Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Transgenic Res ; 30(5): 701-707, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34117597

RESUMEN

Chronic cholestatic liver diseases including primary sclerosing cholangitis (PSC) present a complex spectrum with regards to the cause, age of manifestation and histopathological features. Current treatment options are severely limited primarily due to a paucity of model systems mirroring the disease. Here, we describe the Keratin 5 (K5)-Cre; Klf5fl/fl mouse that spontaneously develops severe liver disease during the postnatal period with features resembling PSC including a prominent ductular reaction, fibrotic obliteration of the bile ducts and secondary degeneration/necrosis of liver parenchyma. Over time, there is an expansion of Sox9+ hepatocytes in the damaged livers suggestive of a hepatocyte-mediated regenerative response. We conclude that Klf5 is required for the normal function of the hepatobiliary system and that the K5-Cre; Klf5fl/fl mouse is an excellent model to probe the molecular events interlinking damage and regenerative response in the liver.


Asunto(s)
Colangitis Esclerosante , Hepatopatías , Animales , Integrasas , Queratina-5 , Factores de Transcripción de Tipo Kruppel/genética , Hígado , Ratones
2.
PLoS One ; 15(3): e0229322, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32176696

RESUMEN

Tetradecylthioacetic acid (TTA) is a synthetic fatty acid with a sulfur substitution in the ß-position. This modification renders TTA unable to undergo complete ß-oxidation and increases its biological activity, including activation of peroxisome proliferator activated receptors (PPARs) with preference for PPARα. This study investigated the effects of TTA on lipid and lipoprotein metabolism in the intestine and liver of mice fed a high fat diet (HFD). Mice receiving HFD supplemented with 0.75% (w/w) TTA had significantly lower body weights compared to mice fed the diet without TTA. Plasma triacylglycerol (TAG) was reduced 3-fold with TTA treatment, concurrent with increase in liver TAG. Total cholesterol was unchanged in plasma and liver. However, TTA promoted a shift in the plasma lipoprotein fractions with an increase in larger HDL particles. Histological analysis of the small intestine revealed a reduced size of lipid droplets in enterocytes of TTA treated mice, accompanied by increased mRNA expression of fatty acid transporter genes. Expression of the cholesterol efflux pump Abca1 was induced in the small intestine, but not in the liver. Scd1 displayed markedly increased mRNA and protein expression in the intestine of the TTA group. It is concluded that TTA treatment of HFD fed mice leads to increased expression of genes involved in uptake and transport of fatty acids and HDL cholesterol in the small intestine with concomitant changes in the plasma profile of smaller lipoproteins.


Asunto(s)
HDL-Colesterol/sangre , Dieta Alta en Grasa/efectos adversos , Lipoproteínas/metabolismo , PPAR alfa/agonistas , Sulfuros/administración & dosificación , Transportador 1 de Casete de Unión a ATP/genética , Transportador 1 de Casete de Unión a ATP/metabolismo , Animales , Peso Corporal/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Estearoil-CoA Desaturasa/genética , Estearoil-CoA Desaturasa/metabolismo , Sulfuros/farmacología , Triglicéridos/sangre
3.
Nat Commun ; 9(1): 1420, 2018 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-29650963

RESUMEN

The chronic skin inflammation psoriasis is crucially dependent on the IL-23/IL-17 cytokine axis. Although IL-23 is expressed by psoriatic keratinocytes and immune cells, only the immune cell-derived IL-23 is believed to be disease relevant. Here we use a genetic mouse model to show that keratinocyte-produced IL-23 is sufficient to cause a chronic skin inflammation with an IL-17 profile. Furthermore, we reveal a cell-autonomous nuclear function for the actin polymerizing molecule N-WASP, which controls IL-23 expression in keratinocytes by regulating the degradation of the histone methyltransferases G9a and GLP, and H3K9 dimethylation of the IL-23 promoter. This mechanism mediates the induction of IL-23 by TNF, a known inducer of IL-23 in psoriasis. Finally, in keratinocytes of psoriatic lesions a decrease in H3K9 dimethylation correlates with increased IL-23 expression, suggesting relevance for disease. Taken together, our data describe a molecular pathway where epigenetic regulation of keratinocytes can contribute to chronic skin inflammation.


Asunto(s)
Epigénesis Genética , N-Metiltransferasa de Histona-Lisina/genética , Subunidad p19 de la Interleucina-23/genética , Psoriasis/genética , Piel/metabolismo , Proteína Neuronal del Síndrome de Wiskott-Aldrich/genética , Adulto , Animales , Modelos Animales de Enfermedad , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Células HEK293 , N-Metiltransferasa de Histona-Lisina/deficiencia , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Inflamación , Interleucina-17/genética , Interleucina-17/metabolismo , Subunidad p19 de la Interleucina-23/deficiencia , Queratinocitos/metabolismo , Queratinocitos/patología , Masculino , Ratones , Ratones Noqueados , Persona de Mediana Edad , Cultivo Primario de Células , Regiones Promotoras Genéticas , Psoriasis/metabolismo , Psoriasis/patología , Transducción de Señal , Piel/patología , Proteína Neuronal del Síndrome de Wiskott-Aldrich/deficiencia
5.
Nat Cell Biol ; 18(12): 1357-1366, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27820601

RESUMEN

DNA double-strand breaks (DSBs) are highly cytotoxic DNA lesions, whose accurate repair by non-homologous end-joining (NHEJ) or homologous recombination (HR) is crucial for genome integrity and is strongly influenced by the local chromatin environment. Here, we identify SCAI (suppressor of cancer cell invasion) as a 53BP1-interacting chromatin-associated protein that promotes the functionality of several DSB repair pathways in mammalian cells. SCAI undergoes prominent enrichment at DSB sites through dual mechanisms involving 53BP1-dependent recruitment to DSB-surrounding chromatin and 53BP1-independent accumulation at resected DSBs. Cells lacking SCAI display reduced DSB repair capacity, hypersensitivity to DSB-inflicting agents and genome instability. We demonstrate that SCAI is a mediator of 53BP1-dependent repair of heterochromatin-associated DSBs, facilitating ATM kinase signalling at DSBs in repressive chromatin environments. Moreover, we establish an important role of SCAI in meiotic recombination, as SCAI deficiency in mice leads to germ cell loss and subfertility associated with impaired retention of the DMC1 recombinase on meiotic chromosomes. Collectively, our findings uncover SCAI as a physiologically important component of both NHEJ- and HR-mediated pathways that potentiates DSB repair efficiency in specific chromatin contexts.


Asunto(s)
Cromosomas de los Mamíferos/metabolismo , Roturas del ADN de Doble Cadena , Reparación del ADN , Factores de Transcripción/metabolismo , Proteína 1 de Unión al Supresor Tumoral P53/metabolismo , Animales , Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Línea Celular , Línea Celular Transformada , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Células Germinativas/citología , Células Germinativas/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Heterocromatina/metabolismo , Recombinación Homóloga/genética , Humanos , Meiosis , Ratones , Unión Proteica , Transducción de Señal , Xenopus
6.
PLoS One ; 11(11): e0166353, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27832170

RESUMEN

Automated blood sampling through a vascular catheter is a frequently utilized technique in laboratory mice. The potential immunological and physiological implications associated with this technique have, however, not been investigated in detail. The present study compared plasma levels of the cytokines IL-1ß, IL-2, IL-6, IL-10, IL-17A, GM-CSF, IFN-γ and TNF-α in male NMRI mice that had been subjected to carotid artery catheterization and subsequent automated blood sampling with age-matched control mice. Body weight and histopathological changes in the surgical area, including the salivary glands, the heart, brain, spleen, liver, kidneys and lungs were compared. Catheterized mice had higher levels of IL-6 than did control mice, but other cytokine levels did not differ between the groups. No significant difference in body weight was found. The histology revealed inflammatory and regenerative (healing) changes at surgical sites of all catheterized mice, with mild inflammatory changes extending into the salivary glands. Several catheterized mice had multifocal degenerative to necrotic changes with inflammation in the heart, kidneys and livers, suggesting that thrombi had detached from the catheter tip and embolized to distant sites. Thus, catheterization and subsequent automated blood sampling may have physiological impact. Possible confounding effects of visceral damage should be assessed and considered, when using catheterized mouse models.


Asunto(s)
Recolección de Muestras de Sangre/efectos adversos , Cateterismo/efectos adversos , Inflamación/etiología , Interleucina-6/sangre , Ratones/inmunología , Animales , Recolección de Muestras de Sangre/métodos , Arterias Carótidas/inmunología , Arterias Carótidas/patología , Cateterismo/métodos , Factor Estimulante de Colonias de Granulocitos y Macrófagos/sangre , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Inflamación/sangre , Inflamación/inmunología , Inflamación/patología , Interferón gamma/sangre , Interferón gamma/inmunología , Interleucina-6/inmunología , Riñón/inmunología , Riñón/patología , Hígado/inmunología , Hígado/patología , Masculino , Ratones Endogámicos , Miocardio/inmunología , Miocardio/patología , Glándulas Salivales/inmunología , Glándulas Salivales/patología , Factor de Necrosis Tumoral alfa/sangre , Factor de Necrosis Tumoral alfa/inmunología
7.
PLoS One ; 11(7): e0159850, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27441847

RESUMEN

Here we characterize a new animal model that spontaneously develops chronic inflammation and fibrosis in multiple organs, the non-obese diabetic inflammation and fibrosis (N-IF) mouse. In the liver, the N-IF mouse displays inflammation and fibrosis particularly evident around portal tracts and central veins and accompanied with evidence of abnormal intrahepatic bile ducts. The extensive cellular infiltration consists mainly of macrophages, granulocytes, particularly eosinophils, and mast cells. This inflammatory syndrome is mediated by a transgenic population of natural killer T cells (NKT) induced in an immunodeficient NOD genetic background. The disease is transferrable to immunodeficient recipients, while polyclonal T cells from unaffected syngeneic donors can inhibit the disease phenotype. Because of the fibrotic component, early on-set, spontaneous nature and reproducibility, this novel mouse model provides a unique tool to gain further insight into the underlying mechanisms mediating transformation of chronic inflammation into fibrosis and to evaluate intervention protocols for treating conditions of fibrotic disorders.


Asunto(s)
Hepatitis Crónica/etiología , Hepatitis Crónica/patología , Cirrosis Hepática/etiología , Cirrosis Hepática/patología , Traslado Adoptivo , Animales , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Biomarcadores , Citocinas/metabolismo , Modelos Animales de Enfermedad , Hepatitis Crónica/metabolismo , Mediadores de Inflamación/metabolismo , Cirrosis Hepática/metabolismo , Activación de Linfocitos/inmunología , Ratones , Células T Asesinas Naturales/inmunología , Células T Asesinas Naturales/metabolismo , Fenotipo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
8.
Stem Cells Dev ; 24(21): 2547-60, 2015 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-26176320

RESUMEN

The hedgehog (Hh) signaling pathway plays fundamental roles during embryonic development and tumorigenesis. Previously, we have shown that ablation of the tumor suppressor and negative regulator, Suppressor of fused (Sufu), within this pathway causes embryonic lethality around E9.5 in the mouse. In this study, we examine how lack of Sufu influences early cell fate determination processes. We established embryonic stem cell (ESC) lines from preimplantation Sufu(-/-) and wild-type mouse embryos and show that these ESCs express the typical pluripotency markers, alkaline phosphatase, SSEA-1, Oct4, Sox2, and Nanog. We demonstrate that these ESCs express all core Hh pathway components and that glioma-associated protein (Gli)1 mRNA levels are increased in Sufu(-/-) ESCs. Upon spontaneous differentiation of Sufu(-/-) ESCs into embryoid bodies (EBs) in vitro, the Hh pathway is strongly upregulated as indicated by an increase in both Gli1 and patched1 (Ptch1) gene expression. Interestingly, developing Sufu(-/-) EBs were smaller than their wild-type counterparts and showed decreased expression of the ectodermal markers, Fgf5 and Sox1. In vivo teratoma formation revealed that Sufu(-/-) ESCs have a limited capacity for differentiation as the resulting tumors lacked the mesodermal derivatives, cartilage and bone. However, Sufu(-/-) ESCs were able to develop into chondrocytes and osteocytes in vitro, which suggests a differential response of ESCs compared with in vivo conditions. Our findings suggest a regulatory function of the Hh signaling pathway in early mesodermal cell fate determination and emphasize the role of Sufu as a key molecule in this process.


Asunto(s)
Diferenciación Celular/fisiología , Transformación Celular Neoplásica/metabolismo , Desarrollo Embrionario/fisiología , Células Madre Embrionarias/citología , Proteínas Hedgehog/metabolismo , Proteínas Represoras/metabolismo , Animales , Diferenciación Celular/genética , Transformación Celular Neoplásica/genética , Desarrollo Embrionario/genética , Genes Supresores de Tumor/fisiología , Ratones Noqueados , Ratones Transgénicos , Transducción de Señal/genética
9.
FASEB J ; 29(8): 3193-205, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-25877214

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a rare premature aging disorder that is most commonly caused by a de novo point mutation in exon 11 of the LMNA gene, c.1824C>T, which results in an increased production of a truncated form of lamin A known as progerin. In this study, we used a mouse model to study the possibility of recovering from HGPS bone disease upon silencing of the HGPS mutation, and the potential benefits from treatment with resveratrol. We show that complete silencing of the transgenic expression of progerin normalized bone morphology and mineralization already after 7 weeks. The improvements included lower frequencies of rib fractures and callus formation, an increased number of osteocytes in remodeled bone, and normalized dentinogenesis. The beneficial effects from resveratrol treatment were less significant and to a large extent similar to mice treated with sucrose alone. However, the reversal of the dental phenotype of overgrown and laterally displaced lower incisors in HGPS mice could be attributed to resveratrol. Our results indicate that the HGPS bone defects were reversible upon suppressed transgenic expression and suggest that treatments targeting aberrant progerin splicing give hope to patients who are affected by HGPS.


Asunto(s)
Huesos/efectos de los fármacos , Silenciador del Gen/fisiología , Mutación/genética , Progeria/tratamiento farmacológico , Progeria/genética , Estilbenos/farmacología , Transgenes/genética , Animales , Huesos/metabolismo , Femenino , Lamina Tipo A/metabolismo , Masculino , Ratones , Osteocitos/efectos de los fármacos , Osteocitos/metabolismo , Fenotipo , Progeria/metabolismo , Resveratrol
10.
Hum Mol Genet ; 24(5): 1305-21, 2015 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-25343989

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a segmental progeroid syndrome with multiple features suggestive of premature accelerated aging. Accumulation of progerin is thought to underlie the pathophysiology of HGPS. However, despite ubiquitous expression of lamin A in all differentiated cells, the HGPS mutation results in organ-specific defects. For example, bone and skin are strongly affected by HGPS, while the brain appears to be unaffected. There are no definite explanations as to the variable sensitivity to progeria disease among different organs. In addition, low levels of progerin have also been found in several tissues from normal individuals, but it is not clear if low levels of progerin contribute to the aging of the brain. In an attempt to clarify the origin of this phenomenon, we have developed an inducible transgenic mouse model with expression of the most common HGPS mutation in brain, skin, bone and heart to investigate how the mutation affects these organs. Ultrastructural analysis of neuronal nuclei after 70 weeks of expression of the LMNA c.1824C>T mutation showed severe distortion with multiple lobulations and irregular extensions. Despite severe distortions in the nuclei of hippocampal neurons of HGPS animals, there were only negligible changes in gene expression after 63 weeks of transgenic expression. Behavioral analysis and neurogenesis assays, following long-term expression of the HGPS mutation, did not reveal significant pathology. Our results suggest that certain tissues are protected from functional deleterious effects of progerin.


Asunto(s)
Envejecimiento/genética , Regulación de la Expresión Génica , Hipocampo/metabolismo , Lamina Tipo A/metabolismo , Células Madre/metabolismo , Envejecimiento Prematuro/genética , Animales , Diferenciación Celular , Femenino , Procesamiento de Imagen Asistido por Computador , Lamina Tipo A/genética , Lamina Tipo B/genética , Lamina Tipo B/metabolismo , Masculino , Ratones , Ratones Transgénicos , Neurogénesis , Neuronas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
11.
PLoS One ; 9(11): e113225, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25426941

RESUMEN

Retro-bulbar sinus puncture and facial vein phlebotomy are two widely used methods for blood sampling in laboratory mice. However, the animal welfare implications associated with these techniques are currently debated, and the possible physiological and pathological implications of blood sampling using these methods have been sparsely investigated. Therefore, this study was conducted to assess and compare the impacts of blood sampling by retro-bulbar sinus puncture and facial vein phlebotomy. Blood was obtained from either the retro-bulbar sinus or the facial vein from male C57BL/6J mice at two time points, and the samples were analyzed for plasma corticosterone. Body weights were measured at the day of blood sampling and the day after blood sampling, and the food consumption was recorded automatically during the 24 hours post-procedure. At the end of study, cheeks and orbital regions were collected for histopathological analysis to assess the degree of tissue trauma. Mice subjected to facial vein phlebotomy had significantly elevated plasma corticosterone levels at both time points in contrast to mice subjected to retro-bulbar sinus puncture, which did not. Both groups of sampled mice lost weight following blood sampling, but the body weight loss was higher in mice subjected to facial vein phlebotomy. The food consumption was not significantly different between the two groups. At gross necropsy, subcutaneous hematomas were found in both groups and the histopathological analyses revealed extensive tissue trauma after both facial vein phlebotomy and retro-bulbar sinus puncture. This study demonstrates that both blood sampling methods have a considerable impact on the animals' physiological condition, which should be considered whenever blood samples are obtained.


Asunto(s)
Bienestar del Animal/ética , Flebotomía/métodos , Estrés Psicológico , Animales , Peso Corporal , Corticosterona/sangre , Ingestión de Alimentos , Venas Yugulares , Masculino , Ratones , Ratones Endogámicos C57BL , Órbita/irrigación sanguínea , Flebotomía/psicología , Punciones/psicología
12.
PLoS One ; 9(10): e109854, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25310607

RESUMEN

Obesity is a global epidemic that contributes to the increasing medical burdens related to type 2 diabetes, cardiovascular disease and cancer. A better understanding of the mechanisms regulating adipose tissue expansion could lead to therapeutics that eliminate or reduce obesity-associated morbidity and mortality. The extracellular matrix (ECM) has been shown to regulate the development and function of numerous tissues and organs. However, there is little understanding of its function in adipose tissue. In this manuscript we describe the role of laminin α4, a specialized ECM protein surrounding adipocytes, on weight gain and adipose tissue function. Adipose tissue accumulation, lipogenesis, and structure were examined in mice with a null mutation of the laminin α4 gene (Lama4-/-) and compared to wild-type (Lama4+/+) control animals. Lama4-/- mice exhibited reduced weight gain in response to both age and high fat diet. Interestingly, the mice had decreased adipose tissue mass and altered lipogenesis in a depot-specific manner. In particular, epididymal adipose tissue mass was specifically decreased in knock-out mice, and there was also a defect in lipogenesis in this depot as well. In contrast, no such differences were observed in subcutaneous adipose tissue at 14 weeks. The results suggest that laminin α4 influences adipose tissue structure and function in a depot-specific manner. Alterations in laminin composition offers insight into the roll the ECM potentially plays in modulating cellular behavior in adipose tissue expansion.


Asunto(s)
Tejido Adiposo/patología , Laminina/deficiencia , Aumento de Peso , Adipocitos/patología , Tejido Adiposo Blanco/patología , Envejecimiento/patología , Animales , Tamaño de la Célula , Dieta Alta en Grasa , Conducta Alimentaria , Laminina/metabolismo , Lipogénesis , Masculino , Ratones Endogámicos C57BL , Obesidad/patología , Grasa Subcutánea/patología
13.
PLoS One ; 8(10): e76330, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24124545

RESUMEN

Multiple Sclerosis (MS) is a progressive autoimmune inflammatory and demyelinating disease of the central nervous system (CNS). T cells play a key role in the progression of neuroinflammation in MS and also in the experimental autoimmune encephalomyelitis (EAE) animal models for the disease. A technology for quantitative and 3 dimensional (3D) spatial assessment of inflammation in this and other CNS inflammatory conditions is much needed. Here we present a procedure for 3D spatial assessment and global quantification of the development of neuroinflammation based on Optical Projection Tomography (OPT). Applying this approach to the analysis of rodent models of MS, we provide global quantitative data of the major inflammatory component as a function of the clinical course. Our data demonstrates a strong correlation between the development and progression of neuroinflammation and clinical disease in several mouse and a rat model of MS refining the information regarding the spatial dynamics of the inflammatory component in EAE. This method provides a powerful tool to investigate the effect of environmental and genetic forces and for assessing the therapeutic effects of drug therapy in animal models of MS and other neuroinflammatory/neurodegenerative disorders.


Asunto(s)
Imagenología Tridimensional/métodos , Esclerosis Múltiple/diagnóstico , Tomografía Óptica/métodos , Animales , Sistema Nervioso Central/inmunología , Sistema Nervioso Central/patología , Enfermedades Desmielinizantes/diagnóstico , Enfermedades Desmielinizantes/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/diagnóstico , Encefalomielitis Autoinmune Experimental/inmunología , Inflamación/diagnóstico , Inflamación/inmunología , Ratones , Esclerosis Múltiple/inmunología , Ratas , Subgrupos de Linfocitos T/metabolismo
14.
J Biol Chem ; 287(40): 33512-22, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22893709

RESUMEN

Hutchinson-Gilford progeria syndrome (HGPS) is a very rare genetic disorder that is characterized by multiple features of premature aging and largely affects tissues of mesenchymal origin. In this study, we describe the development of a tissue-specific mouse model that overexpresses the most common HGPS mutation (LMNA, c.1824C>T, p.G608G) in osteoblasts. Already at the age of 5 weeks, HGPS mutant mice show growth retardation, imbalanced gait and spontaneous fractures. Histopathological examination revealed an irregular bone structure, characterized by widespread loss of osteocytes, defects in mineralization, and a hypocellular red bone marrow. Computerized tomography analysis demonstrated impaired skeletal geometry and altered bone structure. The skeletal defects, which resemble the clinical features reported for bone disease in HGPS patients, was associated with an abnormal osteoblast differentiation. The osteoblast-specific expression of the HGPS mutation increased DNA damage and affected Wnt signaling. In the teeth, irregular dentin formation, as was previously demonstrated in human progeria cases, caused severe dental abnormalities affecting the incisors. The observed phenotype also shows similarities to reported bone abnormalities in aging mice and may therefore help to uncover general principles of the aging process.


Asunto(s)
Mutación , Osteoblastos/metabolismo , Progeria/genética , Progeria/metabolismo , Envejecimiento , Animales , Fenómenos Biomecánicos , Huesos/metabolismo , Daño del ADN , Femenino , Humanos , Lamina Tipo A , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Osteoblastos/citología , Osteocitos/citología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Transducción de Señal , Proteínas Wnt/metabolismo
15.
Neurobiol Dis ; 46(1): 41-51, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22405424

RESUMEN

Neuronal disorders, like Huntington's disease (HD), are difficult to study, due to limited cell accessibility, late onset manifestations, and low availability of material. The establishment of an in vitro model that recapitulates features of the disease may help understanding the cellular and molecular events that trigger disease manifestations. Here, we describe the generation and characterization of a series of induced pluripotent stem (iPS) cells derived from patients with HD, including two rare homozygous genotypes and one heterozygous genotype. We used lentiviral technology to transfer key genes for inducing reprogramming. To confirm pluripotency and differentiation of iPS cells, we used PCR amplification and immunocytochemistry to measure the expression of marker genes in embryoid bodies and neurons. We also analyzed teratomas that formed in iPS cell-injected mice. We found that the length of the pathological CAG repeat did not increase during reprogramming, after long term growth in vitro, and after differentiation into neurons. In addition, we observed no differences between normal and mutant genotypes in reprogramming, growth rate, caspase activation or neuronal differentiation. However, we observed a significant increase in lysosomal activity in HD-iPS cells compared to control iPS cells, both during self-renewal and in iPS-derived neurons. In conclusion, we have established stable HD-iPS cell lines that can be used for investigating disease mechanisms that underlie HD. The CAG stability and lysosomal activity represent novel observations in HD-iPS cells. In the future, these cells may provide the basis for a powerful platform for drug screening and target identification in HD.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Lisosomas/genética , Proteínas del Tejido Nervioso/genética , Células Madre Pluripotentes/metabolismo , Animales , Línea Celular , Fibroblastos/citología , Fibroblastos/fisiología , Heterocigoto , Homocigoto , Humanos , Proteína Huntingtina , Enfermedad de Huntington/patología , Lisosomas/metabolismo , Ratones , Ratones SCID , Mutación , Proteínas del Tejido Nervioso/metabolismo , Fenotipo , Teratoma/genética , Teratoma/metabolismo , Activación Transcripcional/fisiología
16.
PLoS One ; 7(2): e31366, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22393362

RESUMEN

Myc plays an important role in tumor development, including acute myeloid leukemia (AML). However, MYC is also a powerful inducer of apoptosis, which is one of the major failsafe programs to prevent cancer development. To clarify the relative importance of the extrinsic (death receptor-mediated) versus the intrinsic (mitochondrial) pathway of apoptosis in MYC-driven AML, we coexpressed MYC together with anti-apoptotic proteins of relevance for AML; BCL-X(L)/BCL-2 (inhibiting the intrinsic pathway) or FLIP(L) (inhibiting the extrinsic pathway), in hematopoietic stems cells (HSCs). Transplantation of HSCs expressing MYC into syngeneic recipient mice resulted in development of AML and T-cell lymphomas within 7-9 weeks as expected. Importantly, coexpression of MYC together with BCL-X(L)/BCL-2 resulted in strongly accelerated kinetics and favored tumor development towards aggressive AML. In contrast, coexpression of MYC and FLIP(L) did neither accelerate tumorigenesis nor change the ratio of AML versus T-cell lymphoma. However, a change in distribution of immature CD4(+)CD8(+) versus mature CD4(+) T-cell lymphoma was observed in MYC/FLIP(L) mice, possibly as a result of increased survival of the CD4+ population, but this did not significantly affect the outcome of the disease. In conclusion, our findings provide direct evidence that BCL-X(L) and BCL-2 but not FLIP(L) acts in synergy with MYC to drive AML development.


Asunto(s)
Apoptosis , Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Animales , Proteínas Bacterianas/metabolismo , Proteína Reguladora de Apoptosis Similar a CASP8 y FADD/metabolismo , Línea Celular Tumoral , Transformación Celular Neoplásica , Femenino , Proteínas Fluorescentes Verdes/metabolismo , Células Madre Hematopoyéticas/citología , Humanos , Cinética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos DBA , Modelos Biológicos , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteína bcl-X/metabolismo
17.
Mol Carcinog ; 51(9): 754-60, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21882258

RESUMEN

Basal cell carcinoma of the skin typically carries genetic alterations in components of the hedgehog (HH) signaling pathway. Previously, we generated a knockout mouse with a loss-of-function mutation in suppressor of fused (Sufu), an essential repressor of the pathway downstream of Hh ligand cell surface reception. Mice heterozygous for the mutated Sufu allele develop a skin phenotype that includes lesions similar to basaloid follicular hamartomas. The purpose of the current study was to test the possibility that the simultaneous loss of the tumor suppressor gene, transformation related protein 53 (Trp53), would aggravate the Sufu skin phenotype since Trp53 loss is known to enhance the growth of other Hh-driven tumors. Consistent with previous reports, medulloblastomas and rhabdomyosarcomas developed in Sufu(+/-) ;Trp53(-/-) mice. However, the characteristic Sufu(+/-) skin phenotype was not altered in the absence of Trp53, and showed no changes in latency, multiplicity, cellular phenotype, or proliferative capacity of the basaloid lesions. This finding was both novel and intriguing and demonstrated a differential, tissue-specific sensitivity to Sufu and Trp53 tumor suppressor gene loss, which may be linked to developmental stage and the degree of proliferative activity in specific cell types.


Asunto(s)
Neoplasias Cerebelosas/etiología , Neoplasias Cerebelosas/patología , Meduloblastoma/etiología , Meduloblastoma/patología , Proteínas Represoras/fisiología , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/patología , Proteína p53 Supresora de Tumor/fisiología , Animales , Neoplasias Cerebelosas/metabolismo , Femenino , Heterocigoto , Técnicas para Inmunoenzimas , Linfoma/etiología , Linfoma/metabolismo , Linfoma/patología , Masculino , Meduloblastoma/metabolismo , Ratones , Ratones Noqueados , Neoplasias Cutáneas/metabolismo , Tasa de Supervivencia
18.
Am J Pathol ; 179(5): 2569-79, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21933656

RESUMEN

Hedgehog (Hh) signaling is a regulator of salivary gland morphogenesis, but its role in postnatal glands has only recently begun to be addressed. To examine the effects of deregulated Hh signaling in the salivary gland, we expressed the Hh effector protein GLI1, in salivary epithelial cells using both cytokeratin 5 and mouse mammary tumor virus (MMTV) transgenic systems. Ectopic pathway activation resulted in restrained acinar differentiation, formation of cystic lesions, and prominent appearance of ductal structures. Moreover, induced expression of GLI1 aids the formation of hyperplastic lesions, which closely resemble GLI1-induced changes in murine skin and mammary glands, suggesting that GLI1 targets cells with similar characteristics in different tissues. Furthermore, GLI1-expressing salivary epithelial cells are actively dividing, and GLI1-induced lesions are proliferative, an incident accompanied by enhanced expression of the Hh target genes, cyclin D1, and Snail. GLI1-induced salivary lesions regress after transgene withdrawal and become histologically normalized. Taken together, our data reveal the ability of GLI1 to modulate salivary acinar differentiation and to promote proliferation of ductal epithelial cells.


Asunto(s)
Proteínas Hedgehog/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Glándulas Salivales/metabolismo , Células Acinares/patología , Animales , Diferenciación Celular/fisiología , Proliferación Celular , Células Epiteliales/metabolismo , Femenino , Hiperplasia/metabolismo , Hiperplasia/patología , Factores de Transcripción de Tipo Kruppel/fisiología , Masculino , Ratones , Ratones Transgénicos , Glándulas Salivales/patología , Transducción de Señal/fisiología , Proteína con Dedos de Zinc GLI1
19.
Int J Clin Exp Pathol ; 4(6): 596-605, 2011 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-21904635

RESUMEN

Hematopoietic stem cell transplantation (HSCT) is a curative treatment for otherwise incurable diseases. Conditioning regimen is an important part of HSCT and consists of chemotherapy with or without irradiation. Conditioning exerts myelosuppressive, immunosuppressive and antitumor effects, but also contributes to HSCT-related complications including graft-versus-host disease (GVHD). Since almost 50% of the transplanted patients are conditioned with cytostatics without irradiation, we developed and characterized a GVHD mouse model following conditioning with busulphan and cyclophosphamide. Recipient Balb/c female mice were treated with busulphan (20 mg/kg/day for 4 days) and cyclophosphamide (100 mg/kg/day for two days). After one day of rest, recipient mice were transplanted with 2×10(7) bone marrow and 3×10(7) spleen cells from male C57BL/6 (allogeneic group) or female Balb/c (syngeneic/control group) mice. The allogeneic, but not syngeneic transplanted mice developed GVHD. Histopathology of the major internal organs (liver, pancreas, spleen, lungs, heart and kidney) was examined before conditioning start, after conditioning's end and 5, 7 and 21 days after transplantation using hematoxylin-eosin staining. Decreased spleen cellularity and diminished glycogen content in the liver were observed after conditioning regimen. Histopathological changes such as vasculitis, inflammation and apoptotic cell forms in liver, spleen, pancreas, lungs and heart were observed in allogeneic transplanted mice, however, only hypocellular spleen and extramedullar hematopoiesis were detected in syngeneic transplanted animals. No morphological changes were observed in kidney in either HSCT setting. This is the first study describing early histopathological changes after conditioning regimen with busulphan/cyclophosphamide and dynamics of GVHD development in several major internal organs.


Asunto(s)
Busulfano/farmacología , Ciclofosfamida/farmacología , Enfermedad Injerto contra Huésped/patología , Agonistas Mieloablativos/farmacología , Acondicionamiento Pretrasplante/métodos , Animales , Apoptosis/efectos de los fármacos , Trasplante de Médula Ósea , Trasplante de Células , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto , Hematopoyesis Extramedular/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Hígado/patología , Glucógeno Hepático/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Páncreas/efectos de los fármacos , Páncreas/patología , Bazo/citología , Pérdida de Peso/efectos de los fármacos
20.
Biochem Biophys Res Commun ; 412(1): 121-6, 2011 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-21802407

RESUMEN

Glutathione (GSH) is present in all mammalian tissues and plays a crucial role in many cellular processes. The second and final step in the synthesis involves the formation of GSH from gamma-glutamylcysteine (γ-GC) and glycine and is catalyzed by glutathione synthetase (GS). GS deficiency is a rare autosomal recessive disorder, and is present in patients with a range of phenotypes, from mild hemolytic anemia and metabolic acidosis to severe neurologic disorders or even death in infancy. The substrate for GS, γ-GC, has been suggested as playing a protective role, by substituting for GSH as an antioxidant in GS deficient patients. To examine the role of GS and GSH metabolites in development, we generated mice deficient in GSH by targeted disruption of the GS gene (Gss). Homozygous mice died before embryonic day (E) 7.5, but heterozygous mice survived with no distinct phenotype. GS protein levels and enzyme activity, as well as GSH metabolites, were investigated in multiple tissues. Protein levels and enzyme activity of GS in heterozygous mice were diminished by 50%, while GSH levels remained intact. γ-GC could not be detected in any investigated tissue. These data demonstrate that GSH is essential for mammalian development, and GSH synthesis via GS is an indispensable pathway for survival.


Asunto(s)
Desarrollo Embrionario/genética , Glutatión Sintasa/deficiencia , Glutatión Sintasa/fisiología , Glutatión/fisiología , Animales , Modelos Animales de Enfermedad , Glutatión/biosíntesis , Glutatión Sintasa/genética , Ratones , Ratones Noqueados , Estrés Oxidativo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...