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.
Science ; 371(6533): 1019-1025, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33674488

RESUMO

In vivo models that recapitulate human erythropoiesis with persistence of circulating red blood cells (RBCs) have remained elusive. We report an immunodeficient murine model in which combined human liver and cytokine humanization confer enhanced human erythropoiesis and RBC survival in the circulation. We deleted the fumarylacetoacetate hydrolase (Fah) gene in MISTRG mice expressing several human cytokines in place of their murine counterparts. Liver humanization by intrasplenic injection of human hepatocytes (huHep) eliminated murine complement C3 and reduced murine Kupffer cell density. Engraftment of human sickle cell disease (SCD)-derived hematopoietic stem cells in huHepMISTRGFah -/- mice resulted in vaso-occlusion that replicated acute SCD pathology. Combined liver-cytokine-humanized mice will facilitate the study of diseases afflicting RBCs, including bone marrow failure, hemoglobinopathies, and malaria, and also preclinical testing of therapies.


Assuntos
Anemia Falciforme/sangue , Circulação Sanguínea , Modelos Animais de Doenças , Eritrócitos/citologia , Eritropoese/fisiologia , Camundongos , Animais , Citocinas/metabolismo , Eritropoese/genética , Feminino , Deleção de Genes , Células-Tronco Hematopoéticas/citologia , Humanos , Hidrolases/genética , Fígado/fisiologia , Camundongos Mutantes , Pessoa de Meia-Idade
2.
Blood ; 129(8): 959-969, 2017 02 23.
Artigo em Inglês | MEDLINE | ID: mdl-28077418

RESUMO

Humanized mice are a powerful tool for the study of human hematopoiesis and immune function in vivo. However, the existing models cannot support robust adaptive immune responses, especially the generation of class-switched, antigen-specific antibody responses. Here we describe a new mouse strain, in which human interleukin 6 (IL-6) gene encoding the cytokine that is important for B- and T-cell differentiation was knocked into its respective mouse locus. The provision of human IL-6 not only enhanced thymopoiesis and periphery T-cell engraftment, but also significantly increased class switched memory B cells and serum immunoglobulin G (IgG). In addition, immunization with ovalbumin (OVA) induced OVA-specific B cells only in human IL-6 knock-in mice. These OVA-specific antibodies displayed the highest frequency of somatic mutation, further suggesting that human IL-6 is important for efficient B-cell activation and selection. We conclude that human IL-6 knock-in mice represent a novel and improved model for human adaptive immunity without relying on complex surgery to transplant human fetal thymus and liver. These mice can therefore be used to exploit or evaluate immunization regimes that would be unethical or untenable in humans.


Assuntos
Imunidade Adaptativa , Formação de Anticorpos , Técnicas de Introdução de Genes , Switching de Imunoglobulina , Interleucina-6/genética , Animais , Linfócitos B/citologia , Linfócitos B/imunologia , Galinhas , Expressão Gênica , Técnicas de Introdução de Genes/métodos , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Imunização , Imunoglobulina G/imunologia , Interleucina-6/imunologia , Camundongos , Ovalbumina/imunologia , Linfócitos T/citologia , Linfócitos T/imunologia
3.
Nat Med ; 22(11): 1351-1357, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27723723

RESUMO

Most human cancers, including myeloma, are preceded by a precursor state. There is an unmet need for in vivo models to study the interaction of human preneoplastic cells in the bone marrow microenvironment with non-malignant cells. Here, we genetically humanized mice to permit the growth of primary human preneoplastic and malignant plasma cells together with non-malignant cells in vivo. Growth was largely restricted to the bone marrow, mirroring the pattern in patients with myeloma. Xenografts captured the genomic complexity of parental tumors and revealed additional somatic changes. Moreover, xenografts from patients with preneoplastic gammopathy showed progressive growth, suggesting that the clinical stability of these lesions may in part be due to growth controls extrinsic to tumor cells. These data demonstrate a new approach to investigate the entire spectrum of human plasma cell neoplasia and illustrate the utility of humanized models for understanding the functional diversity of human tumors.


Assuntos
Medula Óssea , Proliferação de Células , Microambiente Celular , Camundongos , Modelos Animais , Mieloma Múltiplo , Plasmócitos/citologia , Lesões Pré-Cancerosas , Microambiente Tumoral , Animais , Humanos , Camundongos Transgênicos , Transplante de Neoplasias
4.
Proc Natl Acad Sci U S A ; 112(22): E2891-9, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26038565

RESUMO

Chitinases are enzymes that cleave chitin, a component of the exoskeleton of many organisms including the house dust mite (HDM). Here we show that knockin mice expressing an enzymatically inactive acidic mammalian chitinase (AMCase), the dominant true chitinase in mouse lung, showed enhanced type 2 immune responses to inhaled HDM. We found that uncleaved chitin promoted the release of IL-33, whereas cleaved chitin could be phagocytosed and could induce the activation of caspase-1 and subsequent activation of caspase-7; this results in the resolution of type 2 immune responses, probably by promoting the inactivation of IL-33. These data suggest that AMCase is a crucial regulator of type 2 immune responses to inhaled chitin-containing aeroallergens.


Assuntos
Asma/imunologia , Asma/prevenção & controle , Asma/parasitologia , Quitinases/imunologia , Modelos Animais de Doenças , Pyroglyphidae/imunologia , Animais , Western Blotting , Quitinases/genética , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Técnicas de Introdução de Genes , Interleucina-33 , Interleucinas/imunologia , Camundongos , Reação em Cadeia da Polimerase
5.
Annu Rev Immunol ; 31: 635-674, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23330956

RESUMO

To directly study complex human hemato-lymphoid system physiology and respective system-associated diseases in vivo, human-to-mouse xenotransplantation models for human blood and blood-forming cells and organs have been developed over the past three decades. We here review the fundamental requirements and the remarkable progress made over the past few years in improving these systems, the current major achievements reached by use of these models, and the future challenges to more closely model and study human health and disease and to achieve predictive preclinical testing of both prevention measures and potential new therapies.


Assuntos
Hematopoese/imunologia , Tecido Linfoide/imunologia , Tecido Linfoide/transplante , Modelos Animais , Animais , Transplante de Células-Tronco Hematopoéticas/métodos , Transplante de Células-Tronco Hematopoéticas/tendências , Humanos , Imunofenotipagem , Tecido Linfoide/patologia , Camundongos , Pesquisa Translacional Biomédica/métodos , Pesquisa Translacional Biomédica/tendências , Transplante Heterólogo
6.
Blood ; 118(11): 3119-28, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21791433

RESUMO

Humanized mouse models are useful tools to understand pathophysiology and to develop therapies for human diseases. While significant progress has been made in generating immunocompromised mice with a human hematopoietic system, there are still several shortcomings, one of which is poor human myelopoiesis. Here, we report that human CSF-1 knockin mice show augmented frequencies and functions of human myeloid cells. Insertion of human CSF1 into the corresponding mouse locus of Balb/c Rag2(-/-) γc(-/-) mice through VELOCIGENE technology resulted in faithful expression of human CSF-1 in these mice both qualitatively and quantitatively. Intra-hepatic transfer of human fetal liver derived hematopoietic stem and progenitor cells (CD34(+)) in humanized CSF-1 (CSF1(h/h)) newborn mice resulted in more efficient differentiation and enhanced frequencies of human monocytes/macrophages in the bone marrow, spleens, peripheral blood, lungs, liver and peritoneal cavity. Human monocytes/macrophages obtained from the humanized CSF-1 mice show augmented functional properties including migration, phagocytosis, activation and responses to LPS. Thus, humanized mice engineered to express human cytokines will significantly help to overcome the current technical challenges in the field. In addition, humanized CSF-1 mice will be a valuable experimental model to study human myeloid cell biology.


Assuntos
Diferenciação Celular/fisiologia , Fator Estimulador de Colônias de Macrófagos/genética , Macrófagos/fisiologia , Animais , Animais Recém-Nascidos , Células Cultivadas , Proteínas de Ligação a DNA/genética , Eficiência , Técnicas de Introdução de Genes , Humanos , Cadeias gama de Imunoglobulina/genética , Fator Estimulador de Colônias de Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Especificidade da Espécie
7.
Proc Natl Acad Sci U S A ; 108(32): 13218-23, 2011 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-21788509

RESUMO

Transplantation of human hematopoietic stem cells into severely immunocompromised newborn mice allows the development of a human hematopoietic and immune system in vivo. NOD/scid/γ(c)(-/-) (NSG) and BALB/c Rag2(-/-)γ(c)(-/-) mice are the most commonly used mouse strains for this purpose and a number of studies have demonstrated the high value of these model systems in areas spanning from basic to translational research. However, limited cross-reactivity of many murine cytokines on human cells and residual host immune function against the xenogeneic grafts results in defective development and maintenance of human cells in vivo. Whereas NSG mice have higher levels of absolute human engraftment than similar mice on a BALB/c background, they have a shorter lifespan and NOD ES cells are unsuitable for the complex genetic engineering that is required to improve human hematopoiesis and immune responses by transgenesis or knockin of human genes. We have generated mice that faithfully express a transgene of human signal regulatory protein alpha (SIRPa), a receptor that negatively regulates phagocytosis, in Rag2(-/-)γ(c)(-/-) mice on a mixed 129/BALB/c background, which can easily be genetically engineered. These mice allow significantly increased engraftment and maintenance of human hematopoietic cells reaching levels comparable to NSG mice. Furthermore, we found improved functionality of the human immune system in these mice. In summary, hSIRPa-transgenic Rag2(-/-)γ(c)(-/-) mice represent a unique mouse strain supporting high levels of human cell engraftment, which can easily be genetically manipulated.


Assuntos
Antígenos de Diferenciação/metabolismo , Proteínas de Ligação a DNA/deficiência , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Subunidade gama Comum de Receptores de Interleucina/deficiência , Receptores Imunológicos/metabolismo , Transgenes/genética , Animais , Antígenos de Diferenciação/genética , Células da Medula Óssea/patologia , Linhagem da Célula , Proteínas de Ligação a DNA/metabolismo , Epitopos/imunologia , Humanos , Imunidade Humoral/imunologia , Subunidade gama Comum de Receptores de Interleucina/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Camundongos Transgênicos , Receptores Imunológicos/genética
8.
Eur J Immunol ; 41(5): 1344-51, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21469125

RESUMO

CD4(+) T lymphocytes are required to induce spontaneous autoimmune diabetes in the NOD mouse. Since pancreatic ß cells upregulate Fas expression upon exposure to pro-inflammatory cytokines, we studied whether the diabetogenic action of CD4(+) T lymphocytes depends on Fas expression on target cells. We assayed the diabetogenic capacity of NOD spleen CD4(+) T lymphocytes when adoptively transferred into a NOD mouse model combining: (i) Fas-deficiency, (ii) FasL-deficiency, and (iii) SCID mutation. We found that CD4(+) T lymphocytes require Fas expression in the recipients' target cells to induce diabetes. IL-1ß has been described as a key cytokine involved in Fas upregulation on mouse ß cells. We addressed whether CD4(+) T cells require IL-1ß to induce diabetes. We also studied spontaneous diabetes onset in NOD/IL-1 converting enzyme-deficient mice, in NOD/IL-1ß-deficient mice, and CD4(+) T-cell adoptively transferred diabetes into NOD/SCID IL-1ß-deficient mice. Neither IL-1ß nor IL-18 are required for either spontaneous or CD4(+) T-cell adoptively transferred diabetes. We conclude that CD4(+) T-cell-mediated ß-cell damage in autoimmune diabetes depends on Fas expression, but not on IL-1ß unveiling the existing redundancy regarding the cytokines involved in Fas upregulation on NOD ß cells in vivo.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Tipo 1/imunologia , Receptor fas/metabolismo , Transferência Adotiva , Animais , Diabetes Mellitus Tipo 1/metabolismo , Diabetes Mellitus Tipo 1/patologia , Proteína Ligante Fas/deficiência , Proteína Ligante Fas/metabolismo , Genótipo , Células Secretoras de Insulina/imunologia , Células Secretoras de Insulina/patologia , Interleucina-18/metabolismo , Interleucina-1beta/genética , Interleucina-1beta/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Camundongos Transgênicos , Receptor fas/deficiência , Receptor fas/genética
9.
Proc Natl Acad Sci U S A ; 108(6): 2390-5, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21262803

RESUMO

Mice with a functional human immune system have the potential to allow in vivo studies of human infectious diseases and to enable vaccine testing. To this end, mice need to fully support the development of human immune cells, allow infection with human pathogens, and be capable of mounting effective human immune responses. A major limitation of humanized mice is the poor development and function of human myeloid cells and the absence of human immune responses at mucosal surfaces, such as the lung. To overcome this, we generated human IL-3/GM-CSF knock-in (hIL-3/GM-CSF KI) mice. These mice faithfully expressed human GM-CSF and IL-3 and developed pulmonary alveolar proteinosis because of elimination of mouse GM-CSF. We demonstrate that hIL-3/GM-CSF KI mice engrafted with human CD34(+) hematopoietic cells had improved human myeloid cell reconstitution in the lung. In particular, hIL-3/GM-CSF KI mice supported the development of human alveolar macrophages that partially rescued the pulmonary alveolar proteinosis syndrome. Moreover, human alveolar macrophages mounted correlates of a human innate immune response against influenza virus. The hIL-3/GM-CSF KI mice represent a unique mouse model that permits the study of human mucosal immune responses to lung pathogens.


Assuntos
Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Imunidade Inata , Vírus da Influenza A Subtipo H1N1/imunologia , Interleucina-3/imunologia , Pulmão/imunologia , Macrófagos Alveolares/imunologia , Infecções por Orthomyxoviridae/imunologia , Animais , Transplante de Células-Tronco de Sangue do Cordão Umbilical , Técnicas de Introdução de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Imunidade nas Mucosas/genética , Interleucina-3/genética , Pulmão/virologia , Macrófagos Alveolares/virologia , Camundongos , Camundongos Transgênicos , Modelos Imunológicos , Infecções por Orthomyxoviridae/genética , Quimeras de Transplante/genética , Quimeras de Transplante/imunologia , Quimeras de Transplante/virologia , Transplante Heterólogo
10.
Proc Natl Acad Sci U S A ; 108(6): 2378-83, 2011 Feb 08.
Artigo em Inglês | MEDLINE | ID: mdl-21262827

RESUMO

Hematopoietic stem cells (HSCs) both self-renew and give rise to all blood cells for the lifetime of an individual. Xenogeneic mouse models are broadly used to study human hematopoietic stem and progenitor cell biology in vivo. However, maintenance, differentiation, and function of human hematopoietic cells are suboptimal in these hosts. Thrombopoietin (TPO) has been demonstrated as a crucial cytokine supporting maintenance and self-renewal of HSCs. We generated RAG2(-/-)γ(c)(-/-) mice in which we replaced the gene encoding mouse TPO by its human homolog. Homozygous humanization of TPO led to increased levels of human engraftment in the bone marrow of the hosts, and multilineage differentiation of hematopoietic cells was improved, with an increased ratio of myelomonocytic verus lymphoid lineages. Moreover, maintenance of human stem and progenitor cells was improved, as demonstrated by serial transplantation. Therefore, RAG2(-/-)γ(c)(-/-) TPO-humanized mice represent a useful model to study human hematopoiesis in vivo.


Assuntos
Hematopoese , Trombopoetina/metabolismo , Animais , Técnicas de Introdução de Genes , Transplante de Células-Tronco Hematopoéticas , Humanos , Camundongos , Camundongos Transgênicos , Trombopoetina/genética , Quimeras de Transplante/genética , Quimeras de Transplante/metabolismo , Transplante Heterólogo
11.
Cell Host Microbe ; 8(4): 369-76, 2010 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-20951970

RESUMO

Salmonella enterica serovar Typhi (S. Typhi) causes typhoid fever, a life-threatening human disease. The lack of animal models due to S. Typhi's strict human host specificity has hindered its study and vaccine development. We find that immunodeficient Rag2(-/-) γc(-/-) mice engrafted with human fetal liver hematopoietic stem and progenitor cells are able to support S. Typhi replication and persistent infection. A S. Typhi mutant in a gene required for virulence in humans was unable to replicate in these mice. Another mutant unable to produce typhoid toxin exhibited increased replication, suggesting a role for this toxin in the establishment of persistent infection. Furthermore, infected animals mounted human innate and adaptive immune responses to S. Typhi, resulting in the production of cytokines and pathogen-specific antibodies. We expect that this mouse model will be a useful resource for understanding S. Typhi pathogenesis and for evaluating potential vaccine candidates against typhoid fever.


Assuntos
Modelos Animais de Doenças , Salmonella typhi/imunologia , Salmonella typhi/patogenicidade , Febre Tifoide/imunologia , Imunidade Adaptativa , Animais , Anticorpos Antibacterianos/sangue , Anticorpos Antibacterianos/imunologia , Proteínas de Transporte de Cátions/imunologia , Citocinas/sangue , Citocinas/imunologia , Células-Tronco Hematopoéticas/microbiologia , Especificidade de Hospedeiro , Humanos , Imunidade Inata , Camundongos , Camundongos Endogâmicos BALB C , Mutação , Salmonella typhi/genética , Virulência
12.
Nat Immunol ; 11(10): 962-8, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20802482

RESUMO

Induced regulatory T cells (iT(reg) cells) can be generated by peripheral dendritic cells (DCs) that mediate T cell unresponsiveness to rechallenge with antigen. The molecular factors required for the function of such iT(reg) cells remain unknown. We report a critical role for the transcription cofactor homeodomain-only protein (Hop; also known as Hopx) in iT(reg) cells to mediate T cell unresponsiveness in vivo. Hopx-sufficient iT(reg) cells downregulated expression of the transcription factor AP-1 complex and suppressed other T cells. In the absence of Hopx, iT(reg) cells had high expression of the AP-1 complex, proliferated and failed to mediate T cell unresponsiveness to rechallenge with antigen. Thus, Hopx is required for the function of T(reg) cells induced by DCs and the promotion of DC-mediated T cell unresponsiveness in vivo.


Assuntos
Células Dendríticas/imunologia , Proteínas de Homeodomínio/imunologia , Linfócitos T Reguladores/imunologia , Animais , Proteínas de Homeodomínio/genética , Tolerância Imunológica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator de Transcrição AP-1/metabolismo
13.
Blood ; 115(3): 530-40, 2010 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-19965648

RESUMO

In humans, interleukin-1beta (IL-1beta) has been suggested as an essential cytokine for developing IL-17- or IL-17A-producing CD4(+) T helper 17 (Th17) cells. However, little is known about the relationship of IL-1 receptor expression and Th17 cell differentiation. We report here the presence of 2 distinct CD4(+) T-cell populations with and without expression of IL-1RI that correlates with the capacity to produce IL-17 in naive and memory CD4(+) T cells of human peripheral blood. IL-1RI(+) memory CD4(+) T cells had increased gene expression of IL17, RORC, and IRF4 even before T-cell receptor triggering, indicating that the effect of IL-1beta is programmed in these cells via IL-1RI. Although CD4(+) T cells from umbilical cord blood did not express IL-1RI, the cytokines IL-7, IL-15, and transforming growth factor-beta (TGF-beta) up-regulated IL-1RI expression on naive CD4(+) T cells, suggesting that IL-1RI(+) naive CD4(+) T cells develop in periphery. Furthermore, IL-17 production from the cytokine-treated naive CD4(+) T cells was induced by IL-1beta and this induction was blocked by IL-1R antagonist. These results indicate that human Th17 cell differentiation is regulated via differential expression of IL-1RI, which is controlled by IL-7 and IL-15.


Assuntos
Diferenciação Celular/genética , Diferenciação Celular/imunologia , Receptores de Interleucina-1/genética , Células Th1/metabolismo , Células Th1/fisiologia , Animais , Linfócitos T CD4-Positivos/citologia , Linfócitos T CD4-Positivos/efeitos dos fármacos , Linfócitos T CD4-Positivos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Memória Imunológica/efeitos dos fármacos , Memória Imunológica/genética , Memória Imunológica/fisiologia , Interleucina-15/farmacologia , Interleucina-15/fisiologia , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucina-1beta/farmacologia , Interleucina-7/farmacologia , Interleucina-7/fisiologia , Camundongos , Receptores de Interleucina-1/metabolismo , Células Th1/efeitos dos fármacos , Células Th1/imunologia , Fator de Crescimento Transformador beta/farmacologia
14.
Cell Host Microbe ; 6(1): 5-9, 2009 Jul 23.
Artigo em Inglês | MEDLINE | ID: mdl-19616761

RESUMO

Over 800 million people worldwide are infected with hepatitis viruses, human immunodeficiency virus (HIV), and malaria, resulting in more than 5 million deaths annually. Here we discuss the potential and challenges of humanized mouse models for developing effective and affordable therapies and vaccines, which are desperately needed to combat these diseases.


Assuntos
Pesquisa Biomédica/tendências , Doenças Transmissíveis , Modelos Animais de Doenças , Animais , Humanos , Camundongos
15.
Proc Natl Acad Sci U S A ; 106(27): 11236-40, 2009 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-19549859

RESUMO

Idd3 is one of many gene regions that affect the development of type 1 diabetes (T1D) in the nonobese diabetic (NOD) mouse. Idd3 has been localized to a 650-kb region on chromosome 3 containing the IL-2 gene. Exon 1 of the IL-2 gene is polymorphic between the susceptible NOD and the protective C57BL/6 (B6) alleles, causing multiple amino acid changes that have been proposed to be responsible for the differing glycosylation status. To address whether this coding polymorphism recapitulates the disease suppression mediated by the B6 Idd3 allele, we generated knockin mice in which exon 1 of the B6 IL-2 allele replaces the homologous region in the NOD allele. We generated these mice by targeting the NOD allele of NOD/129 F(1) ES cells. IL-2 protein from the knockin mice showed the glycosylation pattern of the B6 IL-2 isoform, confirming that the amino acid differences encoded within exon 1 affect the glycosylation of the IL-2 protein. However, unlike NOD.B6 Idd3 congenic mice, the knockin mice were not protected from T1D. Furthermore, the difference in amino acid sequence in the IL-2 protein did not affect the level of expression of IL-2. This approach provides a general method for the determination of a functional role of a given genomic sequence in a disease process. Further, our result demonstrates that the variants in exon 1 of the IL-2 gene are not responsible for T1D suppression in NOD.B6 Idd3 mice, thereby supporting the hypothesis that variants in the regulatory region affecting expression levels are causative.


Assuntos
Aminoácidos/genética , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/prevenção & controle , Interleucina-2/genética , Interleucina-2/metabolismo , Polimorfismo de Nucleotídeo Único/genética , Alelos , Animais , Western Blotting , Linhagem Celular , Células Clonais , Cruzamentos Genéticos , Diabetes Mellitus Tipo 1/patologia , Células-Tronco Embrionárias/citologia , Células-Tronco Embrionárias/metabolismo , Feminino , Técnicas de Introdução de Genes , Glicosilação , Espaço Intracelular/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout
16.
Int Immunol ; 19(1): 1-10, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17101709

RESUMO

Constitutive expression of IL9 in the lungs of transgenic (Tg) mice resulted in an asthma-like phenotype consisting of lymphocytic and eosinophilic lung inflammation, mucus hypersecretion and mast cell hyperplasia. Several T(h)2 cytokines including IL4, IL5 and IL13 were expressed in the lung in response to Tg IL9. IL13 was absolutely necessary for the development of lung pathology. To understand how IL9 induces IL13-dependent lung inflammation and mucus production, we sought the IL13-producing cells. Surprisingly, we found that the absence of T cells and B cells in recombinase-activating gene 1 (RAG1)-deficient IL9 Tg mice enhanced lung inflammation and dramatically enhanced IL13 production. In addition, the lack of mast cells or eosinophils in IL9 Tg mice did not affect IL13 levels in the lung. In situ hybridization for IL13 on lung sections from RAG1-/- IL9 Tg mice revealed that airway epithelial cells were the major IL13-producing cell type. Our results implicate the lung epithelium as a potentially important source of inflammatory cytokines in asthma.


Assuntos
Células Epiteliais/efeitos dos fármacos , Interleucina-13/metabolismo , Interleucina-9/metabolismo , Pulmão/efeitos dos fármacos , Pneumonia/metabolismo , Animais , Células Epiteliais/metabolismo , Genes RAG-1/genética , Genes RAG-1/fisiologia , Interleucina-5/fisiologia , Interleucina-9/genética , Pulmão/patologia , Linfócitos/imunologia , Mastócitos/imunologia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Pneumonia/induzido quimicamente , Pneumonia/patologia , Fator de Transcrição STAT6/metabolismo
17.
Mol Cell Biol ; 26(13): 5180-9, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16782901

RESUMO

The FAT10 gene encodes a diubiquitin-like protein containing two tandem head-to-tail ubiquitin-like domains. There is a high degree of similarity between murine and human FAT10 sequences at both the mRNA and protein levels. In various cell lines, FAT10 expression was shown to be induced by gamma interferon or by tumor necrosis factor alpha. In addition, FAT10 expression was found to be up-regulated in some Epstein-Barr virus-infected B-cell lines, in activated dendritic cells, and in several epithelial tumors. However, forced expression of FAT10 in cultured cells was also found to produce apoptotic cell death. Overall, these findings suggest that FAT10 may modulate cellular growth or cellular viability. Here we describe the steps to generate, by genetic targeting, a FAT10 gene knockout mouse model. The FAT10 knockout homozygous mice are viable and fertile. No gross lesions or obvious histological differences were found in these mutated mice. Examination of lymphocyte populations from spleen, thymus, and bone marrow did not reveal any abnormalities. However, flow cytometry analysis demonstrated that the lymphocytes of FAT10 knockout mice were, on average, more prone to spontaneous apoptotic death. Physiologically, these mice demonstrated a high level of sensitivity toward endotoxin challenge. These findings indicate that FAT10 may function as a survival factor.


Assuntos
Apoptose , Linfócitos/fisiologia , Ubiquitinas/fisiologia , Sequência de Aminoácidos , Animais , Apoptose/genética , Células da Medula Óssea/citologia , Células Dendríticas/citologia , Citometria de Fluxo , Humanos , Lipopolissacarídeos/toxicidade , Linfócitos/citologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Dados de Sequência Molecular , Fenótipo , Sepse/genética , Baço/citologia , Timo/citologia , Ubiquitinas/genética
18.
Cell ; 122(1): 2-4, 2005 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-16009124

RESUMO

The human gut is host to hundreds of different species of commensal bacteria that live in peaceful partnership with the host immune system. These commensal bacteria are far from neutral bystanders as they are intimately involved in the development of the immune system. Reporting in this issue of Cell, Kasper and colleagues (Mazmanian et al., 2005) reveal that a bacterial polysaccharide, PSA, produced by the commensal bacterium Bacteroides fragilis directs development of the immune system of the mouse host.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Polissacarídeos Bacterianos/biossíntese , Animais , Bacteroides fragilis/química , Bacteroides fragilis/imunologia , Humanos , Sistema Imunitário/efeitos dos fármacos , Sistema Imunitário/crescimento & desenvolvimento , Sistema Imunitário/imunologia , Camundongos , Polissacarídeos Bacterianos/imunologia , Polissacarídeos Bacterianos/farmacologia , Baço/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...