Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
BMC Res Notes ; 10(1): 573, 2017 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-29116018

RESUMO

OBJECTIVE: Gain-of-function (GOF) mutations in RHBDF2 cause tylosis. Patients present with hyperproliferative skin, and keratinocytes from tylosis patients' skin show an enhanced wound-healing phenotype. The curly bare mouse model of tylosis, carrying a GOF mutation in the Rhbdf2 gene (Rhbdf2 cub ), presents with epidermal hyperplasia and shows accelerated cutaneous wound-healing phenotype through enhanced secretion of the epidermal growth factor receptor family ligand amphiregulin. Despite these advances in our understanding of tylosis, key questions remain. For instance, it is not known whether the disease is skin-specific, whether the immune system or the surrounding microenvironment plays a role, and whether mouse genetic background influences the hyperproliferative-skin and wound-healing phenotypes observed in Rhbdf2 cub mice. RESULTS: We performed bone marrow transfers and reciprocal skin transplants and found that bone marrow transfer from C57BL/6 (B6)-Rhbdf2 cub/cub donor mice to B6 wildtype recipient mice failed to transfer the hyperproliferative-skin and wound-healing phenotypes in B6 mice. Furthermore, skin grafts from B6 mice to the dorsal skin of B6-Rhbdf2 cub/cub mice maintained the phenotype of the donor mice. To test the influence of mouse genetic background, we backcrossed Rhbdf2 cub onto the MRL/MpJ strain and found that the hyperproliferative-skin and wound-healing phenotypes caused by the Rhbdf2 cub mutation persisted on the MRL/MpJ strain.


Assuntos
Proteínas de Transporte/fisiologia , Queratinócitos , Ceratodermia Palmar e Plantar Difusa/genética , Transplante de Pele , Cicatrização/genética , Animais , Transplante de Medula Óssea , Proliferação de Células/genética , Modelos Animais de Doenças , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Fenótipo
2.
Comp Med ; 67(4): 335-343, 2017 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-28830580

RESUMO

Segmented filamentous bacterium (SFB) a gram-positive, anaerobic, and intestinal commensal organism directly influences the development of Th17 helper cells in the small intestine of mice. In NOD mice, SFB colonization interferes with the development of type 1 diabetes (T1D), a T-cell-mediated autoimmune disease, suggesting that SFB may influence Th17 cells to inhibit Th1 populations associated with the anti-ß-cell immune response. This effect is a serious concern for investigators who use NOD mice for diabetes research because the expected incidence of disease decreases markedly when they are colonized by SFB. A room housing mice for T1D studies at The Jackson Laboratory was determined by fecal PCR testing to have widespread SFB colonization of multiple NOD strains after a steady decline in the incidence of T1D was noted. Rederivation of all NOD-related mouse strains was not feasible; therefore an alternative treatment using antibiotics to eliminate SFB from colonized mice was undertaken. After antibiotic treatment, soiled bedding from NOD mouse strains housed in SFB-free high-health-status production barrier rooms was used to reintroduce the gastrointestinal microbiota. Over the past 16 mo since treating the mice and disinfecting the mouse room, regular PCR testing has shown that no additional SFB colonization of mice has occurred, and the expected incidence of T1D has been reestablished in the offspring of treated mice.


Assuntos
Ampicilina/farmacologia , Antibacterianos/farmacologia , Diabetes Mellitus Tipo 1/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Bactérias Gram-Positivas/efeitos dos fármacos , Infecções por Bactérias Gram-Positivas/veterinária , Intestinos/efeitos dos fármacos , Criação de Animais Domésticos/métodos , Animais , Descontaminação/métodos , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Modelos Animais de Doenças , Monitoramento Ambiental/métodos , Fezes/microbiologia , Predisposição Genética para Doença , Bactérias Gram-Positivas/classificação , Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/tratamento farmacológico , Infecções por Bactérias Gram-Positivas/imunologia , Infecções por Bactérias Gram-Positivas/microbiologia , Interações Hospedeiro-Patógeno , Intestinos/imunologia , Intestinos/microbiologia , Camundongos Endogâmicos NOD , Fenótipo , Células Th1/imunologia , Células Th1/microbiologia , Células Th17/imunologia , Células Th17/microbiologia , Fatores de Tempo
3.
Endocrinology ; 158(9): 2707-2712, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28633442

RESUMO

Previous studies with continuous glucose monitoring in mice have been limited to several days or weeks, with the mouse's physical attachment to the equipment affecting behavior and measurements. In the current study, we measured blood glucose and body temperature at 10-second intervals for 12 weeks in a cohort of NOD/ShiLtJ female mice using wireless telemetry. This allowed us to obtain a high-resolution profile of the circadian rhythm of these two parameters and the onset of hyperglycemic development in real time. The most striking observations were the elevated nocturnal concentrations of glucose into the diabetic range days before elevations in diurnal glucose (when glucose concentrations are historically measured) and the strong, negative correlation between elevated blood glucose concentrations and body temperature with a steady decline of the body temperature with diabetes development. Taken together, this technological advancement provides improved resolution in the study of the disease trajectory of diabetes in mouse models, including relevant translatability to the current technologies of continuous glucose monitoring now regularly used in patients.


Assuntos
Glicemia/análise , Temperatura Corporal/fisiologia , Ritmo Circadiano/fisiologia , Telemetria/métodos , Tecnologia sem Fio , Animais , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Progressão da Doença , Feminino , Teste de Tolerância a Glucose , Hiperglicemia/metabolismo , Hiperglicemia/patologia , Camundongos , Camundongos Endogâmicos NOD
4.
Cold Spring Harb Protoc ; 2014(7): 737-40, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24987138

RESUMO

Multiple sites can be used for the engraftment of primary human cells and tissues into murine hosts. For example, leukemias are usually best engrafted intravenously, but they can also be engrafted directly into the bone marrow cavity. Some solid tumors such as colon tumors grow successfully following subcutaneous engraftment, sometimes requiring provision of a Matrigel artificial basement membrane. In certain cases (e.g., human bladder cancer and ductal breast carcinoma), the use of the autochthonous site (bladder and mammary duct, respectively) is often most efficient, whereas the tumors can grow poorly when transplanted subcutaneously or heterochthonously. Here, we present a protocol for the surgical implantation of tissues under the kidney capsule. The kidney is especially suited for the transplantation of normal as well as malignant cells and tissues. It is very accessible, and transplanted tissues are well contained under the renal capsule in a highly vascularized site. Furthermore, the retroperitoneal location of the kidney, together with its separation from other organs, is advantageous both for imaging and biopsy.


Assuntos
Rim/cirurgia , Transplante Heterólogo/métodos , Animais , Cápsula Glomerular/cirurgia , Humanos , Camundongos , Transplante de Neoplasias
5.
Cold Spring Harb Protoc ; 2014(7): 694-708, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24987146

RESUMO

Since the discovery of the "nude" mouse more than 40 years ago, investigators have attempted to model human tumor growth in immunodeficient mice. Here, we summarize how the field has advanced over the ensuing years owing to improvements in the murine recipients of human tumors. These improvements include the discovery of the scid mutation and development of targeted mutations in the recombination-activating genes 1 and 2 (Rag1(null), Rag2(null)) that severely cripple the adaptive immune response of the murine host. More recently, mice deficient in adaptive immunity have been crossed with mice bearing targeted mutations designed to weaken the innate immune system, ultimately leading to the development of immunodeficient mice bearing a targeted mutation in the gene encoding the interleukin 2 (IL2) receptor common γ chain (IL2rg(null), also known in humans as cytokine receptor common subunit γ). The IL2rg(null) mutation has been used to develop several immunodeficient strains of mice, including the NOD-scid IL2rg(null) (NSG) strain. Using NSG mice as human xenograft recipients, it is now possible to grow almost all types of primary human tumors in vivo, including most solid tumors and hematological malignancies that maintain characteristics of the primary tumor in the patient. Programs to optimize patient-specific therapy using patient-derived xenograft tumor growth in NSG mice have been established at several institutions, including The Jackson Laboratory. Moreover, NSG mice can be engrafted with functional human immune systems, permitting for the first time the potential to study primary human tumors in vivo in the presence of a human immune system.


Assuntos
Modelos Animais de Doenças , Neoplasias/patologia , Neoplasias/terapia , Animais , Humanos , Sistema Imunitário/patologia , Camundongos , Camundongos SCID , Neoplasias/imunologia , Pesquisa/tendências , Transplante Heterólogo/tendências
6.
PLoS One ; 8(1): e53426, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23301070

RESUMO

"Hairpatches" (Hpt) is a naturally occurring, autosomal semi-dominant mouse mutation. Hpt/Hpt homozygotes die in utero, while Hpt/+ heterozygotes exhibit progressive renal failure accompanied by patchy alopecia. This mutation is a model for the rare human disorder "glomerulonephritis with sparse hair and telangiectases" (OMIM 137940). Fine mapping localized the Hpt locus to a 6.7 Mb region of Chromosome 4 containing 62 known genes. Quantitative real time PCR revealed differential expression for only one gene in the interval, T-cell acute lymphocytic leukemia 1 (Tal1), which was highly upregulated in the kidney and skin of Hpt/+ mice. Southern blot analysis of Hpt mutant DNA indicated a new EcoRI site in the Tal1 gene. High throughput sequencing identified an endogenous retroviral class II intracisternal A particle insertion in Tal1 intron 4. Our data suggests that the IAP insertion in Tal1 underlies the histopathological changes in the kidney by three weeks of age, and that glomerulosclerosis is a consequence of an initial developmental defect, progressing in severity over time. The Hairpatches mouse model allows an investigation into the effects of Tal1, a transcription factor characterized by complex regulation patterns, and its effects on renal disease.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Elementos de DNA Transponíveis/genética , Nefropatias/metabolismo , Mutação , Proteínas Proto-Oncogênicas/genética , Alopecia/genética , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Mapeamento Cromossômico , Cruzamentos Genéticos , Modelos Animais de Doenças , Feminino , Regulação da Expressão Gênica , Ligação Genética , Nefropatias/virologia , Camundongos , Proteínas Proto-Oncogênicas/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Proteína 1 de Leucemia Linfocítica Aguda de Células T , Fatores de Tempo , Fatores de Transcrição , Regulação para Cima
7.
Curr Protoc Mouse Biol ; 2(3): 263-71, 2012 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26069014

RESUMO

Personnel performing surgery on mice commonly lack formal medical training and most training in mouse surgery focuses on the mechanical skills required for a procedure with less emphasis on perioperative techniques. Consequently, a basic concept that underpins successful surgery, aseptic technique, may not be fully understood or incorporated into the training process. This unit provides a framework in which to plan and carry out surgical procedures in mice using appropriate aseptic technique. Curr. Protoc. Mouse Biol. 2:263-271 © 2012 by John Wiley & Sons, Inc.

8.
Blood ; 115(6): 1267-76, 2010 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-19846887

RESUMO

The spontaneous mouse mutation "thrombocytopenia and cardiomyopathy" (trac) causes macrothrombocytopenia, prolonged bleeding times, anemia, leukopenia, infertility, cardiomyopathy, and shortened life span. Homozygotes show a 20-fold decrease in platelet numbers and a 3-fold increase in platelet size with structural alterations and functional impairments in activation and aggregation. Megakaryocytes in trac/trac mice are present in increased numbers, have poorly developed demarcation membrane systems, and have decreased polyploidy. The thrombocytopenia is not intrinsic to defects at the level of hematopoietic progenitor cells but is associated with a microenvironmental abnormality. The trac mutation maps to mouse chromosome 17, syntenic with human chromosome 2p21-22. A G to A mutation in exon 10 of the adenosine triphosphate (ATP)-binding cassette subfamily G, member 5 (Abcg5) gene, alters a tryptophan codon (UGG) to a premature stop codon (UAG). Crosses with mice doubly transgenic for the human ABCG5 and ABCG8 genes rescued platelet counts and volumes. ABCG5 and ABCG8 form a functional complex that limits dietary phytosterol accumulation. Phytosterolemia in trac/trac mice confirmed a functional defect in the ABCG5/ABCG8 transport system. The trac mutation provides a new clinically significant animal model for human phytosterolemia and provides a new means for studying the role of phytosterols in hematologic diseases and testing therapeutic interventions.


Assuntos
Transportadores de Cassetes de Ligação de ATP/fisiologia , Cardiomiopatias/genética , Modelos Animais de Doenças , Erros Inatos do Metabolismo Lipídico/genética , Lipoproteínas/fisiologia , Mutação/genética , Fitosteróis/metabolismo , Sitosteroides/metabolismo , Trombocitopenia/genética , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Membro 8 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Tempo de Sangramento , Cardiomiopatias/patologia , Células Cultivadas , Ensaio de Unidades Formadoras de Colônias , Cruzamentos Genéticos , Feminino , Feto/citologia , Feto/metabolismo , Erros Inatos do Metabolismo Lipídico/patologia , Lipoproteínas/genética , Fígado/citologia , Fígado/metabolismo , Masculino , Megacariócitos/citologia , Megacariócitos/metabolismo , Camundongos , Camundongos Endogâmicos A , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Trombocitopenia/patologia
9.
Invest Ophthalmol Vis Sci ; 47(3): 1201-9, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16505059

RESUMO

PURPOSE: Viable motheaten mutant mice (abbreviated allele symbol me(v)) are deficient in Src-homology 2-domain phosphatase (SHP)-1, a critical negative regulator of signal transduction in hematopoietic cells. These mice exhibit immune dysfunction, hyperproliferation of myeloid cells, and regenerative anemia. This study focused on the role of SHP-1 in retinal homeostasis. METHODS: Ophthalmoscopy, histology, transmission electron microscopy (TEM), electroretinography (ERG), immunohistochemistry, Western blot, bone marrow transplantation, and genetic crosses were performed for phenotypic characterization and functional studies of retinal degeneration (RD) in me(v)/me(v) mice. RESULTS: Fundus examinations of me(v)/me(v) mice revealed numerous, small white spots. Histologic examination demonstrated photoreceptor loss beginning at 3 weeks of age, and TEM revealed disorganization and reduction in the number of outer segments, as well as the presence of phagocytic cells in the subretinal space. Rod- and cone-mediated ERGs were abnormal. SHP-1 protein was expressed in mouse and human retinal lysates and was localized to the outer nuclear layer of the retina in me(v)/me(v) and control mice. Autoantibodies are not necessary for RD, as B-cell-deficient me(v)/me(v) Igh-6(tm1Cgn) mice had no attenuation of photoreceptor cell loss compared with age-matched me(v)/me(v) mice. Histologic examination of lungs and retinas from normal recipients of me(v)/me(v) marrow revealed the classic acidophilic macrophage pneumonia of me(v)/me(v) mice, but no retinal degeneration. CONCLUSIONS: me(v)/me(v) mice exhibit normal retinal development with the onset of RD at 3 weeks of age and a rapidly progressive loss of photoreceptors. These findings support the hypothesis that SHP-1 plays a critical role in retinal homeostasis.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/deficiência , Proteínas Tirosina Fosfatases/deficiência , Degeneração Retiniana/enzimologia , Animais , Apoptose , Western Blotting , Transplante de Medula Óssea , Cruzamentos Genéticos , Eletrorretinografia , Feminino , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Oftalmoscopia , Células Fotorreceptoras de Vertebrados/enzimologia , Células Fotorreceptoras de Vertebrados/fisiologia , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Degeneração Retiniana/fisiopatologia
10.
J Exp Med ; 203(2): 275-80, 2006 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-16476768

RESUMO

Rheumatoid arthritis (RA) is a complex autoimmune disease with a poorly understood pathogenesis. The disease is associated with polyclonal B cell activation and the production of autoantibodies (autoAbs), but there is a longstanding controversy as to whether such Abs contribute to, or are secondary to, the pathogenesis of RA. To address the potential pathogenicity of human RA-associated Abs, we developed a passive transfer model involving mice deficient in the low-affinity inhibitory Fc receptor, FcgammaRIIB. We report that plasma or serum from patients with active RA can induce inflammation and histological lesions in FcgammaRIIB-/- mice consistent with arthritis, and that this pathogenic activity is caused by the immunoglobulin G-rich fraction. Our results suggest that humoral autoimmunity can contribute directly to autoimmune arthritis, and that FcgammaRIIB-/- mice are a promising model to evaluate the arthritogenic potential of human autoAbs.


Assuntos
Artrite Experimental/imunologia , Autoanticorpos/toxicidade , Sítios de Ligação de Anticorpos , Receptores de IgG/deficiência , Receptores de IgG/genética , Animais , Artrite Experimental/genética , Artrite Experimental/patologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Sítios de Ligação de Anticorpos/genética , Feminino , Humanos , Injeções Intraperitoneais , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Invest Ophthalmol Vis Sci ; 46(11): 4097-106, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16249486

RESUMO

PURPOSE: The present study was conducted to investigate healing of alkali-burned corneas in MRL/MpJ (MRL) mice. METHODS: Gross, clinical, and histologic criteria were used to compare healing of alkali-burned corneas in MRL and control C57BL/6J (B6) mice. Effects of neutrophil depletion of B6 mice and allogeneic reconstitution of B6 mice with MRL bone marrow on wound healing were evaluated. Gene expression patterns in normal and wounded corneas were surveyed with array-based quantitative real-time RT-PCR (AQPCR). RESULTS: MRL mice showed accelerated reepithelialization and decreased corneal opacity compared with B6 mice after alkali wounding. Marked inflammatory cell infiltration and fibrosis were evident in the corneas and anterior chambers of B6 mice. MRL mice showed less severe lesions, except for stromal edema. Rapid reepithelialization and reduced keratitis/iritis were also observed in neutrophil-depleted B6 mice, but not in B6 mice reconstituted with MRL bone marrow. AQPCR showed transcriptional changes of fewer genes associated with inflammation and corneal tissue homeostasis in alkali-burned corneas from MRL mice. Increased expression of an anti-inflammatory gene, Socs1, and a gene associated with healing, Mmp1a, were evident in MRL corneas. CONCLUSIONS: Alkali-burned corneas heal faster and more completely in MRL mice than in B6 mice, by means of rapid reepithelialization, reduced inflammation, and reduced fibrosis. Reduced inflammation, including decreased neutrophil infiltrates and the lack of a robust proinflammatory gene expression signature correlates with the rapid healing. However, the rapid-healing phenotype is not intrinsic to MRL hematopoietic progenitor cells.


Assuntos
Queimaduras Químicas/metabolismo , Doenças da Córnea/metabolismo , Queimaduras Oculares/induzido quimicamente , Cicatrização/fisiologia , Animais , Transplante de Medula Óssea , Queimaduras Químicas/imunologia , Queimaduras Químicas/patologia , Queimaduras Químicas/cirurgia , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Colagenases/genética , Colagenases/metabolismo , Doenças da Córnea/imunologia , Doenças da Córnea/patologia , Doenças da Córnea/cirurgia , Modelos Animais de Doenças , Epitélio Corneano/fisiologia , Citometria de Fluxo , Regulação da Expressão Gênica/fisiologia , Masculino , Metaloproteinase 1 da Matriz , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos MRL lpr , Neutrófilos/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Hidróxido de Sódio , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/genética , Proteínas Supressoras da Sinalização de Citocina/metabolismo
12.
Exp Biol Med (Maywood) ; 230(9): 659-67, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16179734

RESUMO

The flaky skin (fsn) mutation in mice causes pleiotropic abnormalities including psoriasiform dermatitis, anemia, hyper-IgE, and anti-dsDNA autoantibodies resembling those detected in systemic lupus erythematosus. The fsn mutation was mapped to an interval of 3.9 kb on chromosome 17 between D17Mit130 and D17Mit162. Resequencing of known and predicted exons and regulatory sequences from this region in fsn/fsn and wild-type mice indicated that the mutation is due to the insertion of an endogenous retrovirus (early transposon class) into intron 14 of the Tetratricopeptide repeat (TPR) domain 7 (Ttc7) gene. The insertion leads to reduced levels of wild-type Ttc7 transcripts in fsn mice and the insertion of an additional exon derived from the retrovirus into the majority of Ttc7 mRNAs. This disrupts one of the TPRs within TTC7 and may affect its interaction with an as-yet unidentified protein partner. The Ttc7 is expressed in multiple types of tissue including skin, kidney, spleen, and thymus, but is most abundant in germinal center B cells and hematopoietic stem cells, suggesting an important role in the development of immune system cells. Its role in immunologic and hematologic disorders should be further investigated.


Assuntos
Anemia/genética , Autoimunidade/genética , Mutação , Proteínas/genética , Psoríase/genética , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA , Expressão Gênica , Ligação Genética , Humanos , Linfócitos/imunologia , Camundongos , Camundongos Endogâmicos , Dados de Sequência Molecular , Fenótipo , Homologia de Sequência de Aminoácidos
13.
J Immunol ; 174(10): 6477-89, 2005 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-15879151

RESUMO

Ethical considerations constrain the in vivo study of human hemopoietic stem cells (HSC). To overcome this limitation, small animal models of human HSC engraftment have been used. We report the development and characterization of a new genetic stock of IL-2R common gamma-chain deficient NOD/LtSz-scid (NOD-scid IL2Rgamma(null)) mice and document their ability to support human mobilized blood HSC engraftment and multilineage differentiation. NOD-scid IL2Rgamma(null) mice are deficient in mature lymphocytes and NK cells, survive beyond 16 mo of age, and even after sublethal irradiation resist lymphoma development. Engraftment of NOD-scid IL2Rgamma(null) mice with human HSC generate 6-fold higher percentages of human CD45(+) cells in host bone marrow than with similarly treated NOD-scid mice. These human cells include B cells, NK cells, myeloid cells, plasmacytoid dendritic cells, and HSC. Spleens from engrafted NOD-scid IL2Rgamma(null) mice contain human Ig(+) B cells and lower numbers of human CD3(+) T cells. Coadministration of human Fc-IL7 fusion protein results in high percentages of human CD4(+)CD8(+) thymocytes as well human CD4(+)CD8(-) and CD4(-)CD8(+) peripheral blood and splenic T cells. De novo human T cell development in NOD-scid IL2Rgamma(null) mice was validated by 1) high levels of TCR excision circles, 2) complex TCRbeta repertoire diversity, and 3) proliferative responses to PHA and streptococcal superantigen, streptococcal pyrogenic exotoxin. Thus, NOD-scid IL2Rgamma(null) mice engrafted with human mobilized blood stem cells provide a new in vivo long-lived model of robust multilineage human HSC engraftment.


Assuntos
Mobilização de Células-Tronco Hematopoéticas , Linfopoese/genética , Linfopoese/imunologia , Mielopoese/genética , Mielopoese/imunologia , Transplante de Células-Tronco de Sangue Periférico/métodos , Receptores de Interleucina-7/deficiência , Receptores de Interleucina-7/genética , Envelhecimento/genética , Envelhecimento/imunologia , Animais , Contagem de Células Sanguíneas , Citotoxicidade Imunológica/genética , Células Dendríticas/citologia , Feminino , Citometria de Fluxo , Mobilização de Células-Tronco Hematopoéticas/métodos , Humanos , Imunoglobulinas/sangue , Imunofenotipagem , Subunidade gama Comum de Receptores de Interleucina , Células Matadoras Naturais/imunologia , Longevidade/genética , Longevidade/imunologia , Ativação Linfocitária/genética , Linfoma/genética , Linfoma/imunologia , Linfoma/prevenção & controle , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Tolerância a Radiação/genética , Tolerância a Radiação/imunologia , Receptores de Interleucina-2/deficiência , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/fisiologia , Receptores de Interleucina-7/fisiologia , Baço/citologia , Baço/imunologia , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/imunologia
14.
Clin Immunol ; 112(3): 273-83, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15308121

RESUMO

Immunodeficient NOD mice engrafted with human peripheral blood mononuclear cells (PBMCs) were used in two models of human islet allograft rejection. Model one: human PBMCs were engrafted into chemically diabetic NOD-scid mice bearing established subrenal human islet allografts. Inflammation and often complete islet allograft rejection were observed. Model 2 incorporated three key advances. First, we developed a new immunodeficient recipient, NOD-RagI(null)Prf1(null) mice. Second, graft-lymphocyte interactions were optimized by intrasplenic co-transplantation of islets and human PBMC. Third, NOD-scid islets expressing human HLA-A2.1 were used as allograft targets. Diabetic NOD-RagI(null)Prf1(null) recipients of HLA-A2.1 transgenic mouse islets, alone or co-engrafted with HLA-A2-positive human PBMC, exhibited durable graft survival and euglycemia. Contrastingly, co-transplantation with HLA-A2-negative human PBMC led to islet graft rejection without evidence of graft-vs.-host disease (GVHD). We propose that diabetic NOD-RagI(null)Prf1(null) mice co-engrafted with HLA-A2 mouse transgenic islets and allogeneic human PBMC provide an effective in vivo model of human islet allograft rejection.


Assuntos
Genes RAG-1/genética , Rejeição de Enxerto/imunologia , Antígeno HLA-A2/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/imunologia , Linfócitos/imunologia , Imunodeficiência Combinada Severa/imunologia , Animais , Glicemia/análise , Diabetes Mellitus/sangue , Diabetes Mellitus/induzido quimicamente , Diabetes Mellitus/imunologia , Diabetes Mellitus/cirurgia , Deleção de Genes , Doença Enxerto-Hospedeiro/imunologia , Humanos , Glicoproteínas de Membrana/deficiência , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos Transgênicos , Perforina , Proteínas Citotóxicas Formadoras de Poros , Baço/imunologia , Transplante Homólogo/imunologia
15.
Exp Biol Med (Maywood) ; 228(9): 1096-104, 2003 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-14530522

RESUMO

Small animal models with the capacity to support engraftment of a functional human immune system are needed to facilitate studies of human alloimmunity. In the present investigation, non-obese diabetic (NOD) severe combined immunodeficient (scid) beta2-microglobulin-null (B2mnull) mice engrafted with human peripheral blood lymphocytes (hu-PBL-NOD-scid B2mnull mice) were used as in vivo models for studying human skin allograft rejection. Hu-PBL-NOD-scid B2mnull mice were established by injection of human spleen cells or PBLs and transplanted with full-thickness allogeneic human skin. Human cell engraftment was enhanced by injection of anti-mouse CD122 antibody. The respective contributions of human CD4+ and CD8+ cells in allograft rejection were determined using depleting antibodies. Human skin grafts on unmanipulated NOD-scid B2mnull mice uniformly survived but on chimeric hu-PBL-NOD-scid B2mnull mice exhibited severe immune-mediated injury that often progressed to complete rejection. The alloaggressive hu-PBLs did not require prior in vitro sensitization to elicit targeted effector cell activity. Extensive mononuclear cell infiltration directed towards human-origin endothelium was associated with thrombosis and fibrin necrosis. No evidence of graft-versus-host disease was detected. Either CD4+ or CD8+ T cells may mediate injury and alloimmune rejection of human skin grafts on hu-PBL-NOD-scid B2mnull mice. It is proposed that Hu-PBL-NOD-scid B2mnull mice engrafted with human skin will provide a useful model for analysis of interventions designed to modulate human allograft rejection.


Assuntos
Doença Enxerto-Hospedeiro , Linfócitos/citologia , Transplante de Pele/imunologia , Animais , Complexo CD3/biossíntese , Antígenos CD4/biossíntese , Antígenos CD8/biossíntese , Feminino , Citometria de Fluxo , Rejeição de Enxerto , Humanos , Imuno-Histoquímica , Antígenos Comuns de Leucócito/biossíntese , Leucócitos Mononucleares/metabolismo , Linfócitos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Receptores de Interleucina-2/biossíntese , Baço/citologia , Microglobulina beta-2/genética
16.
Exp Hematol ; 31(3): 234-43, 2003 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-12644021

RESUMO

OBJECTIVE: Viable motheaten mice (abbreviated gene symbol me(v)) are deficient in SHP-1, a critical negative regulator of signal transduction in hematopoietic cells. These mice exhibit severe immune dysfunction accompanied by hyperproliferation of myeloid cells, widespread inflammatory lesions, and regenerative anemia. The aim of this study was to investigate the mechanisms underlying anemia in me(v)/me(v) mice. MATERIALS AND METHODS: Multiple hematologic parameters, osmotic fragility, and erythropoietin levels were measured to characterize the anemia in me(v)/me(v) mice. B-cell-deficient me(v)/me(v) Igh-6(null) mice were generated to assess the role of anti-erythrocyte antibodies. Coombs assays and flow cytometry were carried out for detection of anti-erythrocyte antibodies. Oxidant production by macrophages, glutathione levels, and lipid peroxidation products in erythrocytes were measured, as was the impact of oxidant on the ultrastructure of me(v)/me(v) erythrocytes. Erythroid maturation and erythrocyte plasma membrane integrity were assessed with flow cytometry by evaluating CD71 expression and annexin V labeling. RESULTS: The regenerative anemia of me(v)/me(v) mice was associated with erythrocyte changes that were independent of the presence of anti-erythrocyte antibodies. Erythrocytes from me(v)/me(v) mice had increased fragility and heightened susceptibility to oxidant damage. Macrophages from me(v)/me(v) mice demonstrated a higher basal level of oxidant production and enhanced production after stimulation. Oxidant damage in me(v)/me(v) erythrocytes was evidenced by a significant elevation of lipid peroxidation and diminished levels of glutathione. CONCLUSION: Our results support the hypothesis that as a consequence of severe inflammatory disease, me(v)/me(v) erythrocytes are subject to exceptionally high oxidative stress resulting in oxidation of phospholipids in the erythrocyte membrane with subsequent hemolysis.


Assuntos
Anemia Hemolítica/etiologia , Proteínas Tirosina Fosfatases/deficiência , Anemia Hemolítica/metabolismo , Anemia Hemolítica/patologia , Animais , Autoanticorpos , Membrana Eritrocítica/química , Membrana Eritrocítica/metabolismo , Eritrócitos/imunologia , Eritrócitos/metabolismo , Eritrócitos/ultraestrutura , Homozigoto , Inflamação/metabolismo , Inflamação/patologia , Peptídeos e Proteínas de Sinalização Intracelular , Peroxidação de Lipídeos , Lipídeos de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fragilidade Osmótica , Oxirredução , Estresse Oxidativo , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/genética , Proteínas Tirosina Fosfatases/fisiologia
17.
Genomics ; 81(1): 6-14, 2003 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-12573256

RESUMO

In the outcrossing of a new recessive mouse mutation causing hair loss, a new wavy-coated phenotype appeared. The two distinct phenotypes were shown to be alternative manifestations of the same gene mutation and attributable to a single modifier locus. The new mutation, curly bare (cub), was mapped to distal Chr 11 and the modifier (mcub) was mapped to Chr 5. When homozygous for the recessive mcub allele, cub/cub mice appear hairless. A single copy of the dominant Mcub allele confers a full, curly coat in cub/cub mice. Reciprocal transfer of full-thickness skin grafts between mutant and control animals showed that the skin phenotype was tissue autonomous. The hairless cub/cub mcub/mcub mice show normal contact sensitivity responses to oxazolone. The similarity of the wavy coat phenotype to those of Tgfa and Egfr mutations and the map positions of cub and mcub suggest candidate genes that interact in the EGF receptor signal transduction pathway.


Assuntos
Mapeamento Cromossômico , Genes Reguladores/genética , Cabelo/anormalidades , Animais , Cruzamentos Genéticos , Cabelo/fisiologia , Hipersensibilidade/genética , Camundongos , Transplante de Pele
18.
Hum Mol Genet ; 12(1): 61-9, 2003 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-12490533

RESUMO

The nature of the wild-type gene product at the mouse ichthyosis (ic) locus has been of great interest because mutations at this locus cause marked abnormalities in nuclear heterochromatin, similar to those observed in Pelger-Huët anomaly (PHA). We recently found that human PHA is caused by mutations in the gene (LBR) encoding lamin B receptor, an evolutionarily conserved inner nuclear membrane protein involved in nuclear assembly and chromatin binding. Mice homozygous for deleterious alleles at the ichthyosis (ic) locus present with a blood phenotype similar to PHA, and develop other phenotypic abnormalities, including alopecia, variable expression of syndactyly and hydrocephalus. The ic locus on mouse chromosome 1 shares conserved synteny with the chromosomal location of the human LBR locus on human chromosome 1. In this study, we identified one nonsense (815ins) and two frameshift mutations (1088insCC and 1884insGGAA) within the Lbr gene of mice homozygous for either of three independent mutations (ic, ic(J) and ic(4J), respectively) at the ichthyosis locus. These allelic mutations are predicted to result in truncated or severely impaired LBR protein. Our studies of mice homozygous for the ic(J) mutation revealed a complete loss of LBR protein as shown by immunofluorescence microscopy and immunoblotting. The findings provide the molecular basis for the heterochromatin clumping and other distinct phenotypes caused by ic mutations. These spontaneous Lbr mutations confirm the molecular basis of human PHA and provide a small animal model for determination of the precise function of LBR in normal and pathological states.


Assuntos
Ictiose/genética , Mutação , Anomalia de Pelger-Huët/genética , Receptores Citoplasmáticos e Nucleares/genética , Animais , Modelos Animais de Doenças , Genótipo , Humanos , Camundongos , Microscopia Eletrônica , Anomalia de Pelger-Huët/patologia , Fenótipo , Análise de Sequência de DNA , Receptor de Lamina B
19.
In Vivo ; 16(4): 223-8, 2002.
Artigo em Inglês | MEDLINE | ID: mdl-12224130

RESUMO

In this paper we describe the establishment and characterization of a transplantable cell line derived from a spontaneously occurring chondroblastic osteosarcoma in a C57BL/6J mouse. The tumor line, MOS-J, forms solid tumors when injected intramuscularly into immunocompetent syngeneic hosts, mimicking endochondral bone development. These transplantable tumors have the capacity to destroy and invade existing bone and invade vessels in close proximity to the tumor. In culture, MOS-J cells form layers of pleomorphic cells with high mitotic activity. These cells have marked alkaline phosphatase activity and form calcified foci in vitro that stain with alizarin red. MOS-J cells also promote osteoclast development in vitro from normal bone marrow cells. These characteristics indicate the potential utility of the MOS-J osteosarcoma cell line as a model for studies of human osteosarcoma and bone biology.


Assuntos
Neoplasias Ósseas/patologia , Transplante de Neoplasias , Osteossarcoma/patologia , Fosfatase Alcalina/metabolismo , Animais , Antraquinonas , Neoplasias Ósseas/diagnóstico por imagem , Diferenciação Celular , Divisão Celular , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos SCID , Microscopia Eletrônica , Osteoclastos/citologia , Osteossarcoma/diagnóstico por imagem , Radiografia , Coloração e Rotulagem , Células Tumorais Cultivadas/transplante , Células Tumorais Cultivadas/ultraestrutura
20.
J Autoimmun ; 18(2): 105-17, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11908943

RESUMO

Mice homozygous for the viable motheaten (Hcph(me-v)) mutation are deficient in SHP-1 protein-tyrosine phosphatase, resulting in severe systemic autoimmunity and immune dysfunction. A high percentage of B-cells in viable motheaten mice express the cell surface glycoprotein CD5, in contrast to wild type mice that express CD5 on only a small percentage of B-cells. CD5(+) B-cells have been associated with autoantibody production. To determine the role of CD5 in the development of the inflammatory disease in me(v)/ me(v) mice, we created a stock of CD5(null)me(v)/ me(v) mice. The longevity of CD5(null)me(v)/ me(v) mice was increased 69% in comparison to me(v)/ me(v) mice on a similar (B6;129) background. The increased lifespan was associated with a marked reduction in pulmonary inflammation. Flow cytometry analysis of spleen cells from CD5(null)me(v)/ me(v) mice at 9-12 weeks of age revealed significant decreases in percentages of IgM/B220 double positive B-cells, Mac-1/Gr-1 double positive cells and CD4(+) T-cells compared with me(v)/ me(v) mice. CD5(null)me(v)/ me(v) mice also had significantly lower serum IgM levels in comparison to me(v)/ me(v) mice. Study of CD5(null)me(v)/ me(v) mice may provide further insight into the role of CD5 in cell signaling and may help explain the observed association of CD5(+) B-cells with autoimmune disease.


Assuntos
Antígenos CD5/metabolismo , Pneumonia/etiologia , Pneumonia/imunologia , Proteínas Tirosina Fosfatases/deficiência , Animais , Autoanticorpos/sangue , Contagem de Células Sanguíneas , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Antígenos CD5/genética , Feminino , Imunoglobulinas/sangue , Peptídeos e Proteínas de Sinalização Intracelular , Longevidade , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes , Pneumonia/enzimologia , Pneumonia/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Proteínas Tirosina Fosfatases/genética , Baço/imunologia , Baço/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...