Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
1.
Nat Commun ; 12(1): 4462, 2021 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-34294718

RESUMEN

RORγt+ lymphocytes, including interleukin 17 (IL-17)-producing gamma delta T (γδT17) cells, T helper 17 (Th17) cells, and group 3 innate lymphoid cells (ILC3s), are important immune regulators. Compared to Th17 cells and ILC3s, γδT17 cell metabolism and its role in tissue homeostasis remains poorly understood. Here, we report that the tissue milieu shapes splenic and intestinal γδT17 cell gene signatures. Conditional deletion of mitochondrial transcription factor A (Tfam) in RORγt+ lymphocytes significantly affects systemic γδT17 cell maintenance and reduces ILC3s without affecting Th17 cells in the gut. In vivo deletion of Tfam in RORγt+ lymphocytes, especially in γδT17 cells, results in small intestine tissue remodeling and increases small intestine length by enhancing the type 2 immune responses in mice. Moreover, these mice show dysregulation of the small intestine transcriptome and metabolism with less body weight but enhanced anti-helminth immunity. IL-22, a cytokine produced by RORγt+ lymphocytes inhibits IL-13-induced tuft cell differentiation in vitro, and suppresses the tuft cell-type 2 immune circuit and small intestine lengthening in vivo, highlighting its key role in gut tissue remodeling.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Intestino Delgado/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Animales , Diferenciación Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Femenino , Perfilación de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Homeostasis/inmunología , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Intestino Delgado/citología , Intestino Delgado/metabolismo , Masculino , Metaboloma , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Organoides , Proteína de la Leucemia Promielocítica con Dedos de Zinc/genética , Proteína de la Leucemia Promielocítica con Dedos de Zinc/metabolismo , Subgrupos de Linfocitos T/citología , Células Th17/citología , Células Th17/inmunología , Células Th17/metabolismo
2.
Nature ; 571(7764): 270-274, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31207604

RESUMEN

Tumour-specific CD8 T cell dysfunction is a differentiation state that is distinct from the functional effector or memory T cell states1-6. Here we identify the nuclear factor TOX as a crucial regulator of the differentiation of tumour-specific T (TST) cells. We show that TOX is highly expressed in dysfunctional TST cells from tumours and in exhausted T cells during chronic viral infection. Expression of TOX is driven by chronic T cell receptor stimulation and NFAT activation. Ectopic expression of TOX in effector T cells in vitro induced a transcriptional program associated with T cell exhaustion. Conversely, deletion of Tox in TST cells in tumours abrogated the exhaustion program: Tox-deleted TST cells did not upregulate genes for inhibitory receptors (such as Pdcd1, Entpd1, Havcr2, Cd244 and Tigit), the chromatin of which remained largely inaccessible, and retained high expression of transcription factors such as TCF-1. Despite their normal, 'non-exhausted' immunophenotype, Tox-deleted TST cells remained dysfunctional, which suggests that the regulation of expression of inhibitory receptors is uncoupled from the loss of effector function. Notably, although Tox-deleted CD8 T cells differentiated normally to effector and memory states in response to acute infection, Tox-deleted TST cells failed to persist in tumours. We hypothesize that the TOX-induced exhaustion program serves to prevent the overstimulation of T cells and activation-induced cell death in settings of chronic antigen stimulation such as cancer.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diferenciación Celular/inmunología , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas de Homeodominio/metabolismo , Neoplasias/inmunología , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Proteínas de Homeodominio/genética , Humanos , Memoria Inmunológica , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Ratones , Neoplasias/patología , Fenotipo , Receptores de Antígenos de Linfocitos T/inmunología , Transcripción Genética
3.
PLoS One ; 13(6): e0199785, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29953487

RESUMEN

FAcilitates Chromatin Transcription (FACT) is a complex of SSRP1 and SPT16 that is involved in chromatin remodeling during transcription, replication, and DNA repair. FACT has been mostly studied in cell-free or single cell model systems because general FACT knockout (KO) is embryonically lethal (E3.5). FACT levels are limited to the early stages of development and stem cell niches of adult tissues. FACT is upregulated in poorly differentiated aggressive tumors. Importantly, FACT inhibition (RNAi) is lethal for tumors but not normal cells, making FACT a lucrative target for anticancer therapy. To develop a better understanding of FACT function in the context of the mammalian organism under normal physiological conditions and in disease, we aimed to generate a conditional FACT KO mouse model. Because SPT16 stability is dependent on the SSRP1-SPT16 association and the presence of SSRP1 mRNA, we targeted the Ssrp1 gene using a CreERT2- LoxP approach to generate the FACT KO model. Here, we highlight the limitations of the CreERT2-LoxP (Rosa26) system that we encountered during the generation of this model. In vitro studies showed an inefficient excision rate of ectopically expressed CreERT2 (retroviral CreERT2) in fibroblasts with homozygous floxed Ssrp1. In vitro and in vivo studies showed that the excision efficiency could only be increased with germline expression of two alleles of Rosa26CreERT2. The expression of one germline Rosa26CreERT2 allele led to the incomplete excision of Ssrp1. The limited efficiency of the CreERT2-LoxP system may be sufficient for studies involving the deletion of genes that interfere with cell growth or viability due to the positive selection of the phenotype. However, it may not be sufficient for studies that involve the deletion of genes supporting growth, or those crucial for development. Although CreERT2-LoxP is broadly used, it has limitations that have not been widely discussed. This paper aims to encourage such discussions.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Técnicas de Inactivación de Genes/métodos , Proteínas del Grupo de Alta Movilidad/deficiencia , Integrasas , Complejos Multiproteicos , Factores de Transcripción , Animales , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Integrasas/genética , Integrasas/metabolismo , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
4.
Antioxid Redox Signal ; 24(7): 345-60, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26725491

RESUMEN

AIMS: The differentiation of embryonic stem (ES) cells into energetically efficient cardiomyocytes contributes to functional cardiac repair and is envisioned to ameliorate progressive degenerative cardiac diseases. Advanced cell maturation strategies are therefore needed to create abundant mature cardiomyocytes. In this study, we tested whether the redox-sensitive heme oxygenase-1/carbon monoxide (HO-1/CO) system, operating through mitochondrial biogenesis, acts as a mechanism for ES cell differentiation and cardiomyocyte maturation. RESULTS: Manipulation of HO-1/CO to enhance mitochondrial biogenesis demonstrates a direct pathway to ES cell differentiation and maturation into beating cardiomyocytes that express adult structural markers. Targeted HO-1/CO interventions up- and downregulate specific cardiogenic transcription factors, transcription factor Gata4, homeobox protein Nkx-2.5, heart- and neural crest derivatives-expressed protein 1, and MEF2C. HO-1/CO overexpression increases cardiac gene expression for myosin regulatory light chain 2, atrial isoform, MLC2v, ANP, MHC-ß, and sarcomere α-actinin and the major mitochondrial fusion regulators, mitofusin 2 and MICOS complex subunit Mic60. This promotes structural mitochondrial network expansion and maturation, thereby supporting energy provision for beating embryoid bodies. These effects are prevented by silencing HO-1 and by mitochondrial reactive oxygen species scavenging, while disruption of mitochondrial biogenesis and mitochondrial DNA depletion by loss of mitochondrial transcription factor A compromise infrastructure. This leads to failure of cardiomyocyte differentiation and maturation and contractile dysfunction. INNOVATION: The capacity to augment cardiomyogenesis via a defined mitochondrial pathway has unique therapeutic potential for targeting ES cell maturation in cardiac disease. CONCLUSION: Our findings establish the HO-1/CO system and redox regulation of mitochondrial biogenesis as essential factors in ES cell differentiation as well as in the subsequent maturation of these cells into functional cardiac cells.


Asunto(s)
Monóxido de Carbono/metabolismo , Diferenciación Celular , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/metabolismo , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Animales , Diferenciación Celular/genética , Línea Celular , Autorrenovación de las Células/genética , ADN Mitocondrial/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Activación Enzimática , Regulación del Desarrollo de la Expresión Génica , Técnicas de Inactivación de Genes , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Ratones , Mitocondrias/genética , Mitocondrias/metabolismo , Mioblastos Cardíacos/citología , Mioblastos Cardíacos/metabolismo , Miocitos Cardíacos/fisiología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Oxidación-Reducción , ARN Mensajero/genética , Ratas , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/metabolismo , Transducción de Señal
5.
Development ; 142(13): 2278-90, 2015 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-26041766

RESUMEN

In the developing mammalian brain, neural stem cells (NSCs) initially expand the progenitor pool by symmetric divisions. NSCs then shift from symmetric to asymmetric division and commence neurogenesis. Although the precise mechanisms regulating the developmental timing of this transition have not been fully elucidated, gradual elongation in the length of the cell cycle and coinciding accumulation of determinants that promote neuronal differentiation might function as a biological clock that regulates the onset of asymmetric division and neurogenesis. We conducted gene expression profiling of embryonic NSCs in the cortical regions and found that expression of high mobility group box transcription factor 1 (Hbp1) was upregulated during neurogenic stages. Induced conditional knockout mice of Hbp1, generated by crossing with Nestin-CreER(T2) mice, exhibited a remarkable dilatation of the telencephalic vesicles with a tangentially expanded ventricular zone and a thinner cortical plate containing reduced numbers of neurons. In these Hbp1-deficient mouse embryos, neural stem/progenitor cells continued to divide with a shorter cell cycle length. Moreover, downstream target genes of the Wnt signaling, such as cyclin D1 (Ccnd1) and c-jun (Jun), were upregulated in the germinal zone of the cortical regions. These results indicate that Hbp1 plays a crucial role in regulating the timing of cortical neurogenesis by elongating the cell cycle and that it is essential for normal cortical development.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Proteínas del Grupo de Alta Movilidad/metabolismo , Neuronas/citología , Neuronas/metabolismo , Proteínas Represoras/metabolismo , Animales , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Ciclo Celular/genética , Diferenciación Celular/genética , Movimiento Celular/genética , Proliferación Celular , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Ciclina D1/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Técnicas de Silenciamiento del Gen , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Ratones Endogámicos ICR , Ratones Noqueados , Morfogénesis , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis/genética , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Proteína de Retinoblastoma/metabolismo , Factores de Tiempo , Regulación hacia Arriba/genética
6.
Nature ; 520(7548): 553-7, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25642965

RESUMEN

Mitochondrial DNA (mtDNA) is normally present at thousands of copies per cell and is packaged into several hundred higher-order structures termed nucleoids. The abundant mtDNA-binding protein TFAM (transcription factor A, mitochondrial) regulates nucleoid architecture, abundance and segregation. Complete mtDNA depletion profoundly impairs oxidative phosphorylation, triggering calcium-dependent stress signalling and adaptive metabolic responses. However, the cellular responses to mtDNA instability, a physiologically relevant stress observed in many human diseases and ageing, remain poorly defined. Here we show that moderate mtDNA stress elicited by TFAM deficiency engages cytosolic antiviral signalling to enhance the expression of a subset of interferon-stimulated genes. Mechanistically, we find that aberrant mtDNA packaging promotes escape of mtDNA into the cytosol, where it engages the DNA sensor cGAS (also known as MB21D1) and promotes STING (also known as TMEM173)-IRF3-dependent signalling to elevate interferon-stimulated gene expression, potentiate type I interferon responses and confer broad viral resistance. Furthermore, we demonstrate that herpesviruses induce mtDNA stress, which enhances antiviral signalling and type I interferon responses during infection. Our results further demonstrate that mitochondria are central participants in innate immunity, identify mtDNA stress as a cell-intrinsic trigger of antiviral signalling and suggest that cellular monitoring of mtDNA homeostasis cooperates with canonical virus sensing mechanisms to fully engage antiviral innate immunity.


Asunto(s)
ADN Mitocondrial/metabolismo , Herpesvirus Humano 1/inmunología , Inmunidad Innata/inmunología , Estrés Fisiológico , Animales , Línea Celular , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Humanos , Factor 3 Regulador del Interferón/metabolismo , Interferón Tipo I/inmunología , Proteínas de la Membrana/metabolismo , Ratones , Nucleotidiltransferasas/metabolismo
7.
J Invest Dermatol ; 135(3): 679-689, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25371971

RESUMEN

Here, we studied how epithelial energy metabolism impacts overall skin development by selectively deleting intraepithelial mtDNA in mice by ablating a key maintenance factor (Tfam(EKO)), which induces loss of function of the electron transport chain (ETC). Quantitative (immuno)histomorphometry demonstrated that Tfam(EKO) mice showed significantly reduced hair follicle (HF) density and morphogenesis, fewer intrafollicular keratin15+ epithelial progenitor cells, increased apoptosis, and reduced proliferation. Tfam(EKO) mice also displayed premature entry into (aborted) HF cycling by apoptosis-driven HF regression (catagen). Ultrastructurally, Tfam(EKO) mice exhibited severe HF dystrophy, pigmentary abnormalities, and telogen-like condensed dermal papillae. Epithelial HF progenitor cell differentiation (Plet1, Lrig1 Lef1, and ß-catenin), sebaceous gland development (adipophilin, Scd1, and oil red), and key mediators/markers of epithelial-mesenchymal interactions during skin morphogenesis (NCAM, versican, and alkaline phosphatase) were all severely altered in Tfam(EKO) mice. Moreover, the number of mast cells, major histocompatibility complex class II+, or CD11b+ immunocytes in the skin mesenchyme was increased, and essentially no subcutis developed. Therefore, in contrast to their epidermal counterparts, pilosebaceous unit stem cells depend on a functional ETC. Most importantly, our findings point toward a frontier in skin biology: the coupling of HF keratinocyte mitochondrial function with the epithelial-mesenchymal interactions that drive overall development of the skin and its appendages.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Folículo Piloso/crecimiento & desarrollo , Mitocondrias/fisiología , Morfogénesis/fisiología , Fenómenos Fisiológicos de la Piel , Animales , Apoptosis/fisiología , Proliferación Celular , ADN Mitocondrial/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Metabolismo Energético/fisiología , Epitelio/fisiología , Folículo Piloso/citología , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/fisiología , Ratones , Ratones Noqueados , Modelos Animales
8.
Gastroenterology ; 146(4): 1097-107, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24361123

RESUMEN

BACKGROUND & AIMS: High mobility group box 1 (HMGB1) is an abundant protein that regulates chromosome architecture and also functions as a damage-associated molecular pattern molecule. Little is known about its intracellular roles in response to tissue injury or during subsequent local and systemic inflammatory responses. We investigated the function of Hmgb1 in mice after induction of acute pancreatitis. METHODS: We utilized a Cre/LoxP system to create mice with pancreas-specific disruption in Hmbg1 (Pdx1-Cre; HMGB1(flox/flox) mice). Acute pancreatitis was induced in these mice (HMGB1(flox/flox) mice served as controls) after injection of l-arginine or cerulein. Pancreatic tissues and acinar cells were collected and analyzed by histologic, immunoblot, and immunohistochemical analyses. RESULTS: After injection of l-arginine or cerulein, Pdx1-Cre; HMGB1(flox/flox) mice developed acute pancreatitis more rapidly than controls, with increased mortality. Pancreatic tissues of these mice also had higher levels of serum amylase, acinar cell death, leukocyte infiltration, and interstitial edema than controls. Pancreatic tissues and acinar cells collected from the Pdx1-Cre; HMGB1(flox/flox) mice after l-arginine or cerulein injection demonstrated nuclear catastrophe with greater nucleosome release when compared with controls, along with increased phosphorylation/activation of RELA nuclear factor κB, degradation of inhibitor of κB, and phosphorylation of mitogen-activated protein kinase. Inhibitors of reactive oxygen species (N-acetyl-l-cysteine) blocked l-arginine-induced DNA damage, necrosis, apoptosis, release of nucleosomes, and activation of nuclear factor κB in pancreatic tissues and acinar cells from Pdx1-Cre; HMGB1(flox/flox) and control mice. Exogenous genomic DNA and recombinant histone H3 proteins significantly induced release of HMGB1 from mouse macrophages; administration of antibodies against H3 to mice reduced serum levels of HMGB1 and increased survival after l-arginine injection. CONCLUSIONS: In 2 mouse models of acute pancreatitis, intracellular HMGB1 appeared to prevent nuclear catastrophe and release of inflammatory nucleosomes to block inflammation. These findings indicate a role for the innate immune response in tissue damage.


Asunto(s)
Proteínas del Grupo de Alta Movilidad/metabolismo , Nucleosomas/metabolismo , Páncreas/metabolismo , Pancreatitis/prevención & control , Proteínas Represoras/metabolismo , Enfermedad Aguda , Animales , Arginina , Muerte Celular , Ceruletida , Daño del ADN , Modelos Animales de Enfermedad , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Histonas/metabolismo , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Estrés Oxidativo , Páncreas/inmunología , Páncreas/patología , Pancreatitis/inducido químicamente , Pancreatitis/genética , Pancreatitis/inmunología , Pancreatitis/metabolismo , Pancreatitis/patología , Especies Reactivas de Oxígeno/metabolismo , Proteínas Represoras/deficiencia , Proteínas Represoras/genética , Transducción de Señal , Factores de Tiempo
9.
Am J Pathol ; 180(1): 24-31, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22056359

RESUMEN

Alterations in mitochondrial oxidative phosphorylation have long been documented in tumors. Other types of mitochondrial dysfunction, including altered reactive oxygen species (ROS) production and apoptosis, also can contribute to tumorigenesis and cancer phenotypes. Furthermore, mutation and altered amounts of mitochondrial DNA (mtDNA) have been observed in cancer cells. However, how mtDNA instability per se contributes to cancer remains largely undetermined. Mitochondrial transcription factor A (TFAM) is required for expression and maintenance of mtDNA. Tfam heterozygous knock-out (Tfam(+/-)) mice show mild mtDNA depletion, but have no overt phenotypes. We show that Tfam(+/-) mouse cells and tissues not only possess less mtDNA but also increased oxidative mtDNA damage. Crossing Tfam(+/-) mice to the adenomatous polyposis coli multiple intestinal neoplasia (APC(Min/+)) mouse cancer model revealed that mtDNA instability increases tumor number and growth in the small intestine. This was not a result of enhancement of Wnt/ß-catenin signaling, but rather appears to involve a propensity for increased mitochondrial ROS production. Direct involvement of mitochondrial ROS in intestinal tumorigenesis was shown by crossing APC(Min/+) mice to those that have catalase targeted to mitochondria, which resulted in a significant reduction in tumorigenesis in the colon. Thus, mitochondrial genome instability and ROS enhance intestinal tumorigenesis and Tfam(+/-) mice are a relevant model to address the role of mtDNA instability in disease states in which mitochondrial dysfunction is implicated, such as cancer, neurodegeneration, and aging.


Asunto(s)
Poliposis Adenomatosa del Colon/etiología , Proteínas de Unión al ADN/fisiología , Genoma Mitocondrial/fisiología , Inestabilidad Genómica/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Enfermedades Mitocondriales/etiología , Especies Reactivas de Oxígeno/metabolismo , Poliposis Adenomatosa del Colon/metabolismo , Animales , Transformación Celular Neoplásica , Daño del ADN/fisiología , ADN Mitocondrial/fisiología , Proteínas de Unión al ADN/deficiencia , Proteínas del Grupo de Alta Movilidad/deficiencia , Ratones , Ratones Noqueados , Enfermedades Mitocondriales/metabolismo
10.
PLoS One ; 6(11): e27727, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22110743

RESUMEN

Mitochondrial dysfunction is a central mediator of disease progression in diverse neurodegenerative diseases that often present with prominent gastrointestinal abnormalities. Gastrointestinal dysfunction in these disorders is related, at least in part, to defects in the enteric nervous system (ENS). The role of mitochondrial deficits in ENS neurodegeneration and their relative contribution to gastrointestinal dysfunction, however, are unclear. To better understand how mitochondrial abnormalities in the ENS influence enteric neurodegeneration and affect intestinal function, we generated mice (Tfam-ENSKOs) with impaired mitochondrial metabolism in enteric neurons and glia through the targeted deletion of the mitochondrial transcription factor A gene (Tfam). Tfam-ENSKO mice were initially viable but, at an early age, they developed severe gastrointestinal motility problems characterized by intestinal pseudo-obstruction resulting in premature death. This gastrointestinal dysfunction was caused by extensive, progressive neurodegeneration of the ENS involving both neurons and glia. Interestingly, mitochondrial defects differentially affected specific subpopulations of enteric neurons and regions of the gastrointestinal tract. Mitochondrial deficiency-related neuronal and glial loss was most prominent in the proximal small intestine, but the first affected neurons, nitrergic inhibitory neurons, had the greatest losses in the distal small intestine. This regional and subtype-specific variability in susceptibility to mitochondrial defects resulted in an imbalance of inhibitory and excitatory neurons that likely accounts for the observed phenotype in Tfam-ENSKO mice. Mitochondrial dysfunction, therefore, is likely to be an important driving force of neurodegeneration in the ENS and contribute to gastrointestinal symptoms in people with neurodegenerative disorders.


Asunto(s)
Sistema Nervioso Entérico/patología , Enfermedades Mitocondriales/patología , Neuronas/patología , Animales , Axones/metabolismo , Axones/patología , Recuento de Células , Replicación del ADN/genética , ADN Mitocondrial/biosíntesis , ADN Mitocondrial/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Susceptibilidad a Enfermedades , Sistema Nervioso Entérico/metabolismo , Sistema Nervioso Entérico/fisiopatología , Motilidad Gastrointestinal/genética , Tracto Gastrointestinal/fisiopatología , Eliminación de Gen , Técnicas de Inactivación de Genes , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Ratones , Mitocondrias/metabolismo , Mitocondrias/patología , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Enfermedades Mitocondriales/fisiopatología , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Enfermedades Neurodegenerativas/patología , Enfermedades Neurodegenerativas/fisiopatología , Neuroglía/metabolismo , Neuroglía/patología , Neuronas/metabolismo , Neuronas Nitrérgicas/metabolismo , Neuronas Nitrérgicas/patología , Especificidad de Órganos , Transcripción Genética/genética
11.
Physiol Res ; 60(Suppl 1): S61-70, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21777023

RESUMEN

Understanding mitochondrial role in normal physiology and pathological conditions has proven to be of high importance as mitochondrial dysfunction is connected with a number of disorders as well as some of the most common diseases (e.g. diabetes or Parkinson's disease). Modeling mitochondrial dysfunction has been difficult mainly due to unique features of mitochondrial genetics. Here we discuss some of the most important mouse models generated so far and lessons learned from them.


Asunto(s)
ADN Mitocondrial/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , Envejecimiento/genética , Animales , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Ratones , Ratones Noqueados , Ratones Transgénicos , Enfermedades Mitocondriales/metabolismo , Mutación , Degeneración Nerviosa/genética
12.
Stem Cells ; 29(9): 1459-68, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21780252

RESUMEN

Tissue stem cells and germ line or embryonic stem cells were shown to have reduced oxidative metabolism, which was proposed to be an adaptive mechanism to reduce damage accumulation caused by reactive oxygen species. However, an alternate explanation is that stem cells are less dependent on specialized cytoplasmic functions compared with differentiated cells, therefore, having a high nuclear-to-cytoplasmic volume ratio and consequently a low mitochondrial content. To determine whether stem cells rely or not on mitochondrial respiration, we selectively ablated the electron transport chain in the basal layer of the epidermis, which includes the epidermal progenitor/stem cells (EPSCs). This was achieved using a loxP-flanked mitochondrial transcription factor A (Tfam) allele in conjunction with a keratin 14 Cre transgene. The epidermis of these animals (Tfam(EKO)) showed a profound depletion of mitochondrial DNA and complete absence of respiratory chain complexes. However, despite a short lifespan due to malnutrition, epidermal development and skin barrier function were not impaired. Differentiation of epidermal layers was normal and no proliferation defect or major increase of apoptosis could be observed. In contrast, mice with an epidermal ablation of prohibitin-2, a scaffold protein in the inner mitochondrial membrane, displayed a dramatic phenotype observable already in utero, with severely impaired skin architecture and barrier function, ultimately causing death from dehydration shortly after birth. In conclusion, we here provide unequivocal evidence that EPSCs, and probably tissue stem cells in general, are independent of the mitochondrial respiratory chain, but still require a functional dynamic mitochondrial compartment.


Asunto(s)
Células Epidérmicas , Mitocondrias/metabolismo , Células Madre/citología , Células Madre/metabolismo , Animales , Apoptosis/fisiología , Diferenciación Celular/fisiología , Procesos de Crecimiento Celular/fisiología , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Transporte de Electrón , Epidermis/metabolismo , Genotipo , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Inmunohistoquímica , Ratones , Ratones Noqueados , Mitocondrias/genética , Especies Reactivas de Oxígeno/metabolismo
13.
Adv Exp Med Biol ; 682: 279-96, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20824532

RESUMEN

Skeletal muscle weakness is an important feature of numerous -pathological conditions and it may also be a component in normal ageing. Decreased muscular strength can be due to decreased muscle mass and/or intrinsic defects in the muscle cells. In this chapter we will discuss decreased force production due to mechanisms intrinsic to skeletal muscle cells. We will mainly use data from mouse disease models to exemplify defects at various sites in the cellular activation-contraction pathway. We will show that depending on the underlying problem, muscle weakness can be due decreased Ca²(+) release from the sarcoplasmic reticulum, reduced myofibrillar Ca²(+) sensitivity and/or decreased ability of the cross-bridges to generate force.


Asunto(s)
Debilidad Muscular/fisiopatología , Músculo Esquelético/fisiopatología , Acetilcolina/farmacología , Animales , Calcio/metabolismo , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Modelos Animales de Enfermedad , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Ratones , Ratones Noqueados , Neuronas Motoras/fisiología , Contracción Muscular/fisiología , Debilidad Muscular/genética , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/fisiología , Miofibrillas/fisiología
14.
Dev Biol ; 348(1): 58-66, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20849842

RESUMEN

Metazoan stem cells repopulate tissues during adult life by dividing asymmetrically to generate another stem cell and a cell that terminally differentiates. Wnt signaling regulates the division pattern of stem cells in flies and vertebrates. While the short-lived nematode C. elegans has no adult somatic stem cells, the lateral epithelial seam cells divide in a stem cell-like manner in each larval stage, usually generating a posterior daughter that retains the seam cell fate and an anterior daughter that terminally differentiates. We show that while wild-type adult animals have 16 seam cells per side, animals with reduced function of the TCF homolog POP-1 have as many as 67 seam cells, and animals with reduced function of the ß-catenins SYS-1 and WRM-1 have as few as three. Analysis of seam cell division patterns showed alterations in their stem cell-like divisions in the L2-L4 stages: reduced Wnt signaling caused both daughters to adopt non-seam fates, while activated Wnt signaling caused both daughters to adopt the seam fate. Therefore, our results indicate that Wnt signaling globally regulates the asymmetric, stem cell-like division of most or all somatic seam cells during C. elegans larval development, and that Wnt pathway regulation of stem cell-like behavior is conserved in nematodes.


Asunto(s)
Proteínas de Caenorhabditis elegans/fisiología , Caenorhabditis elegans/crecimiento & desarrollo , Proteínas del Citoesqueleto/fisiología , Proteínas de Unión al ADN/fisiología , Células Epiteliales/citología , Proteínas del Grupo de Alta Movilidad/fisiología , Transducción de Señal/fisiología , Factores de Transcripción/fisiología , Proteínas Wnt/fisiología , Animales , Caenorhabditis elegans/citología , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/antagonistas & inhibidores , Proteínas de Caenorhabditis elegans/genética , Recuento de Células , Diferenciación Celular , División Celular , Proteínas del Citoesqueleto/deficiencia , Proteínas del Citoesqueleto/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Células Epiteliales/metabolismo , Genes Reporteros , Proteínas del Grupo de Alta Movilidad/antagonistas & inhibidores , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Larva , Interferencia de ARN , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/fisiología , Proteínas Represoras/fisiología , Transducción de Señal/genética , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética
15.
Genes Brain Behav ; 9(2): 173-81, 2010 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-20002202

RESUMEN

The MitoPark mouse, in which the mitochondrial transcription factor Tfam is selectively removed in midbrain dopamine (DA) neurons, is a genetic model for Parkinson's disease (PD) that replicates the slow and progressive development of key symptoms. To further validate this model, we have extended both behavioral and biochemical analyses in these animals. We found that vertical movements decline earlier and faster than horizontal movements, possibly modeling the early occurrence of axial, postural instability in PD. L-DOPA induces different locomotor responses depending on the age: in young MitoPark mice the L-DOPA-induced motor activation is small; middle-aged MitoPark mice respond in a dose-dependent manner to L-DOPA, whereas aged MitoPark mice display a double-peaked locomotor response to a high dose of L-DOPA that includes an intermittent period of very low motor activity, similar to the 'on-off' phenomenon in PD. To correlate behavior with biochemical data, we analyzed monoamine levels in three different brain areas that are highly innervated by the DA system: striatum, anterior cortex and olfactory bulb. DA levels declined earlier and faster in striatum than in cortex; only at the latest time-point analyzed, DA levels were found to be significantly lower than control levels in the olfactory bulb. Interestingly, the ratio between homovanillic acid (HVA) and DA differed between regions over time. In striatum and olfactory bulb, the ratio increased steeply indicating increased DA turnover. In contrast, the ratio decreased over time in cortex, revealing important differences between DA cells in substantia nigra and the ventral tegmental area.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Proteínas del Grupo de Alta Movilidad/deficiencia , Levodopa/uso terapéutico , Enfermedad de Parkinson/tratamiento farmacológico , Enfermedad de Parkinson/genética , Animales , Emparejamiento Base , Cartilla de ADN , Proteínas de Unión al ADN/genética , Progresión de la Enfermedad , Relación Dosis-Respuesta a Droga , Femenino , Proteínas del Grupo de Alta Movilidad/genética , Masculino , Mesencéfalo/metabolismo , Mesencéfalo/patología , Ratones , Ratones Noqueados , Modelos Genéticos , Actividad Motora , Enfermedad de Parkinson/fisiopatología , Reacción en Cadena de la Polimerasa , Postura
16.
J Cell Biochem ; 108(2): 508-18, 2009 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-19639603

RESUMEN

A possible role for structure-specific recognition protein 1 (SSRP1) in replication-associated repair processes has previously been suggested based on its interaction with several DNA repair factors and the replication defects observed in SSRP1 mutants. In this study, we investigated the potential role of SSRP1 in association with DNA repair mediated by homologous recombination (HR), one of the pathways involved in repairing replication-associated DNA damage, in mammalian cells. Surprisingly, over-expression of SSRP1 reduced the number of hprt(+) recombinants generated via HR both spontaneously and upon hydroxyurea (HU) treatment, whereas knockdown of SSRP1 resulted in an increase of HR events in response to DNA double-strand break formation. In correlation, we found that the depletion of SSRP1 in HU-treated human cells elevated the number of Rad51 and H2AX foci, while over-expression of the wild-type SSRP1 markedly reduced HU-induced Rad51 foci formation. We also found that SSRP1 physically interacts with a key HR repair protein, Rad54 both in vitro and in vivo. Further, branch migration studies demonstrated that SSRP1 inhibits Rad54-promoted branch migration of Holliday junctions in vitro. Taken together, our data suggest a functional role for SSRP1 in spontaneous and replication-associated DNA damage response by suppressing avoidable HR repair events.


Asunto(s)
Reparación del ADN , Proteínas de Unión al ADN/fisiología , Proteínas del Grupo de Alta Movilidad/fisiología , Recombinación Genética , Factores de Elongación Transcripcional/fisiología , Animales , Western Blotting , Cricetinae , Cricetulus , Roturas del ADN de Doble Cadena , Daño del ADN , ADN Helicasas , ADN Cruciforme/metabolismo , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Técnicas de Silenciamiento del Gen , Genes Reporteros , Proteínas del Grupo de Alta Movilidad/biosíntesis , Proteínas del Grupo de Alta Movilidad/deficiencia , Proteínas del Grupo de Alta Movilidad/genética , Histonas/metabolismo , Humanos , Hidroxiurea , Proteínas Mutantes/aislamiento & purificación , Proteínas Mutantes/metabolismo , Proteínas Nucleares/metabolismo , Fragmentos de Péptidos , Plásmidos , Unión Proteica , ARN Interferente Pequeño , Recombinasa Rad51/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Proteínas Recombinantes de Fusión/metabolismo , Factores de Elongación Transcripcional/biosíntesis , Factores de Elongación Transcripcional/deficiencia , Factores de Elongación Transcripcional/genética , Transfección
17.
Proc Natl Acad Sci U S A ; 105(34): 12337-42, 2008 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-18719100

RESUMEN

A major goal for developmental biologists is to define the behaviors and molecular contents of differentiating cells. We have devised a strategy for isolating cells from diverse embryonic regions and stages in the zebrafish, using computer-guided laser photoconversion of injected Kaede protein and flow cytometry. This strategy enabled us to perform a genome-wide transcriptome comparison of germ layer precursor cells. Mesendoderm and ectoderm precursors cells isolated by this method differentiated appropriately in transplantation assays. Microarray analysis of these cells reidentified known genes at least as efficiently as previously reported strategies that relied on artificial mesendoderm activation or inhibition. We also identified a large set of uncharacterized mesendoderm-enriched genes as well as ectoderm-enriched genes. Loss-of-function studies revealed that one of these genes, the MAP kinase inhibitor dusp4, is essential for early development. Embryos injected with antisense morpholino oligonucleotides that targeted Dusp4 displayed necrosis of head tissues. Marker analysis during late gastrulation revealed a specific loss of sox17, but not of other endoderm markers, and analysis at later stages revealed a loss of foregut and pancreatic endoderm. This specific loss of sox17 establishes a new class of endoderm specification defect.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Fosfatasas de Especificidad Dual/genética , Estratos Germinativos/citología , Proteínas del Grupo de Alta Movilidad/deficiencia , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/genética , Factores de Transcripción/deficiencia , Transcripción Genética , Proteínas de Pez Cebra/deficiencia , Proteínas de Pez Cebra/genética , Animales , Fosfatasas de Especificidad Dual/fisiología , Ectodermo/citología , Ectodermo/embriología , Embrión no Mamífero , Desarrollo Embrionario/genética , Inducción Embrionaria/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Estratos Germinativos/embriología , Mesodermo/citología , Mesodermo/embriología , Fosfatasas de la Proteína Quinasa Activada por Mitógenos/fisiología , Factores de Transcripción SOXF , Proteínas de Pez Cebra/fisiología
18.
Exp Eye Res ; 86(6): 895-907, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18423449

RESUMEN

We describe that Sox11, a member of the group C of the Sox transcription factor family, is critically required during the morphogenetic processes of early eye development, and that lack of Sox11 results in ocular anterior segment dysgenesis (ASD). Sox11-deficient mice show a persistent lens stalk, a delay in lens formation, and the phenotypes of Peters' anomaly and microphthalmia at birth. In addition, the optic fissure does not close in the anterior halves of the eyes resulting in anterior coloboma. The delay in lens formation is associated with a reduced mitotic activity in the lens placode during its invagination into the optic cup. No changes in Pax6 expression are observed in the developing eyes of Sox11-/- mice, whereas the expression of Sox11 is reduced in optic cup, optic vesicle and lens placode of Pax6+/- embryos and in the optic vesicle of Pax6-/- mice. Transfection experiments show an increase in Sox11 expression when higher doses of Pax6 are present. Considerably smaller amounts of BMP7 are expressed in lens and optic cup of Sox11-/- mice as compared to their wild-type littermates. We conclude that Sox11 is required during separation of the lens vesicle from the surface ectoderm and the closure of the anterior optic fissure. The expression of Sox11 in early eye development is under control of Pax6, and changes in BMP7-signalling appear to be involved in the effects of Sox11 on anterior eye development. Our findings suggest that SOX11 might similarly be involved in the pathogenesis of ASD in human patients.


Asunto(s)
Segmento Anterior del Ojo/anomalías , Proteínas del Grupo de Alta Movilidad/deficiencia , Animales , Segmento Anterior del Ojo/patología , Western Blotting/métodos , Proteína Morfogenética Ósea 7 , Proteínas Morfogenéticas Óseas/metabolismo , Coloboma/genética , Coloboma/patología , Proteínas del Ojo/fisiología , Desarrollo Fetal , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/metabolismo , Proteínas del Grupo de Alta Movilidad/fisiología , Proteínas de Homeodominio/fisiología , Cristalino/embriología , Ratones , Ratones Endogámicos C57BL , Microftalmía/genética , Microftalmía/patología , Mitosis , Factor de Transcripción PAX6 , Factores de Transcripción Paired Box/fisiología , ARN Mensajero/genética , Proteínas Represoras/fisiología , Factores de Transcripción SOXC , Factor de Crecimiento Transformador beta/metabolismo
19.
Int J Dev Biol ; 52(4): 323-32, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18415932

RESUMEN

The transcription factor Sox9 plays an important role during chondrogenesis. After early conditional inactivation of Sox9 in mesenchymal limb bud cells of mice, mesenchymal condensations as well as cartilage and bone are completely absent in the developing limbs. We analyzed chondrogenic differentiation of Sox9-/- mouse embryonic stem cells in vitro, using two clones with different targeted mutations. We found that the development of mature and hypertrophic chondrocytes is completely inhibited in the absence of Sox9 confirming that Sox9 is required for the formation of cartilage. In contrast, Sox9+/- mouse embryonic stem cells showed continuous but reduced differentiation into mature chondrocytes. Interestingly, the formation of early chondrogenic condensations expressing characteristic marker genes such as scleraxis, Sox5 and Sox6 was not inhibited in the absence of Sox9 in vitro. Thus, we propose that the earliest step of chondrogenesis could be regulated by a non cell-autonomous function of Sox9.


Asunto(s)
Condrocitos/química , Condrocitos/metabolismo , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Proteínas del Grupo de Alta Movilidad/deficiencia , Factores de Transcripción/deficiencia , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cartílago/citología , Cartílago/crecimiento & desarrollo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Condrogénesis/genética , Condrogénesis/fisiología , Células Clonales , Colágeno Tipo II/genética , ADN/genética , Exones , Regulación del Desarrollo de la Expresión Génica , Marcación de Gen , Proteínas del Grupo de Alta Movilidad/genética , Proteínas del Grupo de Alta Movilidad/fisiología , Ratones , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción SOX9 , Factores de Transcripción/genética , Factores de Transcripción/fisiología
20.
Development ; 135(4): 637-46, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18184726

RESUMEN

Specification of the myelin-forming oligodendrocytes of the central nervous system requires the Sox9 transcription factor, whereas terminal differentiation depends on the closely related Sox10. Between specification and terminal differentiation, Sox9 and Sox10 are co-expressed in oligodendrocyte precursors and are believed to exert additional functions. To identify such functions, we have deleted Sox9 specifically in already specified oligodendrocyte precursors of the spinal cord. In the absence of Sox9, oligodendrocyte precursors developed normally and started terminal differentiation on schedule. However, when Sox10 was additionally deleted, oligodendrocyte precursors exhibited an altered migration pattern and were present in reduced numbers because of increased apoptosis rates. Remaining precursors continued to express many characteristic oligodendroglial markers. Aberrant expression of astrocytic and neuronal markers was not observed. Strikingly, we failed to detect PDGF receptor alpha expression in the mutant oligodendrocyte precursors, arguing that PDGF receptor alpha is under transcriptional control of Sox9 and Sox10. Altered PDGF receptor alpha expression is furthermore sufficient to explain the observed phenotype, as PDGF is both an important survival factor and migratory cue for oligodendrocyte precursors. We thus conclude that Sox9 and Sox10 are required in a functionally redundant manner in oligodendrocyte precursors for PDGF-dependent survival and migration.


Asunto(s)
Movimiento Celular , Proteínas de Unión al ADN/metabolismo , Proteínas del Grupo de Alta Movilidad/metabolismo , Oligodendroglía/citología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Médula Espinal/citología , Células Madre/citología , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores/metabolismo , Recuento de Células , Linaje de la Célula , Proliferación Celular , Supervivencia Celular , Transdiferenciación Celular , Proteínas de Unión al ADN/deficiencia , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica , Proteínas del Grupo de Alta Movilidad/deficiencia , Integrasas/metabolismo , Ratones , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos , Oligodendroglía/metabolismo , Factor de Transcripción SOX9 , Factores de Transcripción SOXE , Médula Espinal/embriología , Médula Espinal/metabolismo , Células Madre/metabolismo , Factores de Transcripción/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...