Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 222
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Transplant ; 16(2): 389-97, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26588186

RESUMEN

Our understanding of the molecular pathways that control immune responses, particularly immunomodulatory molecules that control the extent and duration of an immune response, have led to new approaches in the field of transplantation immunology to induce allograft survival. These molecular pathways are being defined precisely in murine models and translated into clinical practice; however, many of the newly available drugs are human-specific reagents. Furthermore, many species-specific differences exist between mouse and human immune systems. Recent advances in the development of humanized mice, namely, immunodeficient mice engrafted with functional human immune systems, have led to the availability of a small animal model for the study of human immune responses. Humanized mice represent an important preclinical model system for evaluation of new drugs and identification of the mechanisms underlying human allograft rejection without putting patients at risk. This review highlights recent advances in the development of humanized mice and their use as preclinical models for the study of human allograft responses.


Asunto(s)
Modelos Animales de Enfermedad , Rechazo de Injerto/prevención & control , Trasplante de Órganos , Inmunología del Trasplante , Animales , Rechazo de Injerto/inmunología , Humanos , Ratones , Pronóstico
2.
Clin Exp Immunol ; 179(3): 398-413, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25302633

RESUMEN

Several ß cell antigens recognized by T cells in the non-obese diabetic (NOD) mouse model of type 1 diabetes (T1D) are also T cell targets in the human disease. While numerous antigen-specific therapies prevent diabetes in NOD mice, successful translation of rodent findings to patients has been difficult. A human leucocyte antigen (HLA)-transgenic mouse model incorporating human ß cell-specific T cells might provide a better platform for evaluating antigen-specific therapies. The ability to study such T cells is limited by their low frequency in peripheral blood and the difficulty in obtaining islet-infiltrating T cells from patients. We have worked to overcome this limitation by using lentiviral transduction to 'reprogram' primary human CD8 T cells to express three T cell receptors (TCRs) specific for a peptide derived from the ß cell antigen islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP265-273 ) and recognized in the context of the human class I major histocompatibility complex (MHC) molecule HLA-A2. The TCRs bound peptide/MHC multimers with a range of avidities, but all bound with at least 10-fold lower avidity than the anti-viral TCR used for comparison. One exhibited antigenic recognition promiscuity. The ß cell-specific human CD8 T cells generated by lentiviral transduction with one of the TCRs released interferon (IFN)-γ in response to antigen and exhibited cytotoxic activity against peptide-pulsed target cells. The cells engrafted in HLA-A2-transgenic NOD-scid IL2rγ(null) mice and could be detected in the blood, spleen and pancreas up to 5 weeks post-transfer, suggesting the utility of this approach for the evaluation of T cell-modulatory therapies for T1D and other T cell-mediated autoimmune diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Vectores Genéticos/genética , Inmunoterapia Adoptiva/métodos , Células Secretoras de Insulina/inmunología , Lentivirus/genética , Linfocitos T Citotóxicos/inmunología , Animales , Linfocitos T CD8-positivos/trasplante , Supervivencia Celular , Glucosa-6-Fosfatasa/inmunología , Antígeno HLA-A2/genética , Antígeno HLA-A2/metabolismo , Humanos , Células Jurkat , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Fragmentos de Péptidos/inmunología , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores de Interleucina-2/genética , Linfocitos T Citotóxicos/trasplante
3.
Gene Ther ; 20(6): 658-69, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23076379

RESUMEN

In vivo delivery is a major barrier to the use of molecular tools for gene modification. Here we demonstrate site-specific gene editing of human cells in vivo in hematopoietic stem cell-engrafted NOD.Cg-Prkdc(scid)IL2rγ(tm1Wjl) (abbreviated NOD-scid IL2rγ(null)) mice, using biodegradable nanoparticles loaded with triplex-forming peptide nucleic acids (PNAs) and single-stranded donor DNA molecules. In vitro screening showed greater efficacy of nanoparticles containing PNAs/DNAs together over PNA-alone or DNA-alone. Intravenous injection of particles containing PNAs/DNAs produced modification of the human CCR5 gene in hematolymphoid cells in the mice, with modification confirmed at the genomic DNA, mRNA and functional levels. Deep sequencing revealed in vivo modification of the CCR5 gene at frequencies of 0.43% in hematopoietic cells in the spleen and 0.05% in the bone marrow: off-target modification in the partially homologous CCR2 gene was two orders of magnitude lower. We also induced specific modification in the ß-globin gene using nanoparticles carrying ß-globin-targeted PNAs/DNAs, demonstrating this method's versatility. In vivo testing in an enhanced green fluorescent protein-ß-globin reporter mouse showed greater activity of nanoparticles containing PNAs/DNAs together over DNA only. Direct in vivo gene modification, such as we demonstrate here, would allow for gene therapy in systemic diseases or in cells that cannot be manipulated ex vivo.


Asunto(s)
ADN/genética , Marcación de Gen , Técnicas de Transferencia de Gen , Nanopartículas/química , Ácidos Nucleicos de Péptidos/genética , Animales , Línea Celular , ADN/administración & dosificación , ADN/química , Terapia Genética , Células Madre Hematopoyéticas/citología , Humanos , Ratones , Nanopartículas/administración & dosificación , Ácidos Nucleicos de Péptidos/administración & dosificación , Ácidos Nucleicos de Péptidos/química , Receptores CCR5/genética
4.
Clin Exp Immunol ; 174(3): 372-88, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23869841

RESUMEN

Immunodeficient mice bearing targeted mutations in the IL2rg gene and engrafted with human immune systems are effective tools for the study of human haematopoiesis, immunity, infectious disease and transplantation biology. The most robust human immune model is generated by implantation of human fetal thymic and liver tissues in irradiated recipients followed by intravenous injection of autologous fetal liver haematopoietic stem cells [often referred to as the BLT (bone marrow, liver, thymus) model]. To evaluate the non-obese diabetic (NOD)-scid IL2rγ(null) (NSG)-BLT model, we have assessed various engraftment parameters and how these parameters influence the longevity of NSG-BLT mice. We observed that irradiation and subrenal capsule implantation of thymus/liver fragments was optimal for generating human immune systems. However, after 4 months, a high number of NSG-BLT mice develop a fatal graft-versus-host disease (GVHD)-like syndrome, which correlates with the activation of human T cells and increased levels of human immunoglobulin (Ig). Onset of GVHD was not delayed in NSG mice lacking murine major histocompatibility complex (MHC) classes I or II and was not associated with a loss of human regulatory T cells or absence of intrathymic cells of mouse origin (mouse CD45(+) ). Our findings demonstrate that NSG-BLT mice develop robust human immune systems, but that the experimental window for these mice may be limited by the development of GVHD-like pathological changes.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas , Timo/trasplante , Animales , Modelos Animales de Enfermedad , Células Madre Hematopoyéticas , Humanos , Inmunoglobulina G/sangre , Inmunoglobulina M/sangre , Subunidad gamma Común de Receptores de Interleucina/genética , Subunidad gamma Común de Receptores de Interleucina/inmunología , Antígenos Comunes de Leucocito/metabolismo , Activación de Linfocitos , Recuento de Linfocitos , Ratones , Ratones Endogámicos NOD , Datos de Secuencia Molecular , Linfocitos T Reguladores/inmunología , Timo/inmunología
5.
Clin Exp Immunol ; 166(2): 269-80, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21985373

RESUMEN

Graft-versus-host disease (GVHD) is a life-threatening complication of human allogeneic haematopoietic stem cell transplantation. Non-obese diabetic (NOD)-scid IL2rγ(null) (NSG) mice injected with human peripheral blood mononuclear cells (PBMC) engraft at high levels and develop a robust xenogeneic (xeno)-GVHD, which reproduces many aspects of the clinical disease. Here we show that enriched and purified human CD4 T cells engraft readily in NSG mice and mediate xeno-GVHD, although with slower kinetics compared to injection of whole PBMC. Moreover, purified human CD4 T cells engraft but do not induce a GVHD in NSG mice that lack murine MHC class II (NSG-H2-Ab1(tm1Gru), NSG-Ab°), demonstrating the importance of murine major histocompatibility complex (MHC) class II in the CD4-mediated xeno-response. Injection of purified human CD4 T cells from a DR4-negative donor into a newly developed NSG mouse strain that expresses human leucocyte antigen D-related 4 (HLA-DR4) but not murine class II (NSG-Ab° DR4) induces an allogeneic GVHD characterized by weight loss, fur loss, infiltration of human cells in skin, lung and liver and a high level of mortality. The ability of human CD4 T cells to mediate an allo-GVHD in NSG-Ab° DR4 mice suggests that this model will be useful to investigate acute allo-GVHD pathogenesis and to evaluate human specific therapies.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped , Receptores de Interleucina-2/inmunología , Animales , Linfocitos T CD4-Positivos/trasplante , Genes MHC Clase II , Enfermedad Injerto contra Huésped/inmunología , Antígeno HLA-DR4/inmunología , Humanos , Leucocitos Mononucleares/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Trasplante Heterólogo
6.
Vet Pathol ; 48(2): 495-9, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20817888

RESUMEN

Spontaneous morbidity primarily affecting female breeders in 3 independent breeding colonies of NSG (NOD.Cg-Prkdc(scid) I12rg(tm1Wjl) /SzJ) mice prompted an investigation to uncover the cause of disease. Necropsies were performed on 264 (157 female and 107 male) spontaneously sick, experimentally unmanipulated NSG mice. In sum, 42 mice (15.9%) had acute or chronic renal inflammatory lesions, of which 12 had concurrent histologic evidence of an ascending urinary tract infection. From 94 kidneys cultured for bacterial organisms, 23 (24.5%) grew Enterococcus sp and 19 (20.2%) grew Klebsiella oxytoca. Female mice were twice more likely than males to present with nephritis. These findings indicate that bacterial nephritis is a major contributor to morbidity in the NSG strain.


Asunto(s)
Animales de Laboratorio , Enterococcus , Klebsiella oxytoca , Nefritis/veterinaria , Infecciones Oportunistas/veterinaria , Enfermedades de los Roedores/epidemiología , Enfermedades de los Roedores/microbiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos , Morbilidad , Nefritis/microbiología , Infecciones Oportunistas/epidemiología , Infecciones Oportunistas/microbiología
7.
Gene Ther ; 17(2): 238-49, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19829316

RESUMEN

Human embryonic stem cells (hESCs) provide a novel source of hematopoietic and other cell populations suitable for gene therapy applications. Preclinical studies to evaluate engraftment of hESC-derived hematopoietic cells transplanted into immunodeficient mice demonstrate only limited repopulation. Expression of a drug-resistance gene, such as Tyr22-dihydrofolate reductase (Tyr22-DHFR), coupled to methotrexate (MTX) chemotherapy has the potential to selectively increase the engraftment of gene-modified, hESC-derived cells in mouse xenografts. Here, we describe the generation of Tyr22-DHFR-GFP-expressing hESCs that maintain pluripotency, produce teratomas and can differentiate into MTXr-hemato-endothelial cells. We demonstrate that MTX administered to nonobese diabetic/severe combined immunodeficient/IL-2Rgammac(null) (NSG) mice after injection of Tyr22-DHFR-hESC-derived cells significantly increases human CD34(+) and CD45(+) cell engraftment in the bone marrow (BM) and peripheral blood of transplanted MTX-treated mice. These results demonstrate that MTX treatment supports selective, long-term engraftment of Tyr22-DHFR cells in vivo, and provides a novel approach for combined human cell and gene therapy.


Asunto(s)
Células Madre Embrionarias/metabolismo , Metotrexato/farmacología , Trasplante de Células Madre/métodos , Tetrahidrofolato Deshidrogenasa/genética , Animales , Médula Ósea , Diferenciación Celular , Línea Celular , Resistencia a Medicamentos , Terapia Genética/métodos , Supervivencia de Injerto , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Teratoma/genética , Tetrahidrofolato Deshidrogenasa/metabolismo
8.
J Exp Med ; 181(6): 2265-70, 1995 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-7760012

RESUMEN

The study of human malaria has been hampered by the lack of small animal models for the human-infecting malarial parasites. To approach this problem, the erythrocytic stages of the human malarial parasite Plasmodium falciparum were adapted to in vitro growth in the presence of ascites fluid from mice homozygous for the severe-combined immunodeficiency (scid) mutation. Human red blood cells (hRBCs) infected with these adapted parasites were then injected i.p. into nonobese diabetic scid/scid (NOD/LtSz-scid) mice. With daily supplemental intraperitoneal boosts of uninfected hRBCs, parasites were detected in the peripheral circulation of these mice for an average of 7 d after injection. Splenectomy of NOD/LtSz-scid mice increased both the level and duration of parasitemia in the periphery, and it also promoted the circulation of mature sexual stage parasites (gametocytes). When Anopheline mosquitoes were allowed to feed on the splenectomized mice, the gametocytes were ingested by the mosquitoes and developed into oocysts in the mosquito midguts. To our knowledge, these results are the first demonstration of human malarial parasite propagation in mice and transmission of these parasites to the invertebrate vector.


Asunto(s)
Culicidae/parasitología , Malaria Falciparum/fisiopatología , Plasmodium falciparum/fisiología , Animales , Eritrocitos/parasitología , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Parasitemia/fisiopatología , Plasmodium falciparum/crecimiento & desarrollo , Plasmodium falciparum/patogenicidad , Factores de Tiempo
9.
J Exp Med ; 191(4): 731-6, 2000 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-10684864

RESUMEN

Host defense against multicellular, extracellular pathogens such as nematode parasites is believed to be mediated largely, if not exclusively, by T lymphocytes. During our investigations into the course of Brugia malayi and Brugia pahangi infections in immunodeficient mouse models, we found that mice lacking B lymphocytes were permissive for Brugian infections, whereas immunocompetent mice were uniformly resistant. Mice bearing the Btk(xid) mutation were as permissive as those lacking all B cells, suggesting that the B1 subset may be responsible for host protection. Reconstitution of immunodeficient recombination activating gene (Rag)-1(-/)- mice with B1 B cells conferred resistance, even in the absence of conventional B2 lymphocytes and most T cells. These results suggest that B1 B cells are necessary to mediate host resistance to Brugian infection. Our data are consistent with a model wherein early resistance to B. malayi is mediated by humoral immune response, with a significant attrition of the incoming infectious larval load. Sterile clearance of the remaining parasite burden appears to require cell-mediated immunity. These data raise the possibility that the identification of molecule(s) recognized by humoral immune mechanisms might help generate prophylactic vaccines.


Asunto(s)
Linfocitos B/inmunología , Brugia Malayi/inmunología , Brugia pahangi/inmunología , Filariasis/inmunología , Animales , Subgrupos de Linfocitos B/inmunología , Filariasis/prevención & control , Citometría de Flujo , Inmunocompetencia , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones SCID , Especificidad de la Especie , Linfocitos T/inmunología
10.
J Exp Med ; 173(3): 659-63, 1991 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-1997651

RESUMEN

The C.B.-17-scid/scid mouse (hereafter referred to as the scid mouse) is homozygous for a recessive mutation at a locus that influences the assembly of intact immunoglobulin and T cell receptor genes. Therefore, scid mice cannot generate functional B or T lymphocytes, are profoundly immunodeficient, and have been reported to be receptive to reconstitution with human immune cells. In the present study, we injected scid mice with infective larvae of the human filarial parasite Brugia malayi. Within 6-10 wk after subcutaneous injection of infective L3 larvae, both male and female worms were observed in various stages of development in 90% of the mice. In animals tested 8 weeks or more after infection, microfilariae were detected in the blood or peritoneal cavity of 52% of the mice examined. Adult worms were observed in the lymphatics of the infected scid mice, where their presence was associated with lymphangitis and lymphangiectasia. These results suggest that the scid mouse model of lymphatic filariasis may be important in investigation of the interaction of the murine, and possibly the human, immune system with the lymphatic filarial parasite.


Asunto(s)
Brugia , Filariasis Linfática/inmunología , Síndromes de Inmunodeficiencia/complicaciones , Animales , Modelos Animales de Enfermedad , Filariasis Linfática/patología , Humanos , Inmunoglobulinas/genética , Síndromes de Inmunodeficiencia/genética , Síndromes de Inmunodeficiencia/patología , Inflamación , Subgrupos Linfocitarios/inmunología , Ratones , Ratones Mutantes , Receptores de Antígenos de Linfocitos T/genética , Bazo/inmunología
11.
J Exp Med ; 180(2): 717-20, 1994 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-8046347

RESUMEN

Interleukin 7 (IL-7) responsive B lineage precursors were greatly expanded in genetically hypogonadal female (HPG/Bm-hpg/hpg) mice that have a secondary deficiency in gonadal steroidogenesis. Estrogen replacement in these mice resulted in a dose-dependent reduction in B cell precursors. More modest increases were documented in genetically normal mice that were surgically castrated. These findings complement other recent observations that B lymphopoiesis selectively declines in pregnant or estrogen-treated animals. Sex steroids have long been known to influence such disparate processes as bone physiology and tumor growth, in addition to their importance for reproductive function. We now show that these hormones are important negative regulators of B lymphopoiesis.


Asunto(s)
Linfocitos B/citología , Estrógenos/fisiología , Hipogonadismo/inmunología , Animales , Linfocitos B/inmunología , División Celular/genética , Terapia de Reemplazo de Estrógeno , Estrógenos/deficiencia , Estrógenos/uso terapéutico , Femenino , Hormona Liberadora de Gonadotropina/genética , Hipogonadismo/tratamiento farmacológico , Hipogonadismo/genética , Interleucina-7/fisiología , Ratones
12.
J Exp Med ; 186(3): 449-54, 1997 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-9236197

RESUMEN

Mast cells are the main effector cells of immediate hypersensitivity and anaphylaxis. Their role in the development of allergen-induced airway hyperresponsiveness (AHR) is controversial and based on indirect evidence. To address these issues, mast cell-deficient mice (W/W v) and their congenic littermates were sensitized to ovalbumin (OVA) by intraperitoneal injection and subsequently challenged with OVA via the airways. Comparison of OVA-specific immunoglobulin E (IgE) levels in the serum and numbers of eosinophils in bronchoalveolar lavage fluid or lung digests showed no differences between the two groups of mice. Further, measurements of airway resistance and dynamic compliance at baseline and after inhalation of methacholine were similar. These data indicate that mast cells or IgE-mast cell activation is not required for the development of eosinophilic inflammation and AHR in mice sensitized to allergen via the intraperitoneal route and challenged via the airways.


Asunto(s)
Hiperreactividad Bronquial/patología , Eosinófilos/patología , Mastocitos/patología , Resistencia de las Vías Respiratorias/inmunología , Animales , Hiperreactividad Bronquial/fisiopatología , Líquido del Lavado Bronquioalveolar/inmunología , Femenino , Inmunoglobulina E/biosíntesis , Inmunoglobulina G/biosíntesis , Pulmón/inmunología , Pulmón/patología , Rendimiento Pulmonar/inmunología , Mastocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ovalbúmina/administración & dosificación , Ovalbúmina/inmunología
13.
J Exp Med ; 185(4): 673-84, 1997 Feb 17.
Artículo en Inglés | MEDLINE | ID: mdl-9034146

RESUMEN

The lytic activity of natural killer (NK) cells is inhibited by the expression of class I major histocompatibility complex (MHC) antigens on target cells. In murine NK cells, Ly-49A mediates inhibition of cytotoxicity in response to the class I MHC antigen H-2Dd. In this report, we studied the function of mouse Ly-49A in both the rat NK cell tumor line, RNK-16, transfected with Ly-49A cDNA, and in primary NK cells. We show that ligation of Ly-49A by H-2Dd inhibits early signaling events during target cell stimulation, including polyphosphoinositide turnover and tyrosine phosphorylation. We also show that Ly-49A directly associates with the cytoplasmic tyrosine phosphatase SHP-1, and that Ly-49A function is impaired in NK cells from SHP-1 mutant viable motheaten mice and from SHP-1-deficient motheaten mice. Finally, we demonstrate that mutational substitution of the tyrosine within the proposed SHP-1 binding motif in Ly-49A completely abrogates inhibition of NK cell cytotoxicity through this receptor. These results demonstrate that Ly-49A interrupts early activating signals in NK cells, and that SHP-1 is an important mediator of Ly-49A function.


Asunto(s)
Citotoxicidad Inmunológica/inmunología , Células Asesinas Naturales/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Proteínas Tirosina Fosfatasas/metabolismo , Transducción de Señal , Animales , Péptidos y Proteínas de Señalización Intracelular , Ratones , Fosfatidilinositoles/metabolismo , Fosforilación , Unión Proteica , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Proteína Tirosina Fosfatasa no Receptora Tipo 6 , Ratas , Tirosina/metabolismo
14.
J Exp Med ; 184(5): 2049-53, 1996 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-8920894

RESUMEN

The T lymphocytes mediating autoimmune destruction of pancreatic beta cells in the nonobese diabetic (NOD) mouse model of insulin-dependent diabetes mellitus (IDDM) may be generated due to functional defects in hematopoietically derived antigen-presenting cells (APC). However, it has not been clear which particular subpopulations of APC (B lymphocytes, macrophages, and dendritic cells) contribute to the development and activation of diabetogenic T cells in NOD mice. In the current study we utilized a functionally inactivated immunoglobulin (Ig) mu allele (Ig mu null) to generate a "speed congenic" stock of B lymphocyte-deficient NOD mice that are fixed for linkage markers delineating previously identified diabetes susceptibility (Idd) genes. These B lymphocyte NOD.Ig mu null mice had normal numbers of T cells but were free of overt IDDM and insulitis resistant, while the frequency of disease in the B lymphocyte intact segregants was equivalent to that of standard NOD mice in our colony. Thus, B lymphocytes play a heretofore unrecognized role that is essential for the initial development and/or activation of beta cell autoreactive T cells in NOD mice.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Linfocitos B/inmunología , Diabetes Mellitus Tipo 1/inmunología , Cadenas mu de Inmunoglobulina/inmunología , Linfocitos T/inmunología , Factores de Edad , Animales , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/genética , Ligamiento Genético , Marcadores Genéticos , Homocigoto , Cadenas mu de Inmunoglobulina/genética , Subgrupos Linfocitarios , Ratones , Ratones Endogámicos NOD , Ratones Mutantes , Bazo/citología , Bazo/inmunología
15.
Clin Exp Immunol ; 157(1): 104-18, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19659776

RESUMEN

Immunodeficient non-obese diabetic (NOD)-severe combined immune-deficient (scid) mice bearing a targeted mutation in the gene encoding the interleukin (IL)-2 receptor gamma chain gene (IL2rgamma(null)) engraft readily with human peripheral blood mononuclear cells (PBMC). Here, we report a robust model of xenogeneic graft-versus-host-like disease (GVHD) based on intravenous injection of human PBMC into 2 Gy conditioned NOD-scid IL2rgamma(null) mice. These mice develop xenogeneic GVHD consistently (100%) following injection of as few as 5 x 10(6) PBMC, regardless of the PBMC donor used. As in human disease, the development of xenogeneic GVHD is highly dependent on expression of host major histocompatibility complex class I and class II molecules and is associated with severely depressed haematopoiesis. Interrupting the tumour necrosis factor-alpha signalling cascade with etanercept, a therapeutic drug in clinical trials for the treatment of human GVHD, delays the onset and progression of disease. This model now provides the opportunity to investigate in vivo mechanisms of xenogeneic GVHD as well as to assess the efficacy of therapeutic agents rapidly.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Subunidad gamma Común de Receptores de Interleucina/genética , Leucocitos Mononucleares/trasplante , Complejo Mayor de Histocompatibilidad , Modelos Animales , Animales , Etanercept , Femenino , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Humanos , Inmunoglobulina G/uso terapéutico , Inmunosupresores/uso terapéutico , Inyecciones Intravenosas , Antígenos Comunes de Leucocito/análisis , Leucocitos Mononucleares/inmunología , Prueba de Cultivo Mixto de Linfocitos , Masculino , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Receptores del Factor de Necrosis Tumoral/uso terapéutico , Distribución Tisular , Trasplante Heterólogo , Irradiación Corporal Total
16.
Curr Top Microbiol Immunol ; 324: 25-51, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18481451

RESUMEN

There is a growing need for effective animal models to carry out experimental studies on human hematopoietic and immune systems without putting individuals at risk. Progress in development of small animal models for the in vivo investigation of human hematopoiesis and immunity has seen three major breakthroughs over the last three decades. First, CB 17-Prkdc(scid) (abbreviated CB 17-scid) mice were discovered in 1983, and engraftment of these mice with human fetal tissues (SCID-Hu model) and peripheral blood mononuclear cells (Hu-PBL-SCID model) was reported in 1988. Second, NOD-scid mice were developed and their enhanced ability to engraft with human hematolymphoid tissues as compared with CB17-scid mice was reported in 1995. NOD-scid mice have been the "gold standard" for studies of human hematolymphoid engraftment in small animal models over the last 10 years. Third, immunodeficient mice bearing a targeted mutation in the IL-2 receptor common gamma chain (IL2rgamma(null)) were developed independently by four groups between 2002 and 2005, and a major increase in the engraftment and function of human hematolymphoid cells as compared with NOD-scid mice has been reported. These new strains of immunodeficient IL2rgamma(null) mice are now being used for studies in human hematopoiesis, innate and adaptive immunity, autoimmunity, infectious diseases, cancer biology, and regenerative medicine. In this chapter, we discuss the current state of development of these strains of mice, the remaining deficiencies, and how approaches used to increase the engraftment and function of human hematolymphoid cells in CB 17-scid mice and in previous models based on NOD-scid mice may enhance human hematolymphoid engraftment and function in NOD-scid IL2rgamma(null) mice.


Asunto(s)
Investigación Biomédica/métodos , Modelos Animales de Enfermedad , Animales , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Ratones , Ratones Endogámicos NOD , Ratones SCID
17.
Curr Top Microbiol Immunol ; 324: 87-94, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18481454

RESUMEN

Biomedical research including immunology and stem cell biology has developed greatly because of the evolving technology of gene modification and conventional transplantation methods using the most common experimental laboratory animal, the mouse. To translate promising research findings based on mouse research into clinical medicine, however, we need to clarify whether similar events take place in humans. In the study of hematology and immunology, humanized mice provide a unique and efficient experimental system to evaluate differentiation, function, and interaction of human blood cells or immune components. Here we review the latest experimental findings in the fields of immunology, stem cell biology, and regenerative medicine using humanized mice.


Asunto(s)
Médula Ósea/fisiología , Diferenciación Celular , Células Madre Hematopoyéticas/fisiología , Modelos Animales , Regeneración , Animales , Trasplante de Células Madre Hematopoyéticas , Humanos , Ratones , Ratones SCID
18.
J Cell Biol ; 114(1): 35-43, 1991 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-1646823

RESUMEN

Secretory defects in abnormal plasma cells, called Mott cells, that appear in lymphoid tissues of spontaneously autoimmune, "viable motheaten" (mev/mev) mice lead to deposition of immunoglobulin in RER-bound vesicles. Such vesicles have been termed Russel bodies. Cells with Russel bodies can also be observed rarely in normal animals, usually as a result of extreme antigenic loads or pathologic states. To understand why these abnormal cells appear commonly in mev/mev mice, we have established a panel of hybridomas that contain Russell bodies. Using immunochemical analysis and immunoelectron microscopy, we have characterized the secretory defects. Although these hybridoma cells synthesize a normal size heavy chain and it associates with light chain, the Russell bodies have many characteristics of inclusion bodies, which commonly appear in cells synthesizing mutant proteins and often are associated with incompletely or abnormally folded proteins. Pulse-chase experiments showed that immunoglobulins synthesized by these hybridomas accumulate rapidly into insoluble complexes and have an intracellular half life approximately 10 time greater than normal immunoglobulins. The defect affected only the immunoglobulin derived from the mev/mev mice and did not affect the secretion of normal immunoglobulin produced by an IgG1-secreting fusion partner. In addition to accumulating intracellular immunoglobulins, many mutant cell lines also secreted immunoglobulin. Endoglycosidase H digestion was used to determine the state of processing of the N-linked carbohydrates on the immunoglobulin molecules. This analysis demonstrated that the N-linked carbohydrates on the secreted immunoglobulin were resistant to endoglycosidase H digestion, indicating that they were processed normally. The insoluble IgM molecules were sensitive to endoglycosidase H, which is consistent with their localization to the RER. We propose several models by which these abnormal immunoglobulin-secreting cells commonly appear in this autoimmune mutant mouse.


Asunto(s)
Enfermedades Autoinmunes/inmunología , Inmunoglobulina M/metabolismo , Cuerpos de Inclusión/inmunología , Células Plasmáticas/inmunología , Acetilglucosaminidasa/metabolismo , Animales , Enfermedades Autoinmunes/patología , Retículo Endoplásmico/metabolismo , Retículo Endoplásmico/ultraestructura , Hibridomas , Inmunoglobulina G/química , Inmunoglobulina G/metabolismo , Inmunoglobulina M/química , Cadenas mu de Inmunoglobulina/química , Cuerpos de Inclusión/ultraestructura , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa , Ratones , Ratones Mutantes , Células Plasmáticas/ultraestructura
19.
Science ; 232(4756): 1423-5, 1986 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-3487115

RESUMEN

Almost all B cells in autoimmune mice with the viable motheaten (mev) mutation express the Ly-1 cell surface antigen, which marks a minor population of B cells constituting a separate lineage in normal mice. Immunoglobulins primarily of the M and G3 classes, which in both normal and mev mice contain high levels of lambda light chain, are produced in excess in mev mice. These and other observations suggest that the development of B cells that express Ly-1 is regulated independently from the development of B cells that do not express Ly-1. B cells bearing the Ly-1 surface antigen may play specialized roles in the normal immune system and in autoimmunity by regulating other B cells via lymphokines, by producing antibodies to self and certain foreign antigens, and by preferentially secreting immunoglobulin M and immunoglobulin G3.


Asunto(s)
Antígenos Ly/inmunología , Enfermedades Autoinmunes/inmunología , Linfocitos B/inmunología , Ratones Mutantes/inmunología , Animales , Enfermedades Autoinmunes/genética , Linfocitos B/clasificación , Citometría de Flujo , Inmunoglobulina D/metabolismo , Inmunoglobulina G/metabolismo , Inmunoglobulina M/metabolismo , Inmunoglobulinas/análisis , Ganglios Linfáticos/citología , Ratones , Cavidad Peritoneal/citología , Bazo/citología
20.
Science ; 241(4873): 1632-9, 1988 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-2971269

RESUMEN

The study of human hematopoietic cells and the human immune system is hampered by the lack of a suitable experimental model. Experimental data are presented showing that human fetal liver hematopoietic cells, human fetal thymus, and human fetal lymph node support the differentiation of mature human T cells and B cells after engraftment into mice with genetically determined severe combined immunodeficiency. The resultant SCID-hu mice are found to have a transient wave of human CD4+ and CD8+ T cells and human IgG (immunoglobulin G) in the peripheral circulation. The functional status of the human immune system within this mouse model is not yet known.


Asunto(s)
Quimera , Células Madre Hematopoyéticas/fisiología , Ratones Mutantes/inmunología , Modelos Biológicos , Animales , Linfocitos B/fisiología , Diferenciación Celular , Humanos , Hígado/citología , Trasplante de Hígado , Ganglios Linfáticos/trasplante , Ratones , Linfocitos T/fisiología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Reguladores/inmunología , Timo/anatomía & histología , Timo/fisiología , Timo/trasplante , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA