Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
2.
Mamm Genome ; 33(1): 203-212, 2022 03.
Article in English | MEDLINE | ID: mdl-34313795

ABSTRACT

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.


Subject(s)
Biomedical Research , Disease Models, Animal , Mice , National Institutes of Health (U.S.) , Animals , Humans , Mice/genetics , Reproducibility of Results , United States
3.
Vet Pathol ; 58(1): 10-33, 2021 01.
Article in English | MEDLINE | ID: mdl-33016246

ABSTRACT

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.


Subject(s)
Central Nervous System Diseases , Peripheral Nervous System Diseases , Animals , Central Nervous System , Central Nervous System Diseases/veterinary , Peripheral Nervous System , Peripheral Nervous System Diseases/diagnosis , Peripheral Nervous System Diseases/veterinary , Spinal Cord
4.
Mol Microbiol ; 100(1): 90-107, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26712165

ABSTRACT

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.


Subject(s)
Bacterial Proteins/metabolism , Membrane Proteins/metabolism , Mycobacterium tuberculosis/metabolism , Animals , Bacterial Proteins/genetics , Cholesterol/metabolism , Disease Models, Animal , Gene Deletion , Gene Order , Membrane Proteins/genetics , Mice , Mutation , Mycobacterium tuberculosis/genetics , Phenotype , Protein Binding , Protein Transport , Tuberculosis/microbiology , Tuberculosis/mortality , Tuberculosis/pathology , Virulence Factors
5.
Cancer Cell ; 12(4): 355-66, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17936560

ABSTRACT

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.


Subject(s)
Gene Expression Regulation, Developmental , Mutagenesis, Site-Directed , Proto-Oncogene Proteins c-mdm2/metabolism , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Amino Acid Substitution , Animals , Cells, Cultured , DNA Damage , Down-Regulation , Embryo, Mammalian , Fibroblasts/enzymology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gamma Rays , Gene Expression Regulation, Developmental/radiation effects , Genotype , Gestational Age , Homozygote , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Phenotype , Proteasome Endopeptidase Complex/metabolism , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/chemistry , Proto-Oncogene Proteins c-mdm2/deficiency , Proto-Oncogene Proteins c-mdm2/genetics , Transcription, Genetic/radiation effects , Tumor Suppressor Protein p53/deficiency , Tumor Suppressor Protein p53/genetics
6.
J Cell Physiol ; 229(1): 44-52, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23757284

ABSTRACT

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.


Subject(s)
B-Lymphocytes/metabolism , Chromatin Assembly and Disassembly/genetics , DNA Helicases , Lymphocyte Activation/genetics , Nuclear Proteins , Transcription Factors , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Cell Differentiation/genetics , Cell Nucleus/genetics , Cell Nucleus/metabolism , DNA Helicases/genetics , DNA Helicases/metabolism , Gene Expression Regulation, Developmental , Humans , Mice , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcriptional Activation
7.
Mamm Genome ; 25(3-4): 95-108, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24487921

ABSTRACT

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.


Subject(s)
Breeding/methods , Disease Models, Animal , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/physiopathology , Mice, Inbred Strains/genetics , Animals , Chromosome Mapping , Crosses, Genetic , DNA Primers/genetics , Genotype , Humans , Mice , Mice, Inbred C57BL , Pedigree , Polymerase Chain Reaction , Quantitative Trait Loci/genetics
8.
Circ Res ; 111(5): e111-22, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22740088

ABSTRACT

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.


Subject(s)
Chromosomal Proteins, Non-Histone/metabolism , DNA Helicases/metabolism , Endothelial Cells/metabolism , Heart Defects, Congenital/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Age Factors , Animals , Catalysis , Cell Death/physiology , Cell Lineage/physiology , Cell Survival/physiology , Chromosomal Proteins, Non-Histone/genetics , Coronary Vessels/embryology , Coronary Vessels/metabolism , Coronary Vessels/pathology , DNA Helicases/genetics , Echocardiography , Endothelial Cells/pathology , Heart/embryology , Heart Defects, Congenital/genetics , Heart Defects, Congenital/pathology , Homeostasis/physiology , Mice , Mice, Transgenic , Myocardial Ischemia/genetics , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Nuclear Proteins/genetics , Pleural Effusion/genetics , Pleural Effusion/metabolism , Pleural Effusion/pathology , Transcription Factors/genetics
9.
Sci Adv ; 8(4): eabl5838, 2022 Jan 28.
Article in English | MEDLINE | ID: mdl-35080986

ABSTRACT

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.
Article in English | MEDLINE | ID: mdl-16230479

ABSTRACT

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.


Subject(s)
Autoimmune Diseases/genetics , Forkhead Transcription Factors/genetics , Lymphatic Diseases/genetics , Mutation/genetics , Thymus Gland/growth & development , Thymus Gland/pathology , Animals , Apoptosis/physiology , Autoimmune Diseases/pathology , Bromodeoxyuridine , DNA Primers , Flow Cytometry , Luciferases , Lymphatic Diseases/pathology , Mice , Mice, Inbred BALB C , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
11.
World Neurosurg ; 148: e242-e251, 2021 04.
Article in English | MEDLINE | ID: mdl-33412322

ABSTRACT

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.


Subject(s)
Carotid Artery Diseases/chemically induced , Disease Models, Animal , Intracranial Aneurysm/complications , Magnetic Resonance Angiography , Pancreatic Elastase/toxicity , Rabbits/physiology , Animals , Aspirin/therapeutic use , Carotid Artery Diseases/diagnostic imaging , Carotid Artery Diseases/pathology , Carotid Artery, Common/diagnostic imaging , Carotid Artery, Common/drug effects , Carotid Artery, Common/pathology , Carotid Artery, Common/physiology , Disease Progression , Elastic Tissue/ultrastructure , Female , Hyperplasia , Infusions, Intra-Arterial , Intracranial Aneurysm/chemically induced , Intracranial Aneurysm/diagnostic imaging , Intracranial Aneurysm/drug therapy , Male , Myocytes, Smooth Muscle/pathology , Necrosis , Pancreatic Elastase/administration & dosage , Rabbits/immunology , Regeneration , Sex Characteristics , Single-Blind Method , Tunica Intima/pathology , Tunica Media/pathology , Vasculitis/drug therapy , Vasculitis/etiology , Vasculitis/pathology
12.
J Exp Med ; 198(12): 1937-49, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-14676303

ABSTRACT

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.


Subject(s)
Nuclear Proteins/physiology , T-Lymphocytes/physiology , Transcription Factors/physiology , Animals , Apoptosis , CD4 Antigens/analysis , CD8 Antigens/analysis , Cell Cycle Proteins/analysis , DNA Helicases , Drosophila Proteins , Helicobacter Infections/complications , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell, gamma-delta/analysis , Rectal Prolapse/etiology , Thymus Gland/physiology , Trans-Activators/analysis
13.
Mol Carcinog ; 48(12): 1139-48, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19676100

ABSTRACT

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.


Subject(s)
Bone Neoplasms/pathology , Chromosomal Proteins, Non-Histone/physiology , Lymphoma, T-Cell/pathology , Osteosarcoma/pathology , Rhabdoid Tumor/pathology , Tumor Suppressor Protein p53/physiology , Animals , Bone Neoplasms/genetics , Cell Cycle Proteins/metabolism , Female , Lymphoma, T-Cell/genetics , Male , Mice , Mice, Knockout , Osteosarcoma/genetics , Rhabdoid Tumor/genetics , SMARCB1 Protein , Survival Rate
14.
Lab Anim (NY) ; 38(1): 24-34, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19112447

ABSTRACT

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.


Subject(s)
Exploratory Behavior/physiology , Fertility/physiology , Housing, Animal , Sexual Behavior, Animal/physiology , Analysis of Variance , Animals , Anxiety/pathology , Body Weight , Female , Litter Size/physiology , Male , Maze Learning/physiology , Mice , Mice, Inbred C57BL , Rotarod Performance Test
15.
Cancer Res ; 67(7): 3002-9, 2007 Apr 01.
Article in English | MEDLINE | ID: mdl-17409406

ABSTRACT

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.


Subject(s)
Choroid Plexus Neoplasms/genetics , Chromosomal Proteins, Non-Histone/genetics , Retinoblastoma Protein/genetics , Rhabdoid Tumor/genetics , Animals , DNA, Neoplasm/genetics , DNA, Neoplasm/isolation & purification , Female , Gene Expression Regulation, Neoplastic , Gene Silencing , Loss of Heterozygosity , Male , Mice , Mice, Inbred C57BL , SMARCB1 Protein
16.
Sci Rep ; 8(1): 10107, 2018 07 04.
Article in English | MEDLINE | ID: mdl-29973630

ABSTRACT

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.


Subject(s)
Breeding/methods , Fecal Microbiota Transplantation/methods , Gastrointestinal Microbiome , Animals , Embryo Transfer/methods , Female , Housing, Animal , Male , Mice , Mice, Inbred C57BL , Models, Animal
17.
Nat Commun ; 9(1): 1988, 2018 05 18.
Article in English | MEDLINE | ID: mdl-29777108

ABSTRACT

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.


Subject(s)
Carcinoma, Squamous Cell/immunology , Factor XIIIa/immunology , Fibrin/chemistry , Lung Neoplasms/immunology , Monocytes/immunology , Animals , Biomarkers, Tumor/chemistry , Biomarkers, Tumor/immunology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Chemokine CCL2/genetics , Chemokine CCL2/immunology , Factor XIIIa/genetics , Female , Fibrin/immunology , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Male , Mice , Mice, Inbred DBA , Neoplasm Invasiveness
18.
Mol Cell Biol ; 23(4): 1269-77, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12556487

ABSTRACT

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.


Subject(s)
Carcinogens/toxicity , Cell Cycle Proteins , Genetic Predisposition to Disease , Neoplasms, Experimental/chemically induced , Tumor Suppressor Proteins/genetics , Adenoma/chemically induced , Adenoma/genetics , Adenoma/pathology , Animals , Carcinoma/chemically induced , Carcinoma/genetics , Carcinoma/pathology , Cyclin-Dependent Kinase Inhibitor p18 , Dimethylamines/toxicity , Enzyme Inhibitors/metabolism , Haplotypes , Hemangiosarcoma/chemically induced , Hemangiosarcoma/genetics , Hemangiosarcoma/pathology , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Lung Neoplasms/chemically induced , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice , Mice, Mutant Strains , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Pituitary Neoplasms/chemically induced , Pituitary Neoplasms/genetics , Pituitary Neoplasms/pathology , Tumor Suppressor Proteins/metabolism
19.
Mol Cell Biol ; 24(20): 8907-16, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15456865

ABSTRACT

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.


Subject(s)
Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , Intestinal Mucosa/pathology , Intestinal Neoplasms/metabolism , Animals , Body Weight , Cell Line , Colon/anatomy & histology , Colon/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/pharmacology , Disease Models, Animal , Epiregulin , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Targeting , Genes, Reporter , Intestinal Mucosa/anatomy & histology , Intestinal Mucosa/drug effects , Intestinal Neoplasms/pathology , Liver Regeneration/physiology , Male , Mice , Mice, Knockout , Tissue Distribution
20.
Lab Anim (NY) ; 36(10): 32-9, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17957178

ABSTRACT

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.


Subject(s)
Behavior, Animal/physiology , Housing, Animal , Reproduction/physiology , Animals , Animals, Newborn/growth & development , Exploratory Behavior/physiology , Female , Litter Size/physiology , Male , Maze Learning , Mice , Mice, Inbred C57BL , Reflex, Startle , Rodent Diseases/mortality , Survival Rate , Weaning , Weight Gain/physiology
SELECTION OF CITATIONS
SEARCH DETAIL