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

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Mamm Genome ; 33(1): 203-212, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34313795

RESUMEN

The Mutant Mouse Resource and Research Center (MMRRC) Program is the pre-eminent public national mutant mouse repository and distribution archive in the USA, serving as a national resource of mutant mice available to the global scientific community for biomedical research. Established more than two decades ago with grants from the National Institutes of Health (NIH), the MMRRC Program supports a Consortium of regionally distributed and dedicated vivaria, laboratories, and offices (Centers) and an Informatics Coordination and Service Center (ICSC) at three academic teaching and research universities and one non-profit genetic research institution. The MMRRC Program accepts the submission of unique, scientifically rigorous, and experimentally valuable genetically altered and other mouse models donated by academic and commercial scientists and organizations for deposition, maintenance, preservation, and dissemination to scientists upon request. The four Centers maintain an archive of nearly 60,000 mutant alleles as live mice, frozen germplasm, and/or embryonic stem (ES) cells. Since its inception, the Centers have fulfilled 13,184 orders for mutant mouse models from 9591 scientists at 6626 institutions around the globe. Centers also provide numerous services that facilitate using mutant mouse models obtained from the MMRRC, including genetic assays, microbiome analysis, analytical phenotyping and pathology, cryorecovery, mouse husbandry, infectious disease surveillance and diagnosis, and disease modeling. The ICSC coordinates activities between the Centers, manages the website (mmrrc.org) and online catalog, and conducts communication, outreach, and education to the research community. Centers preserve, secure, and protect mutant mouse lines in perpetuity, promote rigor and reproducibility in scientific experiments using mice, provide experiential training and consultation in the responsible use of mice in research, and pursue cutting edge technologies to advance biomedical studies using mice to improve human health. Researchers benefit from an expansive list of well-defined mouse models of disease that meet the highest standards of rigor and reproducibility, while donating investigators benefit by having their mouse lines preserved, protected, and distributed in compliance with NIH policies.


Asunto(s)
Investigación Biomédica , Modelos Animales de Enfermedad , Ratones , National Institutes of Health (U.S.) , Animales , Humanos , Ratones/genética , Reproducibilidad de los Resultados , Estados Unidos
3.
Vet Pathol ; 58(1): 10-33, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33016246

RESUMEN

The peripheral nervous system (PNS) relays messages between the central nervous system (brain and spinal cord) and the body. Despite this critical role and widespread distribution, the PNS is often overlooked when investigating disease in diagnostic and experimental pathology. This review highlights key features of neuroanatomy and physiology of the somatic and autonomic PNS, and appropriate PNS sampling and processing techniques. The review considers major classes of PNS lesions including neuronopathy, axonopathy, and myelinopathy, and major categories of PNS disease including toxic, metabolic, and paraneoplastic neuropathies; infectious and inflammatory diseases; and neoplasms. This review describes a broad range of common PNS lesions and their diagnostic criteria and provides many useful references for pathologists who perform PNS evaluations as a regular or occasional task in their comparative pathology practice.


Asunto(s)
Enfermedades del Sistema Nervioso Central , Enfermedades del Sistema Nervioso Periférico , Animales , Sistema Nervioso Central , Enfermedades del Sistema Nervioso Central/veterinaria , Sistema Nervioso Periférico , Enfermedades del Sistema Nervioso Periférico/diagnóstico , Enfermedades del Sistema Nervioso Periférico/veterinaria , Médula Espinal
4.
Mol Microbiol ; 100(1): 90-107, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26712165

RESUMEN

Mycobacterium tuberculosis proteins that are exported out of the bacterial cytoplasm are ideally positioned to be virulence factors; however, the functions of individual exported proteins remain largely unknown. Previous studies identified Rv0199 as an exported membrane protein of unknown function. Here, we characterized the role of Rv0199 in M. tuberculosis virulence using an aerosol model of murine infection. Rv0199 appears to be a member of a Mce-associated membrane (Mam) protein family leading us to rename it OmamA, for orphaned Mam protein A. Consistent with a role in Mce transport, we showed OmamA is required for cholesterol import, which is a Mce4-dependent process. We further demonstrated a function for OmamA in stabilizing protein components of the Mce1 transporter complex. These results indicate a function of OmamA in multiple Mce transporters and one that may be analogous to the role of VirB8 in stabilizing Type IV secretion systems, as structural similarities between Mam proteins and VirB8 proteins are predicted by the Phyre 2 program. In this study, we provide functional information about OmamA and shed light on the function of Mam family proteins in Mce transporters.


Asunto(s)
Proteínas Bacterianas/metabolismo , Proteínas de la Membrana/metabolismo , Mycobacterium tuberculosis/metabolismo , Animales , Proteínas Bacterianas/genética , Colesterol/metabolismo , Modelos Animales de Enfermedad , Eliminación de Gen , Orden Génico , Proteínas de la Membrana/genética , Ratones , Mutación , Mycobacterium tuberculosis/genética , Fenotipo , Unión Proteica , Transporte de Proteínas , Tuberculosis/microbiología , Tuberculosis/mortalidad , Tuberculosis/patología , Factores de Virulencia
5.
Cancer Cell ; 12(4): 355-66, 2007 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-17936560

RESUMEN

It is believed that Mdm2 suppresses p53 in two ways: transcriptional inhibition by direct binding, and degradation via its E3 ligase activity. To study these functions physiologically, we generated mice bearing a single-residue substitution (C462A) abolishing the E3 function without affecting p53 binding. Unexpectedly, homozygous mutant mice died before E7.5, and deletion of p53 rescued the lethality. Furthermore, reintroducing a switchable p53 by crossing with p53ER(TAM) mice surprisingly demonstrated that the mutant Mdm2(C462A) was rapidly degraded in a manner indistinguishable from that of the wild-type Mdm2. Hence, our data indicate that (1) the Mdm2-p53 physical interaction, without Mdm2-mediated p53 ubiquitination, cannot control p53 activity sufficiently to allow early mouse embryonic development, and (2) Mdm2's E3 function is not required for Mdm2 degradation.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Mutagénesis Sitio-Dirigida , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Transcripción Genética , Proteína p53 Supresora de Tumor/metabolismo , Sustitución de Aminoácidos , Animales , Células Cultivadas , Daño del ADN , Regulación hacia Abajo , Embrión de Mamíferos , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibroblastos/efectos de la radiación , Rayos gamma , Regulación del Desarrollo de la Expresión Génica/efectos de la radiación , Genotipo , Edad Gestacional , Homocigoto , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas c-mdm2/química , Proteínas Proto-Oncogénicas c-mdm2/deficiencia , Proteínas Proto-Oncogénicas c-mdm2/genética , Transcripción Genética/efectos de la radiación , Proteína p53 Supresora de Tumor/deficiencia , Proteína p53 Supresora de Tumor/genética
6.
J Cell Physiol ; 229(1): 44-52, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23757284

RESUMEN

Widespread changes in gene expression underlie B cell development and activation, yet our knowledge of which chromatin-remodeling factors are essential is limited. Here, we demonstrate that the BRG1 catalytic subunit of SWI/SNF complexes was dispensable for murine B cell development but played an important, albeit selective, role during activation. Although BRG1 was dispensable for CD69 induction and differentiation into plasma cells based on the ability of mutant B cells to undergo hypertrophy and secrete IgM antibodies, it was required for robust cell proliferation in response to activation. Accordingly, BRG1 was required for only ∼100 genes to be expressed at normal levels in naïve B cells but >1,000 genes during their activation. BRG1 upregulated fivefold more genes than it downregulated, and the toll-like receptor pathway and JAK/STAT cytokine-signaling pathways were particularly dependent on BRG1. The importance of BRG1 in B cell activation was underscored by the occurrence of opportunistic Pasteurella infections in conditionally mutant mice. B cell activation has long served as a model of inducible gene expression, and the results presented here identify BRG1 as a chromatin-remodeling factor that upregulates the transcriptome of B cells during their activation to promote rapid cell proliferation and to mount an effective immune response.


Asunto(s)
Linfocitos B/metabolismo , Ensamble y Desensamble de Cromatina/genética , ADN Helicasas , Activación de Linfocitos/genética , Proteínas Nucleares , Factores de Transcripción , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Linfocitos B/fisiología , Diferenciación Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , ADN Helicasas/genética , ADN Helicasas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Ratones , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
7.
Mamm Genome ; 25(3-4): 95-108, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24487921

RESUMEN

Inflammatory bowel disease (IBD) is an immune-mediated condition driven by improper responses to intestinal microflora in the context of environmental and genetic background. GWAS in humans have identified many loci associated with IBD, but animal models are valuable for dissecting the underlying molecular mechanisms, characterizing environmental and genetic contributions and developing treatments. Mouse models rely on interventions such as chemical treatment or introduction of an infectious agent to induce disease. Here, we describe a new model for IBD in which the disease develops spontaneously in 20-week-old mice in the absence of known murine pathogens. The model is part of the Collaborative Cross and came to our attention due to a high incidence of rectal prolapse in an incompletely inbred line. Necropsies revealed a profound proliferative colitis with variable degrees of ulceration and vasculitis, splenomegaly and enlarged mesenteric lymph nodes with no discernible anomalies of other organ systems. Phenotypic characterization of the CC011/Unc mice with homozygosity ranging from 94.1 to 99.8% suggested that the trait was fixed and acted recessively in crosses to the colitis-resistant C57BL/6J inbred strain. Using a QTL approach, we identified four loci, Ccc1, Ccc2, Ccc3 and Ccc4 on chromosomes 12, 14, 1 and 8 that collectively explain 27.7% of the phenotypic variation. Surprisingly, we also found that minute levels of residual heterozygosity in CC011/Unc have significant impact on the phenotype. This work demonstrates the utility of the CC as a source of models of human disease that arises through new combinations of alleles at susceptibility loci.


Asunto(s)
Cruzamiento/métodos , Modelos Animales de Enfermedad , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/fisiopatología , Ratones Endogámicos/genética , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Cartilla de ADN/genética , Genotipo , Humanos , Ratones , Ratones Endogámicos C57BL , Linaje , Reacción en Cadena de la Polimerasa , Sitios de Carácter Cuantitativo/genética
8.
Circ Res ; 111(5): e111-22, 2012 Aug 17.
Artículo en Inglés | MEDLINE | ID: mdl-22740088

RESUMEN

RATIONALE: Mating type switching/sucrose non-fermenting (SWI/SNF) chromatin-remodeling complexes utilize either BRG1 or BRM as a catalytic subunit to alter nucleosome position and regulate gene expression. BRG1 is required for vascular endothelial cell (VEC) development and embryonic survival, whereas BRM is dispensable. OBJECTIVE: To circumvent embryonic lethality and study Brg1 function in adult tissues, we used conditional gene targeting. To evaluate possible Brg1-Brm redundancy, we analyzed Brg1 mutant mice on wild-type and Brm-deficient backgrounds. METHODS AND RESULTS: The inducible Mx1-Cre driver was used to mutate Brg1 in adult mice. These conditional-null mutants exhibited a tissue-specific phenotype and unanticipated functional compensation between Brg1 and Brm. Brg1 single mutants were healthy and had a normal lifespan, whereas Brg1/Brm double mutants exhibited cardiovascular defects and died within 1 month. BRG1 and BRM were required for the viability of VECs but not other cell types where both genes were also knocked out. The VEC phenotype was most evident in the heart, particularly in the microvasculature of the outer myocardium, and was recapitulated in primary cells ex vivo. VEC death resulted in vascular leakage, cardiac hemorrhage, secondary death of cardiomyocytes due to ischemia, and ventricular dissections. CONCLUSIONS: BRG1-catalyzed SWI/SNF complexes are particularly important in cardiovascular tissues. However, in contrast to embryonic development, in which Brm does not compensate, Brg1 is required in adult VECs only when Brm is also mutated. These results demonstrate for the first time that Brm functionally compensates for Brg1 in vivo and that there are significant changes in the relative importance of BRG1- and BRM-catalyzed SWI/SNF complexes during the development of an essential cell lineage.


Asunto(s)
Proteínas Cromosómicas no Histona/metabolismo , ADN Helicasas/metabolismo , Células Endoteliales/metabolismo , Cardiopatías Congénitas/metabolismo , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Factores de Edad , Animales , Catálisis , Muerte Celular/fisiología , Linaje de la Célula/fisiología , Supervivencia Celular/fisiología , Proteínas Cromosómicas no Histona/genética , Vasos Coronarios/embriología , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , ADN Helicasas/genética , Ecocardiografía , Células Endoteliales/patología , Corazón/embriología , Cardiopatías Congénitas/genética , Cardiopatías Congénitas/patología , Homeostasis/fisiología , Ratones , Ratones Transgénicos , Isquemia Miocárdica/genética , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/patología , Proteínas Nucleares/genética , Derrame Pleural/genética , Derrame Pleural/metabolismo , Derrame Pleural/patología , Factores de Transcripción/genética
9.
Sci Adv ; 8(4): eabl5838, 2022 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-35080986

RESUMEN

The therapeutic potential of CDK4/6 inhibitors for brain tumors has been limited by recurrence. To address recurrence, we tested a nanoparticle formulation of CDK4/6 inhibitor palbociclib (POx-Palbo) in mice genetically-engineered to develop SHH-driven medulloblastoma, alone or in combination with specific agents suggested by our analysis. Nanoparticle encapsulation reduced palbociclib toxicity, enabled parenteral administration, improved CNS pharmacokinetics, and extended mouse survival, but recurrence persisted. scRNA-seq identified up-regulation of glutamate transporter Slc1a2 and down-regulation of diverse ribosomal genes in proliferating medulloblastoma cells in POx-Palbo-treated mice, suggesting mTORC1 signaling suppression, subsequently confirmed by decreased 4EBP1 phosphorylation. Combining POx-Palbo with the mTORC1 inhibitor sapanisertib produced mutually enhancing effects and prolonged mouse survival compared to either agent alone, contrasting markedly with other tested drug combinations. Our data show the potential of nanoparticle formulation and scRNA-seq analysis of resistance to improve brain tumor treatment and identify POx-Palbo + Sapanisertib as effective combinatorial therapy for SHH medulloblastoma.

10.
J Exp Med ; 202(8): 1141-51, 2005 Oct 17.
Artículo en Inglés | MEDLINE | ID: mdl-16230479

RESUMEN

The Scurfy mutation of the FoxP3 gene (FoxP3(sf)) in the mouse and analogous mutations in human result in lethal autoimmunity. The mutation of FoxP3 in the hematopoietic cells impairs the development of regulatory T cells. In addition, development of the Scurfy disease also may require mutation of the gene in nonhematopoietic cells. The T cell-extrinsic function of FoxP3 has not been characterized. Here we show that the FoxP3(sf) mutation leads to defective thymopoiesis, which is caused by inactivation of FoxP3 in the thymic stromal cells. FoxP3 mutation also results in overexpression of ErbB2 in the thymic stroma, which may be involved in defective thymopoiesis. Our data reveal a novel T cell-extrinsic function of FoxP3. In combination, the T cell-intrinsic and -extrinsic defects provide plausible explanation for the severity of the autoimmune diseases in the scurfy mice and in patients who have immunodysregulation, polyendocrinopathy, enteropathy, and X-linked syndrome.


Asunto(s)
Enfermedades Autoinmunes/genética , Factores de Transcripción Forkhead/genética , Enfermedades Linfáticas/genética , Mutación/genética , Timo/crecimiento & desarrollo , Timo/patología , Animales , Apoptosis/fisiología , Enfermedades Autoinmunes/patología , Bromodesoxiuridina , Cartilla de ADN , Citometría de Flujo , Luciferasas , Enfermedades Linfáticas/patología , Ratones , Ratones Endogámicos BALB C , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células del Estroma/metabolismo , Timo/citología , Timo/metabolismo
11.
World Neurosurg ; 148: e242-e251, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33412322

RESUMEN

OBJECTIVE: Aneurysmal subarachnoid hemorrhage remains a devastating event with poorly understood pathophysiology. Previous studies have suggested that aneurysm wall inflammation may play a part in the development and potential rupture of aneurysms. The rabbit elastase aneurysm model is a well-established model, which produces aneurysms closely mimicking human cerebral aneurysms in flow dynamics and histopathology. The primary aim of this study was to correlate inflammatory changes after aneurysm formation using sequential vessel wall imaging with histopathologic analysis. A secondary aim was to evaluate the potential effect of gender and anti-inflammatory treatment with aspirin on this inflammatory response. METHODS: Twenty-seven New Zealand rabbits underwent surgery to create an aneurysm using elastase infusion at the right common carotid artery origin. Vessel wall imaging and histopathologic analysis was obtained at different time points after aneurysm creation. The rabbits were also randomized by gender and to treatment groups with or without aspirin. RESULTS: Histopathologic analysis revealed 3 distinct phases after aneurysm formation. These phases were an initial inflammatory phase, followed by a regeneration phase, and finally a connective tissue deposition phase. Vessel wall imaging demonstrated 2 distinct imaging patterns. No appreciable differences were seen in histology or imaging when comparing gender or treatment with aspirin. CONCLUSIONS: Inflammatory changes induced by the rabbit elastase aneurysm model can be correlated with histopathologic findings and observed on noninvasive vessel wall imaging. This may provide a method to study the inflammatory pathway as it pertains to aneurysmal development and subsequent rupture.


Asunto(s)
Enfermedades de las Arterias Carótidas/inducido químicamente , Modelos Animales de Enfermedad , Aneurisma Intracraneal/complicaciones , Angiografía por Resonancia Magnética , Elastasa Pancreática/toxicidad , Conejos/fisiología , Animales , Aspirina/uso terapéutico , Enfermedades de las Arterias Carótidas/diagnóstico por imagen , Enfermedades de las Arterias Carótidas/patología , Arteria Carótida Común/diagnóstico por imagen , Arteria Carótida Común/efectos de los fármacos , Arteria Carótida Común/patología , Arteria Carótida Común/fisiología , Progresión de la Enfermedad , Tejido Elástico/ultraestructura , Femenino , Hiperplasia , Infusiones Intraarteriales , Aneurisma Intracraneal/inducido químicamente , Aneurisma Intracraneal/diagnóstico por imagen , Aneurisma Intracraneal/tratamiento farmacológico , Masculino , Miocitos del Músculo Liso/patología , Necrosis , Elastasa Pancreática/administración & dosificación , Conejos/inmunología , Regeneración , Caracteres Sexuales , Método Simple Ciego , Túnica Íntima/patología , Túnica Media/patología , Vasculitis/tratamiento farmacológico , Vasculitis/etiología , Vasculitis/patología
12.
J Exp Med ; 198(12): 1937-49, 2003 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-14676303

RESUMEN

Mammalian SWI-SNF-related complexes use brahma-related gene 1 (Brg1) as a catalytic subunit to remodel nucleosomes and regulate transcription. Recent biochemical data has linked Brg1 function to genes important for T lymphocyte differentiation. To investigate the role of SWI-SNF-related complexes in this lineage, we ablated Brg1 function in T lymphocytes. T cell-specific Brg1-deficient mice showed profound thymic abnormalities, CD4 derepression at the double negative (DN; CD4- CD8-) stage, and a developmental block at the DN to double positive (CD4+ CD8+) transition. 5'-bromo-2'-deoxyuridine incorporation and annexin V staining establish a role for Brg1 complexes in the regulation of thymocyte cell proliferation and survival. This Brg1-dependent cell survival is specific for developing thymocytes as indicated by the presence of Brg1-deficient mature T lymphocytes that have escaped the developmental block in the thymus. However, reductions in peripheral T cell populations lead to immunodeficiency and compromised health of mutant mice. These results highlight the importance of chromatin-remodeling complexes at different stages in the development of a mammalian cell lineage.


Asunto(s)
Proteínas Nucleares/fisiología , Linfocitos T/fisiología , Factores de Transcripción/fisiología , Animales , Apoptosis , Antígenos CD4/análisis , Antígenos CD8/análisis , Proteínas de Ciclo Celular/análisis , ADN Helicasas , Proteínas de Drosophila , Infecciones por Helicobacter/complicaciones , Activación de Linfocitos , Ratones , Receptores de Antígenos de Linfocitos T gamma-delta/análisis , Prolapso Rectal/etiología , Timo/fisiología , Transactivadores/análisis
13.
Mol Carcinog ; 48(12): 1139-48, 2009 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-19676100

RESUMEN

Malignant rhabdoid tumors (MRTs) are poorly differentiated pediatric cancers that arise in various anatomical locations and have a very poor outcome. The large majority of these malignancies are caused by loss of function of the SNF5/INI1 component of the SWI/SNF chromatin remodeling complex. However, the mechanism of tumor development associated with SNF5 loss remains unclear. Multiple studies have demonstrated a role for SNF5 in the regulation of cyclin D1, p16(INK4A), and pRb(f) activities suggesting it functions through the SWI/SNF complex to affect transcription of genes involved in cell cycle control. Previous studies in genetically engineered mouse models (GEMM) have shown that loss of SNF5 on a p53-null background significantly accelerates tumor development. Here, we use established GEMM to further define the relationship between the SNF5 and p53 tumor suppressor pathways. Combined haploinsufficiency of p53 and Snf5 leads to decreased latency for MRTs arising in alternate anatomical locations but not for the standard facial MRTs. We also observed acceleration in the appearance of T-cell lymphomas in the p53(+/-);Snf5(+/-) mice. Our studies suggest that loss of SNF5 activity does not bestow a selective advantage on the p53 spectrum of tumors in the p53(+/-);Snf5(+/-) mice. However, reduced p53 expression specifically accelerated the growth of a subset of MRTs in these mice.


Asunto(s)
Neoplasias Óseas/patología , Proteínas Cromosómicas no Histona/fisiología , Linfoma de Células T/patología , Osteosarcoma/patología , Tumor Rabdoide/patología , Proteína p53 Supresora de Tumor/fisiología , Animales , Neoplasias Óseas/genética , Proteínas de Ciclo Celular/metabolismo , Femenino , Linfoma de Células T/genética , Masculino , Ratones , Ratones Noqueados , Osteosarcoma/genética , Tumor Rabdoide/genética , Proteína SMARCB1 , Tasa de Supervivencia
14.
Lab Anim (NY) ; 38(1): 24-34, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19112447

RESUMEN

The authors examined the effects of cage size and enrichment on mouse breeding performance and behavior. Breeding trios of C57BL/6Tac mice were housed in cages of two different sizes ('standard' and 'large' cages with 82 in(2) and 124 in(2) floor space, respectively). Half of the cages of each size contained four enrichment items (Nestlet, plastic tunnel, nylon rings and running wheel), whereas the remaining cages had no enrichment. The authors measured the following reproductive parameters: litter size, number of pups that survived to weaning age, average pup weights at 21 d after birth and number of days between births of litters. A subset of weaned male and female pups from each cage size and enrichment condition completed a suite of behavioral tests. Pups raised in large cages weighed less than those raised in standard cages. Enrichment and cage size had certain behavioral effects, which were dependent on gender and behavioral measure. Male pups born in enriched cages showed more anxiety-like behavior and less exploration than did males born in non-enriched cages. Though being raised in enriched or large cages did not clearly improve pups' performance in behavioral tests, enrichment (regardless of cage size) did significantly benefit reproductive performance; pups from non-enriched cages weighed less than pups from enriched cages, and fewer survived to weaning age.


Asunto(s)
Conducta Exploratoria/fisiología , Fertilidad/fisiología , Vivienda para Animales , Conducta Sexual Animal/fisiología , Análisis de Varianza , Animales , Ansiedad/patología , Peso Corporal , Femenino , Tamaño de la Camada/fisiología , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Ratones Endogámicos C57BL , Prueba de Desempeño de Rotación con Aceleración Constante
15.
Cancer Res ; 67(7): 3002-9, 2007 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-17409406

RESUMEN

Malignant rhabdoid tumors (MRT) are rare aggressive cancers that occur in young children. Seventy-five percent of sporadic MRTs harbor inactivating SNF5 mutations, and mice heterozygous for an Snf5-null allele develop MRTs with partial penetrance. The diagnosis of choroid plexus carcinomas (CPC) in addition to MRTs in families with a single mutant SNF5 allele prompted us to assess the role of SNF5 loss in CPC in genetically engineered mice. With high frequency, TgT(121) mice develop CPCs that are initiated by inactivation of retinoblastoma protein (pRb) and related proteins p107 and p130. However, CPC penetrance and latency were not significantly affected by Snf5 heterozygosity, consistent with recent evidence that CPCs in SNF5 families were, in many cases, misdiagnosed MRTs. Surprisingly, although the CPC phenotype was unaffected, TgT(121);Snf5(+/-) mice developed MRTs with increased penetrance and decreased latency compared with TgT(121);Snf5(+/+) littermates. MRTs expressed the T(121) protein with a concomitant increase in mitotic activity. The predominant appearance of TgT(121);Snf5(+/-) MRTs in the spinal cord led to the discovery that these tumors likely arose from a subset of spinal cord neural progenitor cells expressing T(121) rather than from transdifferentiation of CPC. Significantly, the target cell type(s) for MRT is unknown. Hence, this study not only shows that pRb(f) and SNF5 inactivation cooperate to induce MRTs but also provides new insight into the MRT target population.


Asunto(s)
Neoplasias del Plexo Coroideo/genética , Proteínas Cromosómicas no Histona/genética , Proteína de Retinoblastoma/genética , Tumor Rabdoide/genética , Animales , ADN de Neoplasias/genética , ADN de Neoplasias/aislamiento & purificación , Femenino , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Pérdida de Heterocigocidad , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína SMARCB1
16.
Sci Rep ; 8(1): 10107, 2018 07 04.
Artículo en Inglés | MEDLINE | ID: mdl-29973630

RESUMEN

Studies indicate that the gut microbiota (GM) can significantly influence both local and systemic host physiologic processes. With rising concern for optimization of experimental reproducibility and translatability, it is essential to consider the GM in study design. However, GM profiles can vary between rodent producers making consistency between models challenging. To circumvent this, we developed outbred CD1 mouse colonies with stable, complex GM profiles that can be used as donors for a variety of GM transfer techniques including rederivation, co-housing, cross-foster, and fecal microbiota transfer (FMT). CD1 embryos were surgically transferred into CD1 or C57BL/6 surrogate dams that varied by GM composition and complexity to establish four separate mouse colonies harboring GM profiles representative of contemporary mouse producers. Using targeted 16S rRNA amplicon sequencing, subsequent female offspring were found to have similar GM profiles to surrogate dams. Furthermore, breeding colonies of CD1 mice with distinct GM profiles were maintained for nine generations, demonstrating GM stability within these colonies. To confirm GM stability, we shipped cohorts of these four colonies to collaborating institutions and found no significant variation in GM composition. These mice are an invaluable experimental resource that can be used to investigate GM effects on mouse model phenotype.


Asunto(s)
Cruzamiento/métodos , Trasplante de Microbiota Fecal/métodos , Microbioma Gastrointestinal , Animales , Transferencia de Embrión/métodos , Femenino , Vivienda para Animales , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales
17.
Nat Commun ; 9(1): 1988, 2018 05 18.
Artículo en Inglés | MEDLINE | ID: mdl-29777108

RESUMEN

Lung cancer is the leading cause of cancer-related deaths worldwide, and lung squamous carcinomas (LUSC) represent about 30% of cases. Molecular aberrations in lung adenocarcinomas have allowed for effective targeted treatments, but corresponding therapeutic advances in LUSC have not materialized. However, immune checkpoint inhibitors in sub-populations of LUSC patients have led to exciting responses. Using computational analyses of The Cancer Genome Atlas, we identified a subset of LUSC tumors characterized by dense infiltration of inflammatory monocytes (IMs) and poor survival. With novel, immunocompetent metastasis models, we demonstrated that tumor cell derived CCL2-mediated recruitment of IMs is necessary and sufficient for LUSC metastasis. Pharmacologic inhibition of IM recruitment had substantial anti-metastatic effects. Notably, we show that IMs highly express Factor XIIIA, which promotes fibrin cross-linking to create a scaffold for LUSC cell invasion and metastases. Consistently, human LUSC samples containing extensive cross-linked fibrin in the microenvironment correlated with poor survival.


Asunto(s)
Carcinoma de Células Escamosas/inmunología , Factor XIIIa/inmunología , Fibrina/química , Neoplasias Pulmonares/inmunología , Monocitos/inmunología , Animales , Biomarcadores de Tumor/química , Biomarcadores de Tumor/inmunología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Quimiocina CCL2/genética , Quimiocina CCL2/inmunología , Factor XIIIa/genética , Femenino , Fibrina/inmunología , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos DBA , Invasividad Neoplásica
18.
Mol Cell Biol ; 23(4): 1269-77, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12556487

RESUMEN

The INK4 family of cyclin-dependent kinase (CDK) inhibitors negatively regulates cyclin D-dependent CDK4 and CDK6 and thereby retains the growth-suppressive function of Rb family proteins. Mutations in the CDK4 gene conferring INK4 resistance are associated with familial and sporadic melanoma in humans and result in a wide spectrum of tumors in mice. Whereas loss of function of other INK4 genes in mice leads to little or no tumor development, targeted deletion of p18(INK4c) causes spontaneous pituitary tumors and lymphoma late in life. Here we show that treatment of p18 null and heterozygous mice with a chemical carcinogen resulted in tumor development at an accelerated rate. The remaining wild-type allele of p18 was neither mutated nor silenced in tumors derived from heterozygotes. Hence, p18 is a haploinsufficient tumor suppressor in mice.


Asunto(s)
Carcinógenos/toxicidad , Proteínas de Ciclo Celular , Predisposición Genética a la Enfermedad , Neoplasias Experimentales/inducido químicamente , Proteínas Supresoras de Tumor/genética , Adenoma/inducido químicamente , Adenoma/genética , Adenoma/patología , Animales , Carcinoma/inducido químicamente , Carcinoma/genética , Carcinoma/patología , Inhibidor p18 de las Quinasas Dependientes de la Ciclina , Dimetilaminas/toxicidad , Inhibidores Enzimáticos/metabolismo , Haplotipos , Hemangiosarcoma/inducido químicamente , Hemangiosarcoma/genética , Hemangiosarcoma/patología , Neoplasias Hepáticas/inducido químicamente , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Neoplasias Pulmonares/inducido químicamente , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Mutantes , Neoplasias Experimentales/genética , Neoplasias Experimentales/patología , Neoplasias Hipofisarias/inducido químicamente , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Proteínas Supresoras de Tumor/metabolismo
19.
Mol Cell Biol ; 24(20): 8907-16, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15456865

RESUMEN

Epiregulin, an epidermal growth factor family member, acts as a local signal mediator and shows dual biological activity, stimulating the proliferation of fibroblasts, hepatocytes, smooth muscle cells, and keratinocytes while inhibiting the growth of several tumor-derived epithelial cell lines. The epiregulin gene (Ereg) is located on mouse chromosome 5 adjacent to three other epidermal growth factor family members, epigen, amphiregulin, and betacellulin. Gene targeting was used to insert a lacZ reporter into the mouse Ereg locus and to ablate its function. Although epiregulin is broadly expressed and regulated both spatially and temporally, Ereg null mice show no overt developmental defects, reproductive abnormalities, or altered liver regeneration. Additionally, in contrast to previous hypotheses, Ereg deficiency does not alter intestinal cancer susceptibility, as assayed in the ApcMin model, despite showing robust expression in developing tumors. However, Ereg null mice are highly susceptible to cancer-predisposing intestinal damage caused by oral administration of dextran sulfate sodium.


Asunto(s)
Factor de Crecimiento Epidérmico/genética , Factor de Crecimiento Epidérmico/metabolismo , Mucosa Intestinal/patología , Neoplasias Intestinales/metabolismo , Animales , Peso Corporal , Línea Celular , Colon/anatomía & histología , Colon/patología , Sulfato de Dextran/administración & dosificación , Sulfato de Dextran/farmacología , Modelos Animales de Enfermedad , Epirregulina , Células Epiteliales/citología , Células Epiteliales/metabolismo , Marcación de Gen , Genes Reporteros , Mucosa Intestinal/anatomía & histología , Mucosa Intestinal/efectos de los fármacos , Neoplasias Intestinales/patología , Regeneración Hepática/fisiología , Masculino , Ratones , Ratones Noqueados , Distribución Tisular
20.
Lab Anim (NY) ; 36(10): 32-9, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17957178

RESUMEN

Scientific research has yet to conclusively determine the optimal cage size for mice. The authors examined the effect of cage size on mouse breeding performance and on offspring behavior, which can serve as indications of overall well-being. They housed breeding trios of C57BL/6Tac mice in standard or large individually ventilated cages and measured four reproductive parameters: litter size; litter survival to weaning age; average pup weight at 7, 14 and 21 days; and the number of days between litter births. They investigated the behavior of a subset of male and female pups from parents housed in cages of each size in the elevated plus maze test, the open field assay and the acoustic startle test. Cage size had no significant effect on any of the reproductive parameters measured and few or inconsistent effects on behavior in weaned pups.


Asunto(s)
Conducta Animal/fisiología , Vivienda para Animales , Reproducción/fisiología , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Conducta Exploratoria/fisiología , Femenino , Tamaño de la Camada/fisiología , Masculino , Aprendizaje por Laberinto , Ratones , Ratones Endogámicos C57BL , Reflejo de Sobresalto , Enfermedades de los Roedores/mortalidad , Tasa de Supervivencia , Destete , Aumento de Peso/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA