intersticial/genética , Nefritis Intersticial/metabolismo , Nefritis Intersticial/patología , Especies Reactivas de Oxígeno/agonistas , Especies Reactivas de Oxígeno/metabolismo , Insuficiencia Renal Crónica/genética , Insuficiencia Renal Crónica/metabolismo , Insuficiencia Renal Crónica/patología , Transducción de Señal , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , beta-Galactosidasa/genética , beta-Galactosidasa/metabolismo
2.
Molecules ; 26(12)2021 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-34208202

RESUMEN

Chronic UVA exposure results in elevated reactive oxygen species in skin which leads to photoaging characterized as upregulated matrix metalloproteinase (MMP)-1 and loss of collagen. Therefore, natural antioxidants are hailed as promising agents to be utilized against photoaging. In the current study, reynosin and santamarine, two known sesquiterpene lactones isolated from Artemisia scoparia, were analyzed for their anti-photoaging properties in UVA-irradiated human dermal fibroblasts (HDFs). Results showed that UVA irradiation (8 J/cm2) upregulated the MMP-1 secretion and expression, and suppressed collagen production, which were significantly reverted by santamarine treatment (10 µM). Although both reynosin and santamarine exhibited ROS scavenging abilities, reynosin failed to significantly diminish UVA-stimulated MMP-1 release. UVA-irradiated HDFs showed increased collagen production when treated with santamarine. As a mechanism to suppress MMP-1, santamarine significantly suppressed the UVA-induced phosphorylation of p38 and JNK and nuclear translocation of p-c-Fos and p-c-Jun. Santamarine promoted collagen I production via relieving the UVA-induced suppression on TGF-ß and its downstream activator Smad2/3 complex. Antioxidant properties of santamarine were also shown to arise from stimulating Nrf2-dependent expression of antioxidant enzymes SOD-1 and HO-1 in UVA-irradiated HDFs. In conclusion, santamarine was found to be a promising natural antioxidant with anti-photoaging properties against UVA-induced damages in HDFs.


Asunto(s)
Fibroblastos/efectos de los fármacos , Sesquiterpenos/farmacología , Envejecimiento de la Piel/efectos de los fármacos , Proteína Smad4/agonistas , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/agonistas , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Células Cultivadas , Colágeno Tipo I/metabolismo , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Humanos , Sistema de Señalización de MAP Quinasas , Metaloproteinasa 1 de la Matriz/metabolismo , Transducción de Señal , Envejecimiento de la Piel/patología , Envejecimiento de la Piel/efectos de la radiación , Rayos Ultravioleta
3.
PLoS One ; 16(3): e0249047, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33765066

RESUMEN

Mitochondria are commonly viewed as highly elongated organelles with regularly spaced mtDNA genomes organized as compact nucleoids that generate the local transcripts essential for production of mitochondrial ribosomes and key components of the respiratory chain. In contrast, A549 human lung carcinoma cells frequently contain apparently swollen mitochondria harboring multiple discrete mtDNA nucleoids and RNA processing granules in a contiguous matrix compartment. While this seemingly aberrant mitochondrial morphology is akin to "mito-bulbs" previously described in cells exposed to a variety of genomic stressors, it occurs in A549 cells under typical culture conditions. We provide a detailed confocal and super-resolution microscopic investigation of the incidence of such mito-bulbs in A549 cells. Most mito-bulbs appear stable, engage in active replication and transcription, and maintain respiration but feature an elevated oxidative environment. High concentrations of glucose and/or L-glutamine in growth media promote a greater incidence of mito-bulbs. Furthermore, we demonstrate that treatment of A549 cells with TGFß suppresses the formation of mito-bulbs while treatment with a specific TGFß pathway inhibitor substantially increases incidence. This striking heterogeneity of mitochondrial form and function may play an important role in a variety of diseases involving mitochondrial dysfunction.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias/metabolismo , Células A549 , ADN Mitocondrial/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Glucosa/farmacología , Glutamina/farmacología , Humanos , Potencial de la Membrana Mitocondrial , Microscopía Confocal , Mitocondrias/genética , Dinámicas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/metabolismo , ARN/metabolismo , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo
4.
Int J Mol Sci ; 22(3)2021 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-33525359

RESUMEN

Nonalcoholic fatty liver disease is defined as the accumulation of excessive fat in the liver in the absence of excessive alcohol consumption or any secondary cause. Although the disease generally remains asymptomatic, chronic liver inflammation leads to fibrosis, liver cirrhosis, and even to the development of hepatocellular carcinoma (HCC). Fibrosis results from epithelial-mesenchymal transition (EMT), which leads to dedifferentiation of epithelial cells into cells with a mesenchymal-like phenotype. During EMT, epithelial cells with high expression of E-cadherin, influenced by growth factors, cytokines, and inflammatory processes, undergo morphological changes via enhanced expression of, e.g., vimentin, fibronectin, and N-cadherin. An inducer of EMT and, consequently, of fibrosis development is transforming growth factor beta (TGFß), a pleiotropic cytokine associated with the progression of hepatocarcinogenesis. However, the understanding of the molecular events that direct the development of steatosis into steatohepatitis and liver fibrosis remains incomplete. Our study revealed that both prolonged exposure of hepatocarcinoma cells to fatty acids in vitro and high-fat diet in mice (20 weeks) result in inflammation. Prolonged treatment with fatty acids increased the levels of TGFß, MMP9, and ß-catenin, important EMT inducers. Moreover, the livers of mice fed a high-fat diet exhibited features of liver fibrosis with increased TGFß and IL-1 levels. Increased expression of IL-1 correlated with a decrease in monocyte chemoattractant protein-induced protein 1 (MCPIP1), a negative regulator of the inflammatory response that regulates the stability of proinflammatory transcripts encoding IL-1. Our study showed that a high-fat diet induced EMT by increasing the levels of EMT-activating transcription factors, including Zeb1, Zeb2, and Snail and changed the protein profile to a profile characteristic of the mesenchymal phenotype.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Hepatocitos/efectos de los fármacos , Enfermedad del Hígado Graso no Alcohólico/genética , Factor de Crecimiento Transformador beta/genética , beta Catenina/genética , Animales , Cadherinas/genética , Cadherinas/metabolismo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal/genética , Fibronectinas/genética , Fibronectinas/metabolismo , Regulación de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Interleucina-1/genética , Interleucina-1/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Metaloproteinasa 9 de la Matriz/genética , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Enfermedad del Hígado Graso no Alcohólico/patología , Ácido Oléico/farmacología , Ribonucleasas/genética , Ribonucleasas/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/metabolismo , Vimentina/genética , Vimentina/metabolismo , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/genética , Caja Homeótica 2 de Unión a E-Box con Dedos de Zinc/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , beta Catenina/agonistas , beta Catenina/metabolismo
5.
Virus Res ; 245: 29-43, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29273341

RESUMEN

Hepatitis B virus (HBV) infection is a leading cause of liver damage and hepatic inflammation. Upon infection, effective antiviral responses by CD8+ T cells, CD4+ T cells, Natural killer (NK) cells, and monocytes can lead to partial or complete eradication of the viral infection. To date, many studies have shown that the production of inhibitory cytokines such as Interleukin 10 (IL-10), Transforming growth factor beta (TGF-ß), along with dysfunction of the dendritic cells (DCs), and the absence of efficient innate immune responses could lead to T cell exhaustion, development of persistent infection, and inability to eradicate the viral infection from liver. Understanding the immunopathogenesis of the virus could be useful in providing further insights toward novel strategies in the eradication of HBV infection.


Asunto(s)
Anergia Clonal/efectos de los fármacos , Vacunas contra Hepatitis B/administración & dosificación , Virus de la Hepatitis B/inmunología , Hepatitis B Crónica/inmunología , Inmunidad Innata/efectos de los fármacos , Antivirales/uso terapéutico , Células Dendríticas/inmunología , Células Dendríticas/virología , Regulación de la Expresión Génica , Vacunas contra Hepatitis B/biosíntesis , Vacunas contra Hepatitis B/síntesis química , Virus de la Hepatitis B/efectos de los fármacos , Virus de la Hepatitis B/patogenicidad , Hepatitis B Crónica/tratamiento farmacológico , Hepatitis B Crónica/patología , Hepatitis B Crónica/prevención & control , Humanos , Interleucina-10/agonistas , Interleucina-10/genética , Interleucina-10/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/virología , Hígado/inmunología , Hígado/virología , Vacunación Masiva/métodos , Monocitos/inmunología , Monocitos/virología , Linfocitos T/inmunología , Linfocitos T/virología , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/inmunología , Carga Viral/efectos de los fármacos , Carga Viral/inmunología
6.
Ann Rheum Dis ; 76(11): 1931-1940, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28801346

RESUMEN

OBJECTIVE: To evaluate the antifibrotic effects of the pan-peroxisome proliferator-activated receptor (PPAR) agonist IVA337 in preclinical mouse models of pulmonary fibrosis and related pulmonary hypertension (PH). METHODS: IVA337 has been evaluated in the mouse model of bleomycin-induced pulmonary fibrosis and in Fra-2 transgenic mice, this latter being characterised by non-specific interstitial pneumonia and severe vascular remodelling of pulmonary arteries leading to PH. Mice received two doses of IVA337 (30 mg/kg or 100 mg/kg) or vehicle administered by daily oral gavage up to 4 weeks. RESULTS: IVA337 demonstrated at a dose of 100 mg/kg a marked protection from the development of lung fibrosis in both mouse models compared with mice receiving 30 mg/kg of IVA337 or vehicle. Histological score was markedly reduced by 61% in the bleomycin model and by 50% in Fra-2 transgenic mice, and total lung hydroxyproline concentrations decreased by 28% and 48%, respectively, as compared with vehicle-treated mice. IVA337 at 100 mg/kg also significantly decreased levels of fibrogenic markers in lesional lungs of both mouse models. In addition, IVA337 substantially alleviated PH in Fra-2 transgenic mice by improving haemodynamic measurements and vascular remodelling. In primary human lung fibroblasts, IVA337 inhibited in a dose-dependent manner fibroblast to myofibroblasts transition induced by TGF-ß and fibroblast proliferation mediated by PDGF. CONCLUSION: We demonstrate that treatment with 100 mg/kg IVA337 prevents lung fibrosis in two complementary animal models and substantially attenuates PH in the Fra-2 mouse model. These findings confirm that the pan-PPAR agonist IVA337 is an appealing therapeutic candidate for these cardiopulmonary involvements.


Asunto(s)
Benzotiazoles/farmacología , Hipertensión Pulmonar/tratamiento farmacológico , Fibrosis Pulmonar/tratamiento farmacológico , Sulfonamidas/farmacología , Animales , Bleomicina , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibroblastos/efectos de los fármacos , Antígeno 2 Relacionado con Fos , Hipertensión Pulmonar/etiología , Ratones , Ratones Transgénicos , Miofibroblastos/efectos de los fármacos , Arteria Pulmonar/efectos de los fármacos , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/complicaciones , Índice de Severidad de la Enfermedad , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/fisiología , Resultado del Tratamiento , Remodelación Vascular/efectos de los fármacos
7.
Biochem J ; 474(3): 399-410, 2017 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-27803247

RESUMEN

Cardiac fibrosis and chronic inflammation are common complications in type 2 diabetes mellitus (T2D). Since nucleotide oligomerization-binding domain 1 (NOD1), an innate immune receptor, is involved in the pathogenesis of insulin resistance and diabetes outcomes, we sought to investigate its involvement in cardiac fibrosis. Here, we show that selective staining of cardiac fibroblasts from T2D (db/db;db) mice exhibits up-regulation and activation of the NOD1 pathway, resulting in enhanced NF-κB and TGF-ß signalling. Activation of the TGF-ß pathway in cardiac fibroblasts from db mice was prevented after inhibition of NF-κB with BAY-11-7082 (BAY). Moreover, fibrosis progression in db mice was also prevented by BAY treatment. Enhanced TGF-ß signalling and cardiac fibrosis of db mice was dependent, at least in part, on the sequential activation of NOD1 and NF-κB since treatment of db mice with a selective NOD1 agonist induced activation of the TGF-ß pathway, but co-administration of a NOD1 agonist plus BAY, or a NOD1 inhibitor prevented the NOD1-induced fibrosis. Therefore, NOD1 is involved in cardiac fibrosis associated with diabetes, and establishes a new mechanism for the development of heart fibrosis linked to T2D.


Asunto(s)
Diabetes Mellitus Experimental/metabolismo , Fibrosis Endomiocárdica/metabolismo , Miocardio/metabolismo , FN-kappa B/metabolismo , Proteína Adaptadora de Señalización NOD1/metabolismo , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Diabetes Mellitus Tipo 2/patología , Ácido Diaminopimélico/análogos & derivados , Ácido Diaminopimélico/farmacología , Fibrosis Endomiocárdica/genética , Fibrosis Endomiocárdica/patología , Fibrosis Endomiocárdica/prevención & control , Regulación de la Expresión Génica , Humanos , Insulina/sangre , Resistencia a la Insulina , Ratones , Ratones Transgénicos , Miocardio/patología , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Células 3T3 NIH , Nitrilos/farmacología , Proteína Adaptadora de Señalización NOD1/agonistas , Proteína Adaptadora de Señalización NOD1/genética , Transducción de Señal , Sulfonas/farmacología , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
8.
Artículo en Inglés | MEDLINE | ID: mdl-27413100

RESUMEN

The discovery of the transforming growth factor ß (TGF-ß) family ligands and the realization that their bioactivities need to be tightly controlled temporally and spatially led to intensive research that has identified a multitude of extracellular modulators of TGF-ß family ligands, uncovered their functions in developmental and pathophysiological processes, defined the mechanisms of their activities, and explored potential modulator-based therapeutic applications in treating human diseases. These studies revealed a diverse repertoire of extracellular and membrane-associated molecules that are capable of modulating TGF-ß family signals via control of ligand availability, processing, ligand-receptor interaction, and receptor activation. These molecules include not only soluble ligand-binding proteins that were conventionally considered as agonists and antagonists of TGF-ß family of growth factors, but also extracellular matrix (ECM) proteins and proteoglycans that can serve as "sink" and control storage and release of both the TGF-ß family ligands and their regulators. This extensive network of soluble and ECM modulators helps to ensure dynamic and cell-specific control of TGF-ß family signals. This article reviews our knowledge of extracellular modulation of TGF-ß growth factors by diverse proteins and their molecular mechanisms to regulate TGF-ß family signaling.


Asunto(s)
Factor de Crecimiento Transformador beta/efectos de los fármacos , Animales , Proteínas de la Matriz Extracelular/metabolismo , Humanos , Ligandos , Proteoglicanos/metabolismo , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo
9.
Part Fibre Toxicol ; 13(1): 27, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27251132

RESUMEN

BACKGROUND: Multi-walled carbon nanotubes (MWCNT) are currently under intense toxicological investigation due to concern on their potential health effects. Current in vitro and in vivo data indicate that MWCNT exposure is strongly associated with lung toxicity (inflammation, fibrosis, granuloma, cancer and airway injury) and their effects might be comparable to asbestos-induced carcinogenesis. Although fibrosis is a multi-origin disease, epithelial-mesenchymal transition (EMT) is recently recognized as an important pathway in cell transformation. It is known that MWCNT exposure induces EMT through the activation of the TGF-ß/Smad signalling pathway thus promoting pulmonary fibrosis, but the molecular mechanisms involved are not fully understood. In the present work we propose a new mechanism involving a TGF-ß-mediated signalling pathway. METHODS: Human bronchial epithelial cells were incubated with two different MWCNT samples at various concentrations for up to 96 h and several markers of EMT were investigated. Quantitative real time PCR, western blot, immunofluorescent staining and gelatin zymographies were performed to detect the marker protein alterations. ELISA was performed to evaluate TGF-ß production. Experiments with neutralizing anti-TGF-ß antibody, specific inhibitors of GSK-3ß and Akt and siRNA were carried out in order to confirm their involvement in MWCNT-induced EMT. In vivo experiments of pharyngeal aspiration in C57BL/6 mice were also performed. Data were analyzed by a one-way ANOVA with Tukey's post-hoc test. RESULTS: Fully characterized MWCNT (mean length < 5 µm) are able to induce EMT in an in vitro human model (BEAS-2B cells) after long-term incubation at sub-cytotoxic concentrations. MWCNT stimulate TGF-ß secretion, Akt activation and GSK-3ß inhibition, which induces nuclear accumulation of SNAIL-1 and its transcriptional activity, thus contributing to switch on the EMT program. Moreover, a significant increment of nuclear ß-catenin - due to E-cadherin repression and following translocation to nucleus - likely reinforces signalling for EMT promotion. In vivo results supported the occurrence of pulmonary fibrosis following MWCNT exposure. CONCLUSIONS: We demonstrate a new molecular mechanism of MWCNT-mediated EMT, which is Smad-independent and involves TGF-ß and its intracellular effectors Akt/GSK-3ß that activate the SNAIL-1 signalling pathway. This finding suggests potential novel targets in the development of therapeutic and preventive approaches.


Asunto(s)
Bronquios/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Nanotubos de Carbono/toxicidad , Mucosa Respiratoria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Factor de Crecimiento Transformador beta/agonistas , Animales , Bronquios/metabolismo , Bronquios/patología , Bronquios/ultraestructura , Pruebas de Carcinogenicidad , Línea Celular , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Humanos , Exposición por Inhalación/efectos adversos , Masculino , Ratones Endogámicos C57BL , Microscopía Electrónica de Rastreo , Microscopía Electrónica de Transmisión , Nanotubos de Carbono/química , Nanotubos de Carbono/ultraestructura , Tamaño de la Partícula , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fibrosis Pulmonar/inducido químicamente , Fibrosis Pulmonar/metabolismo , Fibrosis Pulmonar/patología , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Mucosa Respiratoria/ultraestructura , Factores de Transcripción de la Familia Snail/metabolismo , Propiedades de Superficie , Factor de Crecimiento Transformador beta/metabolismo
10.
J Physiol Biochem ; 72(3): 393-404, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27121159

RESUMEN

Arteriogenesis is a main defense mechanism to prevent heart and local tissues dysfunction in occlusive artery disease. TGF-ß and angiostatin have a pivotal role in arteriogenesis. We tested the hypothesis that aerobic training and l-arginine supplementation promotes cardiac and skeletal muscles arteriogenesis after myocardial infarction (MI) parallel to upregulation of TGF-ß and downregulation of angiostatin. For this purpose, 4 weeks after LAD occlusion, 50 male Wistar rats were randomly distributed into five groups: (1) sham surgery without MI (sham, n = 10), (2) control-MI (Con-MI, n = 10), (3) l-arginine-MI (La-MI, n = 10), (4) exercise training-MI (Ex-MI, n = 10), and (5) exercise and l-arginine-MI (Ex + La-MI). Exercise training groups running on a treadmill for 10 weeks with moderate intensity. Rats in the l-arginine-treated groups drank water containing 4 % l-arginine. Arteriolar density with different diameters (11-25, 26-50, 51-75, and 76-150 µm), TGF-ß, and angiostatin gene expression were measured in cardiac (area at risk) and skeletal (soleus and gastrocnemius) muscles. Smaller arterioles decreased in cardiac after MI. Aerobic training and l-arginine increased the number of cardiac arterioles with 11-25 and 26-50 µm diameters parallel to TGF-ß overexpression. In gastrocnemius muscle, the number of arterioles/mm(2) was only increased in the 11 to 25 µm in response to training with and without l-arginine parallel to angiostatin downregulation. Soleus arteriolar density with different size was not different between experimental groups. Results showed that 10 weeks aerobic exercise training and l-arginine supplementation promotes arteriogenesis of heart and gastrocnemius muscles parallel to overexpression of TGF-ß and downregulation of angiostatin in MI rats.


Asunto(s)
Arginina/uso terapéutico , Vasos Coronarios/fisiopatología , Suplementos Dietéticos , Músculo Esquelético/irrigación sanguínea , Infarto del Miocardio/rehabilitación , Neovascularización Fisiológica , Condicionamiento Físico Animal , Inductores de la Angiogénesis/uso terapéutico , Angiostatinas/antagonistas & inhibidores , Angiostatinas/genética , Angiostatinas/metabolismo , Animales , Arteriolas/fisiopatología , Arterioloesclerosis/dietoterapia , Arterioloesclerosis/fisiopatología , Arterioloesclerosis/terapia , Terapia Combinada , Regulación de la Expresión Génica , Corazón/fisiopatología , Miembro Posterior , Masculino , Actividad Motora , Músculo Esquelético/metabolismo , Músculo Esquelético/fisiopatología , Infarto del Miocardio/etiología , Infarto del Miocardio/metabolismo , Infarto del Miocardio/fisiopatología , Miocardio/metabolismo , Distribución Aleatoria , Ratas Wistar , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
11.
Sci Rep ; 5: 9718, 2015 Apr 13.
Artículo en Inglés | MEDLINE | ID: mdl-25864432

RESUMEN

It is desirable to obtain unlimited supplies of endothelial cells for research and therapeutics. However, current methods of deriving endothelial cells from humans suffer from issues, such as limited supplies, contamination from animal substances, and lengthy/complicated procedures. In this article we developed a way to differentiate human iPS and ES cells to highly pure endothelial cells in 5 days. The chemically defined system is robust, easy to perform, and free of animal substances. Using the system, we verified that combined TGFß and canonical Wnt agonists are essential and sufficient for iPS/ES cell-to-mesoderm transition. Besides, VEGF-KDR signaling alone is required for endothelial formation at high density while supplementation with FGF allows for colonial endothelial differentiation. Finally, anti-adsorptive agents could enrich the endothelial output by allowing selective attachment of the endothelial precursors. The system was validated to work on multiple iPS/ES cells lines to produce endothelial cells capable of forming capillary-like structures in vitro and integrating into host vasculature in vivo. In sum, the simple yet robust differentiation system permits the unlimited supply of human endothelial cells. The defined and animal substance-free nature of the system is compatible with clinical applications and characterization of endothelial differentiation in an unbiased manner.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Células Endoteliales/citología , Células Endoteliales/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Animales , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Madre Embrionarias/efectos de los fármacos , Células Endoteliales/efectos de los fármacos , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Glucógeno Sintasa Quinasas/antagonistas & inhibidores , Humanos , Células Madre Pluripotentes Inducidas/efectos de los fármacos , Mesodermo/embriología , Mesodermo/metabolismo , Ratones , Factor de Crecimiento Transformador beta/agonistas , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Factores de Crecimiento Endotelial Vascular/metabolismo , Proteínas Wnt/agonistas
12.
Mol Vis ; 21: 138-47, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25684979

RESUMEN

PURPOSE: Previously, we demonstrated that scleral stem/progenitor cells (SSPCs) from mice have a chondrogenic differentiation potential, which is stimulated by transforming growth factor-ß (TGF-ß). In the present study, we hypothesized that chondrogenesis in the sclera could be a possible mechanism in myopia development. Therefore, we investigated the association of form-deprivation myopia (FDM) with expressions in mice sclera representing the chondrogenic phenotype: collagen type II (Col2) and α-smooth muscle actin (α-SMA). METHODS: The mRNA levels of α-SMA and Col2 in cultured murine SSPCs during chondrogenesis stimulated by TGF-ß2 were determined by real-time quantitative RT-PCR (qRT-PCR). The expression patterns of α-SMA and Col2 were assessed by immunohistochemistry in a three dimensional pellet culture. In an FDM mouse model, a western blot analysis and immunofluorescence study were used to detect the changes in the α-SMA and Col2 protein expressions in the sclera. In the RPE-choroid complex, qRT-PCR was used to detect any changes in the TGF-ß mRNA expression. RESULTS: The treatment of SSPCs in vitro with TGF-ß2 for 24 h at 1 or 10 ng/ml led to increased levels of both the α-SMA and Col2 expressions. In addition, we observed the formation of cartilage-like pellets from TGF-ß2-treated SSPCs. Both α-SMA and Col2 were expressed in the pellet. In an in-vivo study, the α-SMA and Col2 protein expressions were significantly increased in the sclera of FDM eyes in comparison to contralateral control eyes. Similarly, the levels of TGF-ß in the RPE-choroid complex of an FDM eye were also significantly elevated. CONCLUSION: Based on the concept of stem cells possessing multipotent differentiation potentials, scleral chondrogenesis induced by SSPCs may play a role in myopia development. The increased expressions of the cartilage-associated proteins Col2 and α-SMA during scleral chondrogenesis may be potential markers for myopia development. In addition, the increased levels of TGF-ß mRNA in the RPE-choroid complex might induce the chondrogenic change in the sclera during myopia development.


Asunto(s)
Condrogénesis/genética , Coroides/patología , Miopía/patología , Epitelio Pigmentado de la Retina/patología , Esclerótica/patología , Células Madre/patología , Actinas/agonistas , Actinas/genética , Actinas/metabolismo , Animales , Células Cultivadas , Coroides/metabolismo , Colágeno Tipo II/agonistas , Colágeno Tipo II/genética , Colágeno Tipo II/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Masculino , Ratones , Ratones Endogámicos C57BL , Miopía/genética , Miopía/metabolismo , ARN Mensajero/agonistas , ARN Mensajero/genética , ARN Mensajero/metabolismo , Epitelio Pigmentado de la Retina/metabolismo , Esclerótica/efectos de los fármacos , Esclerótica/metabolismo , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta2/farmacología
13.
Biochem Biophys Res Commun ; 422(3): 488-93, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22583901

RESUMEN

Transforming growth factor-ß (TGF-ß) is a critical regulator of bone development and remodeling. TGF-ß must be activated from its latent form in order to signal. Thrombospondin-1 (TSP1) is a major regulator of latent TGF-ß activation and TSP1 control of TGF-ß activation is critical for regulation of TGF-ß activity in multiple diseases. Bone marrow-derived mesenchymal stem cells (MSCs) have osteogenic potential and they participate in bone remodeling in injury and in response to tumor metastasis. Since both TSP1 and TGF-ß inhibit osteoblast differentiation, we asked whether TSP1 blocks osteoblast differentiation of MSCs through its ability to stimulate TGF-ß activation. TSP1 added to human bone marrow-derived MSCs under growth conditions increases active TGF-ß. Cultured MSCs express TSP1 and both TSP1 expression and TGF-ß activity decrease during osteoblast differentiation. TSP1 and active TGF-ß block osteoblast differentiation of MSCs grown in osteogenic media as measured by decreased Runx2 and alkaline phosphatase expression. The inhibitory effect of TSP1 on osteoblast differentiation is due to its ability to activate latent TGF-ß, since a peptide which blocks TSP1 TGF-ß activation reduced TGF-ß activity and restored osteoblast differentiation as measured by increased Runx2 and alkaline phosphatase expression. Anti-TGF-ß neutralizing antibody also increased alkaline phosphatase expression in the presence of TSP1. These studies show that TSP1 regulated TGF-ß activity is a critical determinant of osteoblast differentiation.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Mesenquimatosas/citología , Osteogénesis/fisiología , Trombospondina 1/fisiología , Factor de Crecimiento Transformador beta/agonistas , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Medios de Cultivo Condicionados/química , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Osteogénesis/efectos de los fármacos , Péptidos/farmacología , Trombospondina 1/análisis , Trombospondina 1/farmacología , Factor de Crecimiento Transformador beta/análisis , Factor de Crecimiento Transformador beta/metabolismo
14.
PLoS One ; 6(10): e26280, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22043313

RESUMEN

Erythropoietin (EPO) is a pleiotropic cytokine originally identified for its role in erythropoiesis. In addition, in various preclinical models EPO exhibited protective activity against tissue injury. There is an urgent need for potent treatments of autoimmune driven disorders of the peripheral nervous system (PNS), such as the Guillain-Barré syndrome (GBS), a disabling autoimmune disease associated with relevant morbidity and mortality. To test the therapeutic potential of EPO in experimental autoimmune neuritis (EAN) - an animal model of human GBS--immunological and clinical effects were investigated in a preventive and a therapeutic paradigm. Treatment with EPO reduced clinical disease severity and if given therapeutically also shortened the recovery phase of EAN. Clinical findings were mirrored by decreased inflammation within the peripheral nerve, and myelin was well maintained in treated animals. In contrast, EPO increased the number of macrophages especially in later stages of the experimental disease phase. Furthermore, the anti-inflammatory cytokine transforming growth factor (TGF)-beta was upregulated in the treated cohorts. In vitro experiments revealed less proliferation of T cells in the presence of EPO and TGF-beta was moderately induced, while the secretion of other cytokines was almost not altered by EPO. Our data suggest that EPO revealed its beneficial properties by the induction of beneficial macrophages and the modulation of the immune system towards anti-inflammatory responses in the PNS. Further studies are warranted to elaborate the clinical usefulness of EPO for treating immune-mediated neuropathies in affected patients.


Asunto(s)
Eritropoyetina/farmacología , Macrófagos/metabolismo , Neuritis Autoinmune Experimental/tratamiento farmacológico , Factor de Crecimiento Transformador beta/efectos de los fármacos , Animales , Citocinas , Eritropoyetina/uso terapéutico , Inflamación , Ratas , Activación Transcripcional/efectos de los fármacos , Factor de Crecimiento Transformador beta/agonistas , Resultado del Tratamiento
15.
J Neurochem ; 115(2): 450-9, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20681951

RESUMEN

Nucleotides act as early signals for microglial recruitment to sites of CNS injury. As microglial motility and activation can be influenced by several local factors at the site of the lesion, we investigated the effects of interferon-gamma, lipopolysaccharide (LPS) or transforming growth factor-ß (TGF-ß) addition to mixed glial cell cultures, on microglial migration in response to ADP, P2Y12 and P2Y1 mRNA expression as well as on the expression of an array of genes associated with the process of microglial activation. First, we demonstrated, by pharmacological inhibition and by using small interfering RNAs, that in addition to P2Y12, P2Y1 is involved in ADP-stimulated microglial migration. The ability of specific agonists to induce Ca(2+) mobilization further confirmed the expression of functional P2Y receptors in microglia. Then, we found that migratory capability and expression of both P2Y receptors were abrogated in microglial cells from LPS-stimulated mixed glial cultures, while TGF-ß increased ADP-induced migration and the expression of P2Y12 and P2Y1 receptors. Interferon-gamma did not influence receptor expression or microglial migration. Finally, the patterns of gene expression induced in microglia by LPS or TGF-ß treatment of mixed glial cultures were clearly distinct. LPS induced a set of classical pro-inflammatory genes, whereas TGF-ß increased the expression of genes associated with atypical microglial phenotype, namely arginase-1 and TGF-ß genes. These results imply that both P2Y1 and P2Y12 may guide microglia toward the lesion. They also suggest that the modulation of microglial purinergic receptors expression by local factors, through direct and/or astrocyte-mediated actions, may represent a novel mechanism affecting neuroinflammatory response.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Lipopolisacáridos/farmacología , Microglía/efectos de los fármacos , Receptores Purinérgicos P2Y1/metabolismo , Receptores Purinérgicos P2/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Adenosina Difosfato/análogos & derivados , Adenosina Difosfato/farmacología , Animales , Animales Recién Nacidos , Calcio/metabolismo , Células Cultivadas , Corteza Cerebral/citología , Regulación de la Expresión Génica/efectos de los fármacos , Interferón gamma/farmacología , Microglía/metabolismo , ARN Interferente Pequeño/farmacología , Ratas , Receptores Purinérgicos P2/genética , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y12 , Tionucleótidos/farmacología , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores
16.
Front Biosci ; 13: 4726-39, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18508541

RESUMEN

Bone morphogenetic protein-7 (BMP7) is a member of the BMP-subfamily of perhaps a dozen proteins within the TGFbeta-superfamily of cysteine-knot fold cytokine-growth factors. BMP7 has pivotal functions during renal and eye development. In adult organisms, BMP7 is heavily expressed in kidney, specifically in podocytes, distal tubules and collecting ducts. The activity of BMP7 is reduced by inhibitors including some members of the dan-cerberus group and CTGF but can be enhanced by endoglin and KCP. Renal BMP7 disappears early in fibrogenic renal diseases which may facilitate progression. Exogenous administration of rhBMP7 or transgenic overexpression reduces renal fibrogenesis and apoptosis as well as transdifferentiation of epithelial cells. BMP7 improves maintenance of nephron function and structural integrity. These antifibrogenic activities result from inhibition of the nuclear translocation of TGFbeta-activated smad3 by smad6 downstream of BMP7-activated smad5. Although at present the beneficial effects of BMP7 have only been studied in rodent models of chronic renal diseases, there is promise for therapeutic utility of rhBMP7 or small molecule BMP7 agonists in patients.


Asunto(s)
Receptores de Proteínas Morfogenéticas Óseas/fisiología , Proteínas Morfogenéticas Óseas/genética , Nefropatías Diabéticas/genética , Regulación de la Expresión Génica , Enfermedades Renales/genética , Factor de Crecimiento Transformador beta/genética , Proteína Morfogenética Ósea 3 , Proteína Morfogenética Ósea 7 , Receptores de Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Morfogenéticas Óseas/agonistas , Proteínas Morfogenéticas Óseas/efectos de los fármacos , Proteínas Morfogenéticas Óseas/fisiología , Enfermedad Crónica , Fibrinógeno/antagonistas & inhibidores , Fibrinógeno/biosíntesis , Humanos , Inhibinas/fisiología , Riñón/fisiología , Riñón/fisiopatología , Insuficiencia Renal/genética , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/efectos de los fármacos
17.
Trends Biotechnol ; 26(4): 173-80, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18295916

RESUMEN

Fibrotic scars deposited during skin wound healing can cause disfiguration and loss of dermal function. Scar differentiation involves inputs from multiple cell types in a predictable and overlapping sequence of cellular events that includes inflammation, migration/proliferation and extracellular matrix deposition. Research into the molecular mechanisms underpinning these processes in embryonic and adult wounds has contributed to the development of a growing number of novel therapeutic approaches for improving scar appearance. This review discusses some of these emerging strategies for shifting the balance of healing from scarring to regeneration in the context of non-pathological wounds. Particular focus is given to potential therapies based on transforming growth factor (TGF)-beta signaling and recent unexpected findings involving targeting of gap junctional connexins. Lessons learned in promoting scarless healing of cutaneous injuries might provide a basis for regenerative healing in other scenarios, such as spinal cord rupture or myocardial infarction.


Asunto(s)
Cicatriz/fisiopatología , Cicatriz/terapia , Piel/patología , Cicatrización de Heridas/fisiología , Adulto , Cicatriz/patología , Colágeno/inmunología , Colágeno/metabolismo , Conexinas/genética , Conexinas/metabolismo , Conexinas/uso terapéutico , Embrión de Mamíferos/inmunología , Embrión de Mamíferos/fisiopatología , Matriz Extracelular/inmunología , Matriz Extracelular/patología , Feto/inmunología , Feto/fisiopatología , Humanos , Inflamación/complicaciones , Inflamación/embriología , Inflamación/inmunología , Inflamación/fisiopatología , Ingeniería de Proteínas , Piel/metabolismo , Piel/fisiopatología , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Factor de Crecimiento Transformador beta/uso terapéutico , Cicatrización de Heridas/efectos de los fármacos
18.
Mol Cell Neurosci ; 37(2): 247-60, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17997109

RESUMEN

Each type of neuron develops a unique morphology critical to its function, but almost all start with the basic plan of one long axon and multiple short, branched dendrites. Though extrinsic signals are known to direct many steps in the development of neuronal structure, little is understood about the initiation of processes, particularly dendrites. We find that Xenopus retinal ganglion cells (RGCs) explanted early will extend axons and not dendrites in dissociated cultures. If RGCs develop longer in vivo prior to culturing, many now extend dendrite-like processes in vitro, suggesting that an extrinsic factor is required to stimulate dendrite initiation. Members of the transforming growth factor beta (TGFbeta) superfamily, bone morphogenetic protein 2 (BMP2), and growth and differentiation factor 11 (GDF11), can signal cultured RGCs to form dendrites. Furthermore, TGFbeta ligands have an endogenous role: blocking BMP/GDF signaling with a secreted antagonist or inhibitory receptors reduces the number of primary dendrites extended in vivo.


Asunto(s)
Diferenciación Celular/fisiología , Dendritas/ultraestructura , Retina/embriología , Retina/metabolismo , Células Ganglionares de la Retina/metabolismo , Factor de Crecimiento Transformador beta/agonistas , Animales , Proteína Morfogenética Ósea 2 , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Dendritas/efectos de los fármacos , Femenino , Factores de Diferenciación de Crecimiento , Oocitos , Retina/citología , Células Ganglionares de la Retina/citología , Células Ganglionares de la Retina/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/metabolismo , Xenopus laevis
19.
Science ; 318(5848): 271-4, 2007 Oct 12.
Artículo en Inglés | MEDLINE | ID: mdl-17761850

RESUMEN

MicroRNAs (miRNAs) repress hundreds of target messenger RNAs (mRNAs), but the physiological roles of specific miRNA-mRNA interactions remain largely elusive. We report that zebrafish microRNA-430 (miR-430) dampens and balances the expression of the transforming growth factor-beta (TGF-beta) Nodal agonist squint and the TGF-beta Nodal antagonist lefty. To disrupt the interaction of specific miRNA-mRNA pairs, we developed target protector morpholinos complementary to miRNA binding sites in target mRNAs. Protection of squint or lefty mRNAs from miR-430 resulted in enhanced or reduced Nodal signaling, respectively. Simultaneous protection of squint and lefty or absence of miR-430 caused an imbalance and reduction in Nodal signaling. These findings establish an approach to analyze the in vivo roles of specific miRNA-mRNA pairs and reveal a requirement for miRNAs in dampening and balancing agonist/antagonist pairs.


Asunto(s)
MicroARNs/metabolismo , ARN Mensajero/metabolismo , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas de Pez Cebra/genética , Pez Cebra/genética , Regiones no Traducidas 3' , Animales , Embrión no Mamífero/fisiología , Desarrollo Embrionario , Regulación de la Expresión Génica , Factores de Determinación Derecha-Izquierda , Mutación , Proteína Nodal , Ligandos de Señalización Nodal , ARN Mensajero/genética , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Pez Cebra/embriología , Pez Cebra/metabolismo
20.
J Biol Chem ; 282(17): 12475-83, 2007 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-17327235

RESUMEN

Keratocytes of the corneal stroma produce transparent extracellular matrix devoid of hyaluronan (HA); however, in corneal pathologies and wounds, HA is abundant. We previously showed primary keratocytes cultured under serum-free conditions to secrete matrix similar to that of normal stroma, but serum and transforming growth factor beta (TGFbeta) induced secretion of fibrotic matrix components, including HA. This study found HA secretion by primary bovine keratocytes to increase rapidly in response to TGFbeta, reaching a maximum in 12 h and then decreasing to <5% of the maximum by 48 h. Cell-free biosynthesis of HA by cell extracts also exhibited a transient peak at 12 h after TGFbeta treatment. mRNA for hyaluronan synthase enzymes HAS1 and HAS2 increased >10- and >50-fold, respectively, in 4-6 h, decreasing to near original levels after 24-48 h. Small interfering RNA against HAS2 inhibited the transient increase of HAS2 mRNA and completely blocked HA induction, but small interfering RNA to HAS1 had no effect on HA secretion. HAS2 mRNA was induced by a variety of mitogens, and TGFbeta acted synergistically to induce HAS2 by as much as 150-fold. In addition to HA synthesis, treatment with TGFbeta induced degradation of fluorescein-HA added to culture medium. These results show HA secretion by keratocytes to be initiated by a rapid transient increase in the HAS2 mRNA pool. The very rapid induction of HA expression in keratocytes suggests a functional role of this molecule in the fibrotic response of keratocytes to wound healing.


Asunto(s)
Córnea/enzimología , Córnea/metabolismo , Glucuronosiltransferasa/biosíntesis , Ácido Hialurónico/metabolismo , ARN Mensajero/biosíntesis , Factor de Crecimiento Transformador beta/farmacología , Cicatrización de Heridas/fisiología , Animales , Bovinos , Células Cultivadas , Córnea/citología , Lesiones de la Cornea , Sinergismo Farmacológico , Fibrosis/enzimología , Hialuronano Sintasas , Mitógenos/agonistas , Mitógenos/farmacología , Factores de Tiempo , Factor de Crecimiento Transformador beta/agonistas , Factor de Crecimiento Transformador beta/metabolismo , Cicatrización de Heridas/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...