Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 485
Filtrar
1.
Immunol Lett ; 237: 3-10, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34174253

RESUMO

Many studies of the autoimmune disease Sjögren's syndrome have been performed using spontaneous mouse models. In the present study, we describe the characteristics of McH/lpr-RA1 mice and propose their use as a novel murine model of autoimmune sialadenitis. The McH/lpr-RA1 mouse is a recombinant congenic strain derived from generation F54 or more of MRL-Faslpr x (MRL- Faslpr x C3H- Faslpr) F1. We show for the first time that this mouse spontaneously develops autoimmune sialadenitis and vasculitis in submandibular gland tissues. Sialadenitis was accompanied by extensive inflammatory cell infiltration and tissue destruction. Immunohistochemical studies revealed that the salivary gland lesions strongly expressed four sialadenitis-related molecules: SSA and SSB (autoantigens of Sjögren's syndrome), gp91phox (an accelerator of reactive oxygen species production) and single strand DNA (a marker of apoptotic cells). In contrast, expression of aquaporin-5 (AQP5), which stimulates salivary secretion was weak or negligible. Statistical correlation analyses indicated that the apoptosis of salivary gland cells provoked by oxidative stress contributed to the severe sialadenitis and reduced expression of AQP5. Our study has demonstrated that McH/lpr-RA1 mice spontaneously develop the pathognomonic features of autoimmune sialadenitis and thus could be used as a new animal model of Sjögren's syndrome.


Assuntos
Doenças Autoimunes/imunologia , Modelos Animais de Doenças , Camundongos Endogâmicos/imunologia , Camundongos Mutantes/imunologia , Sialadenite/imunologia , Síndrome de Sjogren , Vasculite/imunologia , Animais , Animais Congênicos , Apoptose , Aquaporina 5/biossíntese , Aquaporina 5/genética , Autoantígenos/biossíntese , Autoantígenos/genética , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , DNA de Cadeia Simples/análise , Feminino , Predisposição Genética para Doença , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos/genética , Camundongos Mutantes/genética , NADPH Oxidase 2/biossíntese , NADPH Oxidase 2/genética , Ribonucleoproteínas/biossíntese , Ribonucleoproteínas/genética , Índice de Gravidade de Doença , Sialadenite/genética , Sialadenite/patologia , Síndrome de Sjogren/genética , Síndrome de Sjogren/imunologia , Glândula Submandibular/metabolismo , Glândula Submandibular/patologia , Vasculite/genética , Vasculite/patologia , Antígeno SS-B
2.
Nat Commun ; 9(1): 3151, 2018 08 08.
Artigo em Inglês | MEDLINE | ID: mdl-30089795

RESUMO

γδ T cells have many known functions, including the regulation of antibody responses. However, how γδ T cells control humoral immunity remains elusive. Here we show that complete Freund's adjuvant (CFA), but not alum, immunization induces a subpopulation of CXCR5-expressing γδ T cells in the draining lymph nodes. TCRγδ+CXCR5+ cells present antigens to, and induce CXCR5 on, CD4 T cells by releasing Wnt ligands to initiate the T follicular helper (Tfh) cell program. Accordingly, TCRδ-/- mice have impaired germinal center formation, inefficient Tfh cell differentiation, and reduced serum levels of chicken ovalbumin (OVA)-specific antibodies after CFA/OVA immunization. In a mouse model of lupus, TCRδ-/- mice develop milder glomerulonephritis, consistent with decreased serum levels of lupus-related autoantibodies, when compared with wild type mice. Thus, modulation of the γδ T cell-dependent humoral immune response may provide a novel therapy approach for the treatment of antibody-mediated autoimmunity.


Assuntos
Diferenciação Celular , Imunidade Humoral/imunologia , Linfócitos Intraepiteliais/imunologia , Linfócitos Intraepiteliais/fisiologia , Linfócitos T Auxiliares-Indutores/imunologia , Linfócitos T Auxiliares-Indutores/fisiologia , Compostos de Alúmen , Animais , Formação de Anticorpos , Autoanticorpos/sangue , Galinhas , Feminino , Adjuvante de Freund/imunologia , Glomerulonefrite , Imunização , Glomérulos Renais/imunologia , Glomérulos Renais/patologia , Linfonodos/imunologia , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes/imunologia , Modelos Animais , Modelos Imunológicos , Ovalbumina/sangue , Ovalbumina/imunologia , Receptores CXCR5/metabolismo
3.
Med Sci (Paris) ; 30(11): 949-51, 2014 Nov.
Artigo em Francês | MEDLINE | ID: mdl-25388574
4.
Glycobiology ; 23(3): 363-80, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23118208

RESUMO

The consortium for functional glycomics (CFG) was a large research initiative providing networking and resources for investigators studying the role of glycans and glycan-binding proteins in health and disease. Starting in 2001, six scientific cores were established to generate data, materials and new technologies. By the end of funding in 2011, the mouse phenotype core (MPC) submitted data to a website from the phenotype screen of 36 mutant mouse strains deficient in a gene for either a glycan-binding protein (GBP) or glycosyltransferase (GT). Each mutant strain was allotted three months for analysis and screened by standard phenotype assays used in the fields of immunology, histology, hematology, coagulation, serum chemistry, metabolism and behavior. Twenty of the deficient mouse strains had been studied in other laboratories, and additional tests were performed on these strains to confirm previous observations and discover new data. The CFG constructed 16 new homozygous mutant mouse strains and completed the initial phenotype screen of the majority of these new mutant strains. In total, >300 phenotype changes were observed, but considering the over 100 assays performed on each strain, most of the phenotypes were unchanged. Phenotype differences include abnormal testis morphology in GlcNAcT9- and Siglec-H-deficient mice and lethality in Pomgnt1-deficient mice. The numerous altered phenotypes discovered, along with the consideration of the significant findings of normality, will provide a platform for future characterization to understand the important roles of glycans and GBPs in the mechanisms of health and disease.


Assuntos
Glicosiltransferases/genética , Lectinas/genética , Camundongos Mutantes/genética , Fenótipo , Animais , Marcação de Genes , Homozigoto , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes/anatomia & histologia , Camundongos Mutantes/imunologia , Camundongos Mutantes/fisiologia , Mutação
5.
J Stud Alcohol Drugs ; 73(6): 933-7, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23036211

RESUMO

OBJECTIVE: The effects of alcohol (ethanol) are well documented and contribute to significant health problems and financial burden on the health care system. Several mouse models have been described that facilitate studies of the effects of alcohol on the mouse immune system. Our goal was to establish a chronic alcohol mouse model using the immunodeficient NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) mouse. This severely immunodeficient model has previously been shown to allow efficient engraftment of human hematopoietic repopulating cells and cancer cells, thereby facilitating diverse studies on human hematopoiesis, immune cell function, and oncogenesis in vivo. METHOD: NSG mice were provided ethanol in their drinking water as the only available fluid, starting at 5% weight/volume (w/v) and subsequently were increased to 10%, 15%, and 20% w/v. Mice were then maintained at 20% w/v, a level that models chronic alcohol use in humans. Alcohol consumption and weight were monitored. RESULTS: NSG mice readily consumed alcohol throughout the study and showed no adverse effects. No significant difference between group mean weights was identified the day before increasing the ethanol dose or at the end of 5 weeks at 20% w/v (p > .28). While the mice were maintained at 20% w/v ethanol, the mean daily ethanol intake was 27.2 g/kg of body weight, 32% of caloric intake. CONCLUSIONS: Here we have established a chronic alcohol mouse model using the powerful immunodeficient NSG mouse. This model should allow for novel studies on the effects of alcohol on engrafted human cells, including studies on the effects of alcohol on hematopoiesis, immunity, and cancer.


Assuntos
Consumo de Bebidas Alcoólicas/fisiopatologia , Consumo de Bebidas Alcoólicas/psicologia , Alcoolismo/psicologia , Modelos Animais de Doenças , Camundongos Mutantes/psicologia , Alcoolismo/fisiopatologia , Animais , Peso Corporal/efeitos dos fármacos , Peso Corporal/fisiologia , Ingestão de Alimentos/efeitos dos fármacos , Ingestão de Alimentos/fisiologia , Ingestão de Energia/efeitos dos fármacos , Ingestão de Energia/fisiologia , Feminino , Camundongos , Camundongos Mutantes/imunologia , Camundongos Mutantes/fisiologia
6.
Med Sci (Paris) ; 28(1): 63-8, 2012 Jan.
Artigo em Francês | MEDLINE | ID: mdl-22289832

RESUMO

The study of human pathologies is often limited by the absence of animal models which are robust, cost-effective and reproduce the hallmarks of human infections. While mice have been frequently employed to study human diseases, many of important pathogens display unique human tropism. These last two decades the graft of human progenitor cells or tissues into -immunodeficient mice has allowed the elaboration of so called humanized mice. Humanized mouse technology has made rapid progress, and it is now possible to achieve high levels of human chimerism in various organs and tissues, particularly the immune system and the liver. The review briefly summarizes the different models of humanized mice available for in vivo experiments. With a focus on lymphotropic, monocytotropic and hepatotropic viruses, we here discuss the current status and future prospects of these models for studying the pathogenesis of infectious diseases. Furthermore, they provide a powerful tool for the development of innovative therapies.


Assuntos
Quimera , Doenças Transmissíveis , Modelos Animais de Doenças , Camundongos Mutantes , Animais , Quimera/genética , Quimera/imunologia , Cruzamentos Genéticos , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Previsões , Hepatócitos/transplante , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Subunidade gama Comum de Receptores de Interleucina/deficiência , Subunidade gama Comum de Receptores de Interleucina/genética , Fígado/embriologia , Transplante de Fígado , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Mutantes/genética , Camundongos Mutantes/imunologia , Quimera por Radiação , Especificidade da Espécie , Timo/embriologia , Timo/transplante , Transplante Heterólogo , Viroses/tratamento farmacológico , Viroses/imunologia , Viroses/virologia
7.
J Immunother ; 30(3): 338-49, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17414325

RESUMO

Humanized antibodies (Abs) are effective drugs against a variety of diseases such as cancer, autoimmune diseases, transplant rejection and others. The most powerful technology to develop humanized Abs is the use of mice that produce humanized Abs. By modifying the genetic background of F004 mice a new mouse substrain was developed for optimized "one step" generation of chimeric humanized monoclonal Abs. The new mice (F004-Jen) demonstrated improved fertility still expressing the human locus at the same level as the parental F004 mouse. The value of these mice for the generation of chimeric Abs was exemplified for a panel of chimeric Abs against the human neural cell adhesion molecule (NCAM): The fully chimeric human IgG1/kappa Ab Ch.MK1 bound to NCAM expressing cells with a K(D)=4.3-8.7 x 10(-8) M and was functionally active as demonstrated by depleting NCAM expressing cells. We also demonstrated that chimeric IgG1/kappa Abs can be induced by hybridoma class switching of IgM producing hybridoma cells, providing an alternative way to chimeric Abs. The present data highlight F004-Jen mice as an efficient tool for "one step" generation of chimeric Abs.


Assuntos
Anticorpos Monoclonais/biossíntese , Camundongos Mutantes/imunologia , Proteínas Mutantes Quiméricas/metabolismo , Moléculas de Adesão de Célula Nervosa/imunologia , Animais , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/farmacologia , Linhagem Celular Tumoral , Humanos , Hibridomas , Switching de Imunoglobulina , Regiões Constantes de Imunoglobulina/genética , Regiões Constantes de Imunoglobulina/metabolismo , Imunoglobulina G/genética , Imunoglobulina G/metabolismo , Imunoglobulina M/genética , Imunoglobulina M/metabolismo , Cadeias kappa de Imunoglobulina/genética , Cadeias kappa de Imunoglobulina/metabolismo , Camundongos , Camundongos Mutantes/genética , Proteínas Mutantes Quiméricas/genética , Proteínas Mutantes Quiméricas/farmacologia , Moléculas de Adesão de Célula Nervosa/análise , Reprodução
8.
Vet Pathol ; 43(4): 401-23, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16846982

RESUMO

Development of the primary and secondary lymphoid organs is a tightly controlled process. These tissues are highly organized to maximize efficiency of the immune response. Spontaneous and targeted mutations in laboratory mice have led to better understanding of the molecular interactions and signaling pathways essential to the development and organization of lymphoid tissues, and the functional consequences of loss or disruption of the normal structures. On the basis of studies of mutations in mice and other species, it has been determined that a wild-type allele of the Foxn1 gene is required for normal thymic development and function. The Tlx1, Bapx1, Tcf21, Wt1 and Dh genes are essential for development of the spleen, while mutations of Nkx2-3, Lta, Ltb, Ltbr, Map3k14, Relb, Tnf, Tnfrsf1a, Cxcl13, Blr1 (Cxcr5), or cpdm genes result in disruption of normal splenic microarchitecture. The requirements for organized lymph nodes vary according to anatomic location, but most rely on Id2 (Idb2) and Rorc, in addition to lymphotoxins and Tnfrsf11a, Tnfsf11, Relb, Map3k14, Cxcl13, and Blr1 genes. Development of Peyer's patches is dependent on Id2 and Rorc genes, lymphotoxins, and Relb, Map3k14, Il7r, and cpdm genes. Less is known about the requirements for nasal-associated lymphoid tissues (NALT), but Id2 is a requirement. Here we review abnormalities of lymphoid organ development in immunodeficient mutant mice, including spontaneous and targeted mutations of Id2, Rorc, Tnf, Tnfrsf1a, Lta, Ltb, Ltbr, Tnfrsf11a, Tnfsf11, Relb, Map3k14, IL7r, Blr1, and Cxcl13 genes.


Assuntos
Síndromes de Imunodeficiência/patologia , Tecido Linfoide/crescimento & desenvolvimento , Camundongos Mutantes/imunologia , Animais , Humanos , Síndromes de Imunodeficiência/genética , Síndromes de Imunodeficiência/imunologia , Tecido Linfoide/imunologia , Camundongos , Camundongos Mutantes/genética
10.
J Immunol Methods ; 257(1-2): 137-43, 2001 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-11687247

RESUMO

Many commercially and privately available congenic strains of laboratory animals were founded decades ago and are likely to differ from one another by dozens of fixed mutational differences at background loci. This problem is often ignored despite growing evidence that such background variation exists. Eliminating this confounding variation can be largely accomplished by crossing congenic strains to produce F2 segregants that are homozygous (or heterozygous) for relevant genes. Discriminating F2 homozygotes can be difficult when strain differences are minor, as are mutant mouse strains differing at single major histocompatibility loci (H2 mutant congenics). Here, we describe a two-step polymerase chain reaction (PCR) method utilizing heteroduplex analysis and sequence specific primers (SSP-PCR) that efficiently discriminates the F2 progeny of two such H2 mutant congenic mice crosses (bm1xB6 and bm1xbm3). A third H2 mutant cross cannot be resolved by heteroduplexing, but is discriminated (albeit less efficiently) with SSP-PCR alone. This sensitive application can be extended to any congenic mutant strains.


Assuntos
Variação Genética , Antígenos H-2/genética , Camundongos Congênicos/genética , Camundongos Congênicos/imunologia , Camundongos Mutantes/genética , Camundongos Mutantes/imunologia , Alelos , Animais , Sequência de Bases , Cruzamentos Genéticos , Primers do DNA/genética , Feminino , Haplótipos , Heterozigoto , Homozigoto , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase
11.
Cancer Res ; 61(11): 4561-8, 2001 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-11389091

RESUMO

ATM, the gene mutated in the human immunodeficiency disorder ataxia-telangiectasia (A-T), plays a central role in recognizing ionizing radiation damage in DNA and in controlling several cell cycle checkpoints. We describe here a murine model in which a nine-nucleotide in-frame deletion has been introduced into the Atm gene by homologous recombination followed by removal of the selectable marker cassette by Cre-loxP site-specific, recombination-mediated excision. This mouse, Atm-DeltaSRI, was designed as a model of one of the most common deletion mutations (7636del9) found in A-T patients. The murine Atm deletion results in the loss of three amino acid residues (SRI; 2556-2558) but produces near full-length detectable Atm protein that lacks protein kinase activity. Radiosensitivity was observed in Atm-DeltaSRI mice, whereas the immunological profile of these mice showed greater heterogeneity of T-cell subsets than observed in Atm(-/-) mice. The life span of Atm-DeltaSRI mice was significantly longer than that of Atm(-/-) mice when maintained under nonspecific pathogen-free conditions. This can be accounted for by a lower incidence of thymic lymphomas in Atm-DeltaSRI mice up to 40 weeks, after which time the animals died of other causes. The thymic lymphomas in Atm-DeltaSRI mice were characterized by extensive apoptosis, which appears to be attributable to an increased number of cells expressing Fas ligand. A variety of other tumors including B-cell lymphomas, sarcomas, and carcinomas not seen in Atm(-/-) mice were observed in older Atm-DeltaSRI animals. Thus, expression of mutant protein in Atm-DeltaSRI knock-in mice gives rise to a discernibly different phenotype to Atm(-/-) mice, which may account for the heterogeneity seen in A-T patients with different mutations.


Assuntos
Camundongos Mutantes/genética , Proteínas Serina-Treonina Quinases/genética , Deleção de Sequência , Animais , Apoptose/genética , Ataxia Telangiectasia/genética , Proteínas Mutadas de Ataxia Telangiectasia , Sequência de Bases , Proteínas de Ciclo Celular , Cruzamentos Genéticos , DNA/genética , Proteínas de Ligação a DNA , Feminino , Humanos , Linfoma/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Mutantes/crescimento & desenvolvimento , Camundongos Mutantes/imunologia , Mutagênese Sítio-Dirigida , Fenótipo , Neoplasias do Timo/genética , Proteínas Supressoras de Tumor , Regulação para Cima
13.
Thromb Haemost ; 85(1): 125-33, 2001 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-11204564

RESUMO

Mice genetically deficient in factor VII (fVIII) are a model of hemophilia A. As a first step to reproduce in this mouse model what occurs over time in hemophilia A patients treated with human fVIII (hfVIII), we have investigated the time course and the characteristics of their immune response to hfVIII, after multiple intravenous injections. Anti-hfVIII antibodies appeared after four to five injections. They were IgG1 and to a lesser extent IgG2, indicating that they were induced by both Th2 and Th1 cells. Inhibitors appeared after six injections. CD4+ enriched splenocytes from hfVIII-treated mice proliferated in response to fVIII and secreted IL-10: in a few mice they secreted also IFN-gamma and in one mouse IL-4, but never IL-2. A hfVIII-specific T cell line derived from hfVIII-treated mice secreted both IL-4 and IFN-gamma, suggesting that it included both Th1 and Th2 cells. CD4+ enriched splenocytes of hfIII-treated mice recognized all hfVIII domains. Thus, hemophilic mice develop an immune response to hfVIII administered intravenously similar to that of hemophilia A patients. Their anti-hfVIII antibodies can be inhibitors and belong to IgG subclasses homologous to those of inhibitors in hemophilic patients; their anti-hfVIII CD4+ cells recognize a complex repertoire and both Th1 and Th2 cytokines, and especially IL-10, may drive the antibody synthesis.


Assuntos
Fator VIII/imunologia , Hemofilia A/sangue , Camundongos Mutantes/imunologia , Animais , Anticorpos/análise , Anticorpos/sangue , Células Produtoras de Anticorpos/efeitos dos fármacos , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Divisão Celular/efeitos dos fármacos , Citocinas/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Epitopos/análise , Fator VIII/administração & dosagem , Fator VIII/farmacologia , Hemofilia A/imunologia , Humanos , Imunoglobulina G/sangue , Injeções Intravenosas , Interleucina-10/metabolismo , Camundongos , Baço/citologia , Baço/imunologia , Fatores de Tempo
14.
Comp Med ; 51(4): 300-13, 2001 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-11924787

RESUMO

Studies of immune deficiencies have a history as long as that of immunology. However, reports of two key spontaneous recessive mutations in mice (nude in 1966-1968 and scid in 1983) laid the foundations for widespread application of immune-deficient rodents to a broad range of research topics. More recently, technologies modifying the mouse genome by transgenesis, gene ablation and crossbreeding for lines with multiple immune deficits have provided a large number of new types of immunologically impaired mice. The primary goals of this overview are to help non-immunologists understand key differences between some of the immunodeficient strains, develop an appreciation for the value of information derived from immunodeficient mouse-based research and to encourage expanded, creative use of these specialized research animals. Secondary goals are to promote greater awareness of unexpected outcomes that can arise when working with genetically immune-deficient mice, the need for vigilance in maintaining these research animals, and the care required in interpretation of the data that immune-deficient modeling provides. Two illustrations on developing appropriate immune deficient animal models for a new research application conclude the review.


Assuntos
Modelos Animais de Doenças , Síndromes de Imunodeficiência/genética , Camundongos Mutantes/imunologia , Animais , Diferenciação Celular , Proteínas de Ligação a DNA/deficiência , Proteínas de Ligação a DNA/genética , Doenças do Cão/parasitologia , Cães , Feminino , Genes RAG-1 , Vida Livre de Germes , Interações Hospedeiro-Parasita , Sistema Imunitário/citologia , Sistema Imunitário/fisiologia , Síndromes de Imunodeficiência/classificação , Síndromes de Imunodeficiência/imunologia , Subpopulações de Linfócitos/imunologia , Subpopulações de Linfócitos/patologia , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes/genética , Camundongos Nus , Camundongos SCID , Camundongos Transgênicos , Infestações por Ácaros/parasitologia , Infestações por Ácaros/veterinária , Ácaros/crescimento & desenvolvimento , Gravidez , Complicações na Gravidez/imunologia , Organismos Livres de Patógenos Específicos , Transplante Heterólogo , Útero/imunologia
15.
Scand J Immunol ; 52(6): 555-62, 2000 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11119260

RESUMO

The T-cell repertoire is shaped by the positive and negative selection of immature CD4(+) CD8(+) double positive (DP) thymocytes. Positive selection of DP T cells to the CD4(+) CD8(-) and CD4(-) CD8(+) simple positive (SP) lineages is a multistep process which involves cellular interactions between thymocytes and stromal cells. Mutant nackt (nkt/nkt) mice have been shown to have a deficiency in the CD4(+) CD8(-) T-cell subset both in the thymus and in the periphery. The present report suggests that nkt/nkt mice present alterations in early steps of positive selection because they show decreases in the percentages of CD69(+) and CD5(+) cells within the DP subset. Experiments involving bone marrow transfer and thymic chimeras demonstrate that the thymic epithelium of nkt/nkt mice is involved in the alterations registered during positive selection and dictates the ultimate fate of CD4(+) SP cells.


Assuntos
Antígenos CD4 , Camundongos Mutantes/imunologia , Subpopulações de Linfócitos T/imunologia , Timo/imunologia , Alopecia/genética , Animais , Antígenos CD , Antígenos de Diferenciação de Linfócitos T , Antígenos CD5 , Antígenos CD8 , Imunidade Celular/imunologia , Lectinas Tipo C , Camundongos
17.
Transplantation ; 69(8): 1683-9, 2000 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-10836381

RESUMO

BACKGROUND: Nonlytic anti-CD4 monoclonal antibody therapy can be used to induce transplantation tolerance in rodent models. Such tolerance is often associated with dominant regulation, mediated by CD4+ cells, and characterized by infectious tolerance and linked suppression. Understanding the mechanisms by which CD4+ regulatory cells function may improve the manner in which current immunosuppressants are applied and may lead to the development of new tolerance-inducing therapeutics. Fas-mediated apoptosis has been characterized as an important mechanism of peripheral self-tolerance and we here examine whether it has any role in anti-CD4 monoclonal antibody-induced dominant tolerance. METHODS: Tolerance to transplanted skin and bone marrow, mismatched for multiple minor histocompatibility antigens, was induced in Fas mutant and control mice using anti-CD4 and anti-CD8 monoclonal antibodies. To test for linked suppression, animals were transplanted with a second graft-bearing tolerated and third party antigens. The ability of splenocytes from tolerant animals to suppress graft rejection was assessed by transfer into partially immunocompromised recipients. RESULTS: Monoclonal antibody therapy rendered Fas mutant mice tolerant of minor disparate skin and bone marrow. Splenocytes from these and control tolerant animals when transferred into partially immunocompromised Fas mutant or control recipients, induced antigen-specific suppression of graft rejection. Additionally, tolerant Fas mutant mice accepted grafts bearing tolerated and third party antigens. CONCLUSIONS: Signal transduction through the Fas receptor plays no essential role in the induction of tolerance using anti-CD4 and anti-CD8 monoclonal antibodies or its maintenance by active regulation.


Assuntos
Anticorpos Monoclonais/uso terapêutico , Genes Dominantes , Tolerância Imunológica/genética , Terapia de Imunossupressão , Glicoproteínas de Membrana/fisiologia , Receptor fas/fisiologia , Animais , Transplante de Medula Óssea/imunologia , Antígenos CD4/imunologia , Transplante de Células , Proteína Ligante Fas , Rejeição de Enxerto/prevenção & controle , Histocompatibilidade , Camundongos , Camundongos Endogâmicos/imunologia , Camundongos Mutantes/imunologia , Transdução de Sinais , Transplante de Pele/imunologia , Baço/citologia
18.
Nat Med ; 6(5): 589-93, 2000 May.
Artigo em Inglês | MEDLINE | ID: mdl-10802718

RESUMO

Systemic infection with Listeria monocytogenes, a Gram-positive intracellular bacterium, has been used extensively to analyze the innate immune response. Macrophages are central to this response, acting as both the host for and principal defense against this bacterium. During pregnancy L. monocytogenes has a predilection for replication at the maternal-placental interface and consequently is an important cause of fetal morbidity and mortality. However, macrophages are mostly excluded from the murine placenta with neutrophils acting as the main immune effector cell against this bacterium. Colony stimulating factor (CSF)-1, a macrophage growth factor, is synthesized in high concentrations by the uterine epithelium during pregnancy, where it is targeted to trophoblast bearing CSF-1-receptors. To define the involvement of CSF-1 in placental immunity, we infected pregnant mice either homozygous or heterozygous for an inactivating recessive mutation in the gene for CSF-1 (osteopetrotic; Csfmop) with L. monocytogenes. CSF-1 was required to recruit neutrophils to the site of listerial infection in the decidua basalis, and infection by Listeria remained unrestrained in its absence. CSF-1 acted by inducing the trophoblast to synthesize the neutrophil chemoattractants (KC) and macrophage inflammatory protein (MIP)-2. Thus, during pregnancy, trophoblast responsive to CSF-1 acts to organize the maternal immune response to bacterial infection at the utero-placental interface. This previously unknown function indicates that the trophoblast acts as a pregnancy-specific component of the innate immune system.


Assuntos
Listeriose/imunologia , Fator Estimulador de Colônias de Macrófagos/genética , Camundongos Mutantes/imunologia , Placenta/imunologia , Prenhez/imunologia , Trofoblastos/imunologia , Animais , Quimiocinas/análise , Quimiotaxia de Leucócito/imunologia , Citocinas/análise , Feminino , Camundongos , Neutrófilos/imunologia , Placenta/microbiologia , Gravidez , Células Th1 , Células Th2
20.
J Exp Med ; 191(5): 899-906, 2000 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-10704470

RESUMO

The combination of hemorrhagic pneumonitis and rapidly progressive glomerulonephritis is a characteristic feature of Goodpasture's syndrome (GPS), an autoimmune disease resulting from the interaction of pathogenic anti-collagen type IV (C-IV) antibodies with alveolar and glomerular basement membranes. Lack of a suitable animal model for this fatal disease has hampered both a basic understanding of its etiology and the development of therapeutic strategies. We now report a novel model for GPS using mice deficient in a central regulatory receptor for immunoglobulin (Ig)G antibody expression and function, the type IIB Fc receptor for IgG (FcgammaRIIB). Mutant mice immunized with bovine C-IV reproducibly develop massive pulmonary hemorrhage with neutrophil and macrophage infiltration and crescentic glomerulonephritis. The distinctive linear, ribbon-like deposition of IgG immune complex seen in GPS was observed along the glomerular and tubulointerstitial membranes of diseased animals. These results highlight the role of FcgammaRIIB in maintaining tolerance and suggest that it may play a role in the pathogenesis of human GPS.


Assuntos
Doença Antimembrana Basal Glomerular/imunologia , Antígenos CD/genética , Colágeno/imunologia , Modelos Animais de Doenças , Camundongos Mutantes/imunologia , Receptores de IgG/genética , Animais , Doença Antimembrana Basal Glomerular/genética , Autoanticorpos/sangue , Hemorragia , Glomérulos Renais/patologia , Pulmão/patologia , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA