Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Diabetes ; 61(5): 1160-8, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22368175

RESUMEN

In earlier studies of the Iddm14 diabetes susceptibility locus in the rat, we identified an allele of the T-cell receptor (TCR) ß-chain, Tcrb-V13S1A1, as a candidate gene. To establish its importance, we treated susceptible rats with a depleting anti-rat Vß13 monoclonal antibody and then exposed them to either polyinosinic:polycytidylic acid or a diabetogenic virus to induce diabetes. The overall frequency of diabetes in the controls was 74% (n = 50), compared with 17% (n = 30) in the anti-Vß13-treated animals, with minimal islet pathology in nondiabetic treated animals. T cells isolated from islets on day 5 after starting induction showed a greater proportion of Vß13(+) T cells than did peripheral lymph node T cells. Vß13 transcripts recovered from day 5 islets revealed focused Jß usage and less CDR3 diversity than did transcripts from peripheral Vß13(+) T cells. CDR3 usage was not skewed in control Vß16 CDR3 transcripts. Anti-rat Vß13 antibody also prevented spontaneous diabetes in BBDP rats. The Iddm14 gene is likely to be Tcrb-V13, indicating that TCR ß-chain usage is a determinant of susceptibility to autoimmune diabetes in rats. It may be possible to prevent autoimmune diabetes by targeting a limited element of the T-cell repertoire.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Diabetes Mellitus Tipo 1/prevención & control , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Alelos , Animales , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Islotes Pancreáticos/citología , Islotes Pancreáticos/metabolismo , Masculino , Poli I-C/toxicidad , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/fisiología
2.
Blood ; 119(12): 2778-88, 2012 Mar 22.
Artículo en Inglés | MEDLINE | ID: mdl-22246028

RESUMEN

Immunodeficient mice engrafted with human HSCs support multidisciplinary translational experimentation, including the study of human hematopoiesis. Heightened levels of human HSC engraftment are observed in immunodeficient mice expressing mutations in the IL2-receptor common γ chain (IL2rg) gene, including NOD-scid IL2rγ(null) (NSG) mice. Engraftment of human HSC requires preconditioning of immunodeficient recipients, usually with irradiation. Such preconditioning increases the expression of stem cell factor (SCF), which is critical for HSC engraftment, proliferation, and survival. We hypothesized that transgenic expression of human membrane-bound stem cell factor Tg(hu-mSCF)] would increase levels of human HSC engraftment in nonirradiated NSG mice and eliminate complications associated with irradiation. Surprisingly, detectable levels of human CD45(+) cell chimerism were observed after transplantation of cord blood-derived human HSCs into nonirradiated adult as well as newborn NSG mice. However, transgenic expression of human mSCF enabled heightened levels of human hematopoietic cell chimerism in the absence of irradiation. Moreover, nonirradiated NSG-Tg(hu-mSCF) mice engrafted as newborns with human HSCs rejected human skin grafts from a histoincompatible donor, indicating the development of a functional human immune system. These data provide a new immunodeficient mouse model that does not require irradiation preconditioning for human HSC engraftment and immune system development.


Asunto(s)
Diferenciación Celular/fisiología , Células Madre Hematopoyéticas/citología , Ratones Transgénicos , Factor de Células Madre/metabolismo , Quimera por Trasplante/fisiología , Animales , Animales Recién Nacidos , Separación Celular , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Factor de Células Madre/genética , Tolerancia al Trasplante/fisiología
3.
Chem Biol ; 18(9): 1189-98, 2011 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-21944757

RESUMEN

Peptide nucleic acids (PNAs) bind duplex DNA in a sequence-specific manner, creating triplex structures that can provoke DNA repair and produce genome modification. CCR5 encodes a chemokine receptor required for HIV-1 entry into human cells, and individuals carrying mutations in this gene are resistant to HIV-1 infection. Transfection of human cells with PNAs targeted to the CCR5 gene, plus donor DNAs designed to introduce stop codons mimicking the naturally occurring CCR5-delta32 mutation, produced 2.46% targeted gene modification. CCR5 modification was confirmed at the DNA, RNA, and protein levels and was shown to confer resistance to infection with HIV-1. Targeting of CCR5 was achieved in human CD34(+) hematopoietic stem cells (HSCs) with subsequent engraftment into mice and persistence of the gene modification more than four months posttransplantation. This work suggests a therapeutic strategy for CCR5 knockout in HSCs from HIV-1-infected individuals, rendering cells resistant to HIV-1 and preserving immune system function.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Ácidos Nucleicos de Péptidos/farmacología , Receptores CCR5/metabolismo , Animales , Antígenos CD34/metabolismo , Secuencia de Bases , Sitios de Unión , Antagonistas de los Receptores CCR5 , Línea Celular , Codón de Terminación , Reparación del ADN , Marcación de Gen/métodos , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , VIH-1/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/virología , Humanos , Ratones , Mutación , Ácidos Nucleicos de Péptidos/química , Receptores CCR5/genética
4.
Mol Pharm ; 8(3): 767-73, 2011 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-21361360

RESUMEN

Noninvasive measurement of human islet cell mass in pancreas or following islet transplantation by nuclear imaging has yet to be achieved. It has been shown using mouse tumor models that pretargeting imaging strategies are sensitive and can greatly increase target to nontarget signal ratios. The objective now is to demonstrate the specific pretargeting of human islet cells in mice. Our pretargeting strategy uses an anti-human islet cell antibody HPi1, conjugated to a phosphorodiamidate morpholino oligomer (MORF) that binds specifically to a (99m)Tc labeled complementary MORF (cMORF). Sensitivity and specificity of the pretargeting were first validated in culture using a human beta cell line (betalox5) and a negative control human cell line (HEK293). Pretargeting was then used to target and visualize these two cell lines and human islets transplanted subcutaneously in NOD-scid IL2rγ(null) mice. In culture, (99m)Tc accumulation on the betalox5 cells pretargeted by MORF-HPi1 was 100-fold higher than on untreated betalox5 cells or following treatment with native HPi1 and much higher than on the MORF-HPi1 pretargeted control HEK293 cells. Small animal imaging readily localized the transplanted betalox5 cells and human islets, but not the HEK293 cells. Ex vivo counting demonstrated 3-fold higher (99m)Tc accumulation in the transplanted betalox5 cells and human islets than in the control HEK293 cells. The target accumulation was also shown to increase linearly with increased numbers of the implanted betalox5 cells. These results demonstrate specific binding of radioactivity and successful imaging of human betalox5 cells and human islets transplanted in mice. Thus MORF/cMORF pretargeting may be useful to measure noninvasively human islet cell mass within the pancreas or following islet transplantation.


Asunto(s)
Anticuerpos/metabolismo , Células Secretoras de Insulina/citología , Células Secretoras de Insulina/metabolismo , Células Secretoras de Insulina/trasplante , Morfolinas/metabolismo , Animales , Cromatografía Líquida de Alta Presión , Células HEK293 , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Morfolinos
5.
Autoimmunity ; 44(2): 137-48, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20695765

RESUMEN

The adipokine, leptin, regulates blood glucose and the insulin secretory function of beta cells, while also modulating immune cell function. We hypothesized that the dual effects of leptin may prevent or suppress the autoreactive destruction of beta cells in a virally induced rodent model of type 1 diabetes. Nearly 100% of weanling BBDR rats treated with the combination of an innate immune system activator, polyinosinic:polycytidylic acid (pIC), and Kilham rat virus (KRV) become diabetic within a predictable time frame. We utilized this model to test the efficacy of leptin in preventing diabetes onset, remitting new onset disease, and preventing autoimmune recurrence in diabetic rats transplanted with syngeneic islet grafts. High doses of leptin delivered via an adenovirus vector (AdLeptin) or alzet pump prevented diabetes in>90% of rats treated with pIC+KRV. The serum hyperleptinemia generated by this treatment was associated with decreased body weight, decreased non-fasting serum insulin levels, and lack of islet insulitis in leptin-treated rats. In new onset diabetics, hyperleptinemia prevented rapid weight loss and diabetic ketoacidosis, and temporarily restored euglycemia. Leptin treatment also prolonged the survival of syngeneic islets transplanted into diabetic BBDR rats. In diverse therapeutic settings, we found leptin treatment to have significant beneficial effects in modulating virally induced diabetes. These findings merit further evaluation of leptin as a potential adjunct therapeutic agent for treatment of human type 1 diabetes.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Leptina/uso terapéutico , Infecciones por Parvoviridae/inmunología , Parvovirus/inmunología , Animales , Glucemia , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/virología , Diabetes Mellitus Tipo 1/patología , Diabetes Mellitus Tipo 1/virología , Cetoacidosis Diabética/prevención & control , Humanos , Trasplante de Islotes Pancreáticos , Leptina/administración & dosificación , Leptina/inmunología , Infecciones por Parvoviridae/virología , Poli I-C/administración & dosificación , Poli I-C/inmunología , Ratas , Ratas Endogámicas BB , Resultado del Tratamiento
6.
Diabetes ; 59(9): 2265-70, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20570944

RESUMEN

OBJECTIVE: To create an immunodeficient mouse model that spontaneously develops hyperglycemia to serve as a diabetic host for human islets and stem cell-derived beta-cells in the absence or presence of a functional human immune system. RESEARCH DESIGN AND METHODS: We backcrossed the Ins2(Akita) mutation onto the NOD-Rag1(null) IL2rgamma(null) strain and determined 1) the spontaneous development of hyperglycemia, 2) the ability of human islets, mouse islets, and dissociated mouse islet cells to restore euglycemia, 3) the generation of a human immune system following engraftment of human hematopoietic stem cells, and 4) the ability of the humanized mice to reject human islet allografts. RESULTS: We confirmed the defects in innate and adaptive immunity and the spontaneous development of hyperglycemia conferred by the IL2rgamma(null), Rag1(null), and Ins2(Akita) genes in NOD-Rag1(null) IL2rgamma(null) Ins2(Akita) (NRG-Akita) mice. Mouse and human islets restored NRG-Akita mice to normoglycemia. Insulin-positive cells in dissociated mouse islets, required to restore euglycemia in chemically diabetic NOD-scid IL2rgamma(null) and spontaneously diabetic NRG-Akita mice, were quantified following transplantation via the intrapancreatic and subrenal routes. Engraftment of human hematopoietic stem cells in newborn NRG-Akita and NRG mice resulted in equivalent human immune system development in a normoglycemic or chronically hyperglycemic environment, with >50% of engrafted NRG-Akita mice capable of rejecting human islet allografts. CONCLUSIONS: NRG-Akita mice provide a model system for validation of the function of human islets and human adult stem cell, embryonic stem cell, or induced pluripotent stem cell-derived beta-cells in the absence or presence of an alloreactive human immune system.


Asunto(s)
Trasplante de Islotes Pancreáticos/inmunología , Trasplante Heterólogo/inmunología , Trasplante Homólogo/inmunología , Inmunidad Adaptativa , Animales , Glucemia/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Cruzamientos Genéticos , Citometría de Flujo , Humanos , Inmunidad Innata , Subunidad gamma Común de Receptores de Interleucina/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Mutación
7.
Diabetes ; 59(1): 110-8, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19794063

RESUMEN

OBJECTIVE: The contribution of antecedent viral infection to the development of type 1 diabetes in humans is controversial. Using a newer rat model of the disease, we sought to 1) identify viruses capable of modulating diabetes penetrance, 2) identify conditions that increase or decrease the diabetogenicity of infection, and 3) determine whether maternal immunization would prevent diabetes. RESEARCH DESIGN AND METHODS: About 2% of LEW*1WR1 rats develop spontaneous autoimmune diabetes, but disease penetrance is much higher if weanling rats are exposed to environmental perturbants including Kilham rat virus (KRV). We compared KRV with other viruses for diabetogenic activity. RESULTS: Both KRV and rat cytomegalovirus (RCMV) induced diabetes in up to 60% of LEW*1WR1 rats, whereas H-1, vaccinia, and Coxsackie B4 viruses did not. Simultaneous inoculation of KRV and RCMV induced diabetes in 100% of animals. Pretreatment of rats with an activator of innate immunity increased the diabetogenicity of KRV but not RCMV and was associated with a moderate rate of diabetes after Coxsackie B4 and vaccinia virus infection. Inoculation of LEW*1WR1 dams with both KRV and RCMV prior to pregnancy protected weanling progeny from virus-induced diabetes in a virus-specific manner. CONCLUSIONS: Exposure to viruses can affect the penetrance of autoimmune diabetes in genetically susceptible animals. The diabetogenicity of infection is virus specific and is modified by immunomodulation prior to inoculation. Maternal immunization protects weanlings from virus-induced diabetes, suggesting that modification of immune responses to infection could provide a means of preventing islet autoimmunity.


Asunto(s)
Diabetes Mellitus Tipo 1/prevención & control , Diabetes Mellitus Tipo 1/virología , Inmunización/métodos , Virosis/complicaciones , Virosis/inmunología , Animales , Infecciones por Citomegalovirus/inmunología , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Enterovirus Humano B/inmunología , Infecciones por Enterovirus/inmunología , Femenino , Humanos , Modelos Animales , Poli I-C/inmunología , Embarazo , Ratas , Ratas Endogámicas Lew , Ratas Endogámicas , Virus Vaccinia/inmunología
8.
Mamm Genome ; 20(3): 162-9, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19205800

RESUMEN

Iddm14 (formerly Iddm4) is a non-MHC-linked genetic locus associated with autoimmune diabetes. Its effects have been well-documented in BB-derived rats in which diabetes is either induced by immunologic perturbation or occurs spontaneously. The role of Iddm14 in non-BB rat strains is unknown. Our goal was to extend the analysis of Iddm14 in new diabetes-susceptible strains and to identify candidate genes in the rat Iddm14 diabetes susceptibility locus that are common to these multiple diabetic strains. To determine if Iddm14 is important in strains other than BB, we first genotyped a (LEW.1WR1 x WF)F2 cohort in which diabetes was induced by perturbation with polyinosinic:polycytidylic acid. We found that Iddm14 is a major determinant of diabetes susceptibility in LEW.1WR1 rats. We then used nucleotide sequencing to establish a strain distribution pattern of polymorphisms (insertions, deletions, and single nucleotide polymorphisms [SNPs]) that predicts susceptibility to diabetes in a panel of inbred and congenic rats. Using the positional information from the congenic strains and the new linkage data, we identified a susceptibility haplotype in the T-cell receptor Vbeta chain (Tcrb-V) locus. This haplotype includes Tcrb-V13, which is identical in five susceptible strains but different in resistant WF and F344 rats. We conclude that Iddm14 is a powerful determinant of both spontaneous and induced autoimmune diabetes in multiple rat strains, and that Tcrb-V13 SNPs constitute a haplotype of gene elements that may be critical for autoimmune diabetes in rats.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Predisposición Genética a la Enfermedad , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Animales , Femenino , Haplotipos , Humanos , Masculino , Polimorfismo de Nucleótido Simple , Ratas , Ratas Endogámicas
9.
Clin Immunol ; 126(3): 303-14, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18096436

RESUMEN

Immunodeficient NOD-scid mice bearing a targeted mutation in the IL2 receptor common gamma chain (Il2rgamma(null)) readily engraft with human stem cells. Here we analyzed human peripheral blood mononuclear cells (PBMC) for their ability to engraft NOD-scid Il2rgamma(null) mice and established engraftment kinetics, optimal cell dose, and the influence of injection route. Even at low PBMC input, NOD-scid Il2rgamma(null) mice reproducibly support high human PBMC engraftment that plateaus within 3-4 weeks. In contrast to previous stocks of immunodeficient mice, we observed low intra- and inter-donor variability of engraftment. NOD-scid Il2rgamma(null) mice rendered hyperglycemic by streptozotocin treatment return to normoglycemia following transplantation with human islets. Interestingly, these human islet grafts are rejected following injection of HLA-mismatched human PBMC as evidenced by return to hyperglycemia and loss of human C-peptide. These data suggest that humanized NOD-scid Il2rgamma(null) mice may represent an important surrogate for investigating in vivo mechanisms of human islet allograft rejection.


Asunto(s)
Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Islotes Pancreáticos/inmunología , Leucocitos Mononucleares/inmunología , Mutación/genética , Animales , Diabetes Mellitus/inmunología , Modelos Animales de Enfermedad , Rechazo de Injerto/inmunología , Humanos , Trasplante de Islotes Pancreáticos/inmunología , Leucocitos Mononucleares/trasplante , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Fenotipo
10.
Ann N Y Acad Sci ; 1103: 90-3, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17376822

RESUMEN

The use of "humanized" mice represents an appealing translational model for studies of the pathogenesis of immune-mediated diseases and for the evaluation of potential therapeutics. The utility of humanized mice depends on their ability to model the human immune system with high fidelity, and, in this respect, previous models have fallen short. The recently developed NOD-scid Il2rgamma(null) mouse, however, exhibits greatly enhanced ability to support the engraftment of human peripheral blood mononuclear cells. Herein, we describe the challenges of recapitulating human immunity in humanized mice and features of NOD-scid Il2rgamma(null) mice that help overcome them.


Asunto(s)
Diabetes Mellitus Tipo 1/inmunología , Islotes Pancreáticos/inmunología , Leucocitos Mononucleares/inmunología , Ratones Endogámicos NOD/genética , Ratones SCID/genética , Animales , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Humanos , Isoantígenos/inmunología , Ratones , Ratones Endogámicos BALB C
11.
Ann N Y Acad Sci ; 1103: 128-31, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17376830

RESUMEN

Iddm4 is a dominant non-major histocompatibility complex (MHC) determinant of diabetes susceptibility in BBDR rats treated with poly I:C, plus depletion of regulatory T cells. In congenic MHC-identical normal WF rats, Iddm4(d) sensitively and specifically predicts induced diabetes. We report a new diabetes-susceptible subcongenic line that carries Iddm4 in a < 2.6 megabase interval. Candidate genes include the T cell receptor beta chain variable (TCRVbeta) family. We found that TCRVbeta4 in WF rats contains a stop codon, whereas 5/5 diabetes-susceptible rat strains express TCRVbeta4. We conclude that Iddm4-mediated diabetes resistance in rats may be due to a recessive protective mutation in TCRVbeta4.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Región Variable de Inmunoglobulina , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Cruzamientos Genéticos , Cartilla de ADN , Modelos Animales de Enfermedad , Femenino , Masculino , Ratas , Ratas Endogámicas
12.
J Immunol ; 177(11): 7820-32, 2006 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17114453

RESUMEN

Biobreeding (BB) rats model type 1 autoimmune diabetes (T1D). BB diabetes-prone (BBDP) rats develop T1D spontaneously. BB diabetes-resistant (BBDR) rats develop T1D after immunological perturbations that include regulatory T cell (Treg) depletion plus administration of low doses of a TLR ligand, polyinosinic-polycytidylic acid. Using both models, we analyzed CD4+CD25+ and CD4+CD45RC- candidate rat Treg populations. In BBDR and control Wistar Furth rats, CD25+ T cells comprised 5-8% of CD4+ T cells. In vitro, rat CD4+CD25+ T cells were hyporesponsive and suppressed T cell proliferation in the absence of TGF-beta and IL-10, suggesting that they are natural Tregs. In contrast, CD4+CD45RC(-) T cells proliferated in vitro in response to mitogen and were not suppressive. Adoptive transfer of purified CD4+CD25+ BBDR T cells to prediabetic BBDP rats prevented diabetes in 80% of recipients. Surprisingly, CD4+CD45RC-CD25- T cells were equally protective. Quantitative studies in an adoptive cotransfer model confirmed the protective capability of both cell populations, but the latter was less potent on a per cell basis. The disease-suppressing CD4+CD45RC-CD25- population expressed PD-1 but not Foxp3, which was confined to CD4+CD25+ cells. We conclude that CD4+CD25+ cells in the BBDR rat act in vitro and in vivo as natural Tregs. In addition, another population that is CD4+CD45RC-CD25- also participates in the regulation of autoimmune diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/metabolismo , Factores de Transcripción Forkhead/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/metabolismo , Traslado Adoptivo , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Proliferación Celular , Diabetes Mellitus Tipo 1/inmunología , Citometría de Flujo , Antígenos Comunes de Leucocito/biosíntesis , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Ratas , Ratas Endogámicas BB , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Subgrupos de Linfocitos T/inmunología , Linfocitos T Reguladores/inmunología
13.
Xenotransplantation ; 13(3): 224-32, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16756565

RESUMEN

BACKGROUND: The induction of xenogeneic hematopoietic chimerism is an attractive approach for overcoming the host response to xenografts, but establishing xenogeneic chimerism requires severe myeloablative conditioning of the recipient. The goal of this study was to determine if co-stimulation blockade would facilitate chimerism and xenograft tolerance in irradiation-conditioned concordant recipients. METHODS: Wistar Furth rat bone marrow (BM) cells were injected into irradiation-conditioned C57BL/6 mice with or without co-administration of anti-mouse CD154 monoclonal antibody (mAb). Chimerism was quantified by flow cytometry, and mice were transplanted with WF rat skin and islet xenografts. RESULTS: Blockade of CD40-CD154 interaction facilitated establishment of xenogeneic chimerism in mice conditioned with 600 cGy irradiation. Anti-CD154 mAb was not required for establishment of chimerism in mice treated with 700 cGy. However, mice irradiated with 700 cGy but not treated with anti-CD154 mAb developed a "graft-versus-host disease (GVHD)-like" wasting syndrome and died, irrespective of their development of chimerism. Xenogeneic chimeras established with irradiation and anti-CD154 mAb treatment exhibited prolonged skin and, in many cases, permanent islet xenograft survival. Chimerism was unstable and eventually lost in most recipients. Skin xenografts were rejected even in mice that remained chimeric, whereas most islet xenografts survived to the end of the observation period. CONCLUSIONS: Blockade of host CD40-CD154 interaction facilitates the establishment of xenogeneic chimerism and prevents wasting disease and death. Chimerism permits prolonged xenograft survival, but chimerism generated in this way is unstable over time. Skin xenografts are eventually rejected, whereas most islet xenografts survive long term and perhaps permanently.


Asunto(s)
Anticuerpos/farmacología , Trasplante de Médula Ósea/fisiología , Ligando de CD40/inmunología , Supervivencia de Injerto/fisiología , Hematopoyesis/inmunología , Trasplante de Islotes Pancreáticos/fisiología , Trasplante de Piel/fisiología , Quimera por Trasplante/inmunología , Trasplante Heterólogo/fisiología , Animales , Trasplante de Médula Ósea/patología , Antígenos CD40/inmunología , Citometría de Flujo , Reacción Huésped-Injerto , Trasplante de Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos C57BL , Ratas , Trasplante de Piel/patología , Ensayo de Capsula Subrrenal , Factores de Tiempo , Acondicionamiento Pretrasplante/métodos , Irradiación Corporal Total
14.
Diabetes ; 54(9): 2727-33, 2005 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16123363

RESUMEN

We describe a new rat model of autoimmune diabetes that arose in a major histocompatibility complex congenic LEW rat. Spontaneous diabetes in LEW.1WR1 rats (RT1(u/u/a)) occurs with a cumulative frequency of approximately 2% at a median age of 59 days. The disease is characterized by hyperglycemia, glycosuria, ketonuria, and polyuria. Both sexes are affected, and islets of acutely diabetic rats are devoid of beta-cells, whereas alpha- and delta-cell populations are spared. The peripheral lymphoid phenotype is normal, including the fraction of ART2(+) regulatory T-cells. We tested the hypothesis that the expression of diabetes would be increased by immunological perturbation of innate or adaptive immunity. Treatment of young rats with depleting anti-ART2.1 monoclonal antibody increased the frequency of diabetes to 50%. Treatment with the toll-like receptor 3 ligand polyinosinic:polycytidylic acid increased the frequency of diabetes to 100%. All diabetic rats exhibited end-stage islets. The LEW.1WR1 rat is also susceptible to collagen-induced arthritis but is free of spontaneous thyroiditis. The LEW.1WR1 rat provides a new model for studying autoimmune diabetes and arthritis in an animal with a genetic predisposition to both disorders that can be amplified by environmental perturbation.


Asunto(s)
Diabetes Mellitus/genética , Diabetes Mellitus/fisiopatología , Animales , Anticuerpos Monoclonales/farmacología , Artritis/inducido químicamente , Colágeno/toxicidad , Modelos Animales de Enfermedad , Femenino , Lipopolisacáridos/farmacología , Masculino , Poli I-C/farmacología , Ratas , Factores de Tiempo
15.
Diabetes ; 54(4): 1233-7, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15793267

RESUMEN

BBDR rats develop autoimmune diabetes only after challenge with environmental perturbants. These perturbants include polyinosinic:polycytidylic acid (poly I:C, a ligand of toll-like receptor 3), agents that deplete regulatory T-cell (Treg) populations, and a non-beta-cell cytopathic parvovirus (Kilham rat virus [KRV]). The dominant diabetes susceptibility locus Iddm4 is required for diabetes induced by treatment with poly I:C plus Treg depletion. Iddm4 is penetrant in congenic heterozygous rats on the resistant WF background and is 79% sensitive and 80% specific as a predictor of induced diabetes. Surprisingly, an analysis of 190 (BBDR x WF)F2 rats treated with KRV after brief exposure to poly I:C revealed that the BBDR-origin allele of Iddm4 is necessary but not entirely sufficient for diabetes expression. A genome scan identified a locus on chromosome 17, designated Iddm20, that is also required for susceptibility to diabetes after exposure to KRV and poly I:C (logarithm of odds score 3.7). These data suggest that the expression of autoimmune diabetes is a complex process that requires both major histocompatibility complex genes that confer susceptibility and additional genes such as Iddm4 and Iddm20 that operate only in the context of specific environmental perturbants, amplifying the immune response and the rate of disease progression.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/virología , Predisposición Genética a la Enfermedad , Ratas Endogámicas BB/genética , Alelos , Animales , Mapeo Cromosómico , Diabetes Mellitus Tipo 1/inmunología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Ligamiento Genético , Activación de Linfocitos , Glicoproteínas de Membrana/antagonistas & inhibidores , Infecciones por Parvoviridae/complicaciones , Poli I-C/farmacología , Ratas , Receptores de Superficie Celular/antagonistas & inhibidores , Linfocitos T , Receptor Toll-Like 3 , Receptores Toll-Like
16.
Transplantation ; 78(11): 1601-8, 2004 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-15591948

RESUMEN

BACKGROUND: Treatment with anti-CD154 monoclonal antibody (mAb) plus a donor-specific transfusion (DST) of spleen cells prolongs skin allograft survival in mice through a mechanism involving deletion of host alloreactive CD8(+) T cells. It is unknown if other lymphohematopoietic cell populations can be used as a DST. METHODS: Murine recipients of allogeneic skin grafts on day 0 were either untreated or given a DST on day -7 plus 4 doses of anti-CD154 mAb on days -7, -4, 0, and +4. Deletion of CD8(+) alloreactive cells was measured using "synchimeric" CBA recipients, which circulate trace populations of TCR transgenic alloreactive CD8(+) T cells. RESULTS: Transfusion of splenocytes, thymocytes, lymph node cells, or buffy coat cells led to prolonged skin allograft survival in recipients treated with anti-CD154 mAb. In contrast, bone marrow DST failed to delete host alloreactive CD8(+) T cells and was associated with brief skin allograft survival. Transfusions consisting of bone marrow-derived dendritic cells or a mixture of splenocytes and bone marrow cells were also ineffective. CONCLUSIONS: Donor-specific transfusions of splenocytes, thymocytes, lymph node cells, or buffy coat cells can prolong skin allograft survival in recipients treated with costimulation blockade. Bone marrow cells fail to serve this function, in part by failing to delete host alloreactive CD8(+) T cells, and they may actively interfere with the function of a spleen cell DST. The data suggest that transplantation tolerance induction protocols that incorporate bone marrow cells to serve as a DST may not be effective.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Transfusión Sanguínea , Células de la Médula Ósea/fisiología , Ligando de CD40/fisiología , Supervivencia de Injerto , Trasplante de Piel , Animales , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/trasplante , Depleción Linfocítica , Metrizamida/farmacología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Bazo/citología , Trasplante Homólogo
17.
Transplantation ; 78(5): 660-7, 2004 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-15371665

RESUMEN

BACKGROUND: Donor-specific transfusion (DST) and a brief course of anti-CD154 monoclonal antibody (mAb) induces permanent islet and prolonged skin allograft survival in mice. Induction of skin allograft survival requires the presence of CD4 cells and deletion of alloreactive CD8 cells. The specific roles of CD4 and CD4CD25 cells and the mechanism(s) by which they act are not fully understood. METHODS: We used skin and islet allografts, a CD8 T cell receptor (TCR) transgenic model system, and in vivo depleting antibodies to analyze the role of CD4 cell subsets in regulating allograft survival in mice treated with DST and anti-CD154 mAb. RESULTS: Deletion of CD4 or CD25 cells during costimulation blockade induced rapid rejection of skin but only minimally shortened islet allograft survival. Deletion of CD4 or CD25 cells had no effect upon survival of healed-in islet allografts, and CD25 cell deletion had no effect upon healed-in skin allograft survival. In the TCR transgenic model, DST plus anti-CD154 mAb treatment deleted alloreactive CD8 T cells, and anti-CD4 mAb treatment prevented that deletion. In contrast, injection of anti-CD25 mAb did not prevent alloreactive CD8 T cell deletion. CONCLUSIONS: These data document that (1) both CD4CD25 and CD4CD25 cells are required for induction of skin allograft survival, (2) CD4CD25 T cells are not required for alloreactive CD8 T cell deletion, and (3) CD4CD25 regulatory cells are not critical for islet allograft tolerance. It appears that skin and islet transplantation tolerance are mediated by different CD4 cell subsets and different mechanisms.


Asunto(s)
Trasplante de Islotes Pancreáticos/inmunología , Trasplante de Piel/inmunología , Subgrupos de Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Transfusión Sanguínea , Linfocitos T CD4-Positivos/inmunología , Ligando de CD40/inmunología , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Experimental/cirugía , Femenino , Terapia de Inmunosupresión/métodos , Activación de Linfocitos , Depleción Linfocítica , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Transgénicos , Modelos Animales , Trasplante Homólogo/inmunología
18.
Mamm Genome ; 15(1): 53-61, 2004 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-14727142

RESUMEN

Iddm4 is one of several susceptibility genes that have been identified in the BB rat model of type 1 diabetes. The BB rat allele of this gene confers dominant predisposition to diabetes induction by immune perturbation in both the diabetes-prone and the diabetes-resistant substrains, whereas the Wistar Furth (WF) allele confers resistance. We have positioned the gene in a 2.8-cM region on rat Chromosome (Chr) 4, proximal to Lyp/Ian4l1. We have produced a radiation hybrid map of the Iddm4-region that includes a number of rat genes with their mouse and human orthologs. We present a comparative map of the rat Iddm4 region in rat, human, and mouse, assigning the gene to a 6.3-Mb segment between PTN and ZYX at 7q32 in the human genome, and to a 5.7-Mb segment between Ptn and Zyx in the mouse genome.


Asunto(s)
Proteínas Portadoras/genética , Mapeo Cromosómico , Diabetes Mellitus Tipo 1/genética , Animales , Cromosomas/genética , Diabetes Mellitus Tipo 1/patología , Marcadores Genéticos , Predisposición Genética a la Enfermedad , Genoma , Humanos , Escala de Lod , Ratones , Repeticiones de Microsatélite , Mapeo de Híbrido por Radiación , Ratas , Ratas Endogámicas BB , Ratas Endogámicas WF
19.
Transplantation ; 76(7): 1036-42, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14557749

RESUMEN

BACKGROUND: A critical need exists for effective small-animal models that accept engraftment of human hematopoietic progenitor cells and mature lymphocytes. The purpose of this study was to determine the phenotypic effects of perforin (Pfp) deficiency on nonobese diabetic (NOD)-Rag1null mice and to evaluate the ability of NOD/LtSz-Rag1nullPfpnull recipients to support engraftment with human hematolymphoid cells. METHODS: A new genetic stock of NOD mice doubly homozygous for targeted mutations at the recombination activating gene (Rag)-1 and Pfp genes was developed. NOD/LtSz-Rag1nullPfpnull mice were studied for immunopathologic and hematologic abnormalities. The ability of these mice to support engraftment with human peripheral blood mononuclear cells (PBMC) and umbilical-cord blood hematopoietic progenitor cells was assessed. RESULTS: NOD/LtSz-Rag1nullPfpnull mice lacked mature B cells, T cells, natural killer (NK) cell cytotoxic activity and were devoid of serum immunoglobulin (Ig) throughout a 37-week lifespan. These mice supported heightened engraftment with human PBMC as compared with NOD/LtSz-Rag1null controls as evidenced by a 4- to 5-fold increase in percentages of human lymphocytes and a 7- to 13-fold increase in percentages of CD4+ T cells in the peripheral blood and spleen. Total numbers of human CD4+ T cells were increased approximately 20-fold in the spleens of NOD/LtSz-Rag1nullPfpnull mice. These mice also showed approximately 12-fold higher levels of engraftment with human umbilical-cord blood cells compared with NOD/LtSz-Rag1null mice. CONCLUSIONS: NOD/LtSz-Rag1nullPfpnull mice are devoid of mature B cell, T cell, and NK cell cytotoxic activity, engraft at high levels with human PBMC, and hematopoietic progenitor cells and provide a new NK cell-deficient model for human hematolymphoid cell engraftment.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Proteínas de Homeodominio/metabolismo , Transfusión de Leucocitos , Glicoproteínas de Membrana/deficiencia , Ratones Endogámicos NOD/metabolismo , Animales , Recuento de Células Sanguíneas , Células Sanguíneas/trasplante , Cruzamiento , Citotoxicidad Inmunológica , Sangre Fetal/citología , Proteínas de Homeodominio/genética , Humanos , Inmunoglobulinas/sangre , Células Asesinas Naturales , Longevidad , Tejido Linfoide/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados/genética , Monocitos/trasplante , Perforina , Fenotipo , Proteínas Citotóxicas Formadoras de Poros , Bazo/patología , Bazo/fisiopatología , Irradiación Corporal Total
20.
Exp Hematol ; 31(6): 551-8, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12829032

RESUMEN

OBJECTIVE: NOD/SCID and NOD/SCID B2m(null) mice are used for the in vivo study of human hematopoietic stem cells (HSC). A previously unrecognized HSC in cord blood, termed short-term repopulating cell (STRC), has been identified using NOD/SCID B2m(null) mice. However, only low levels of STRC engraft in NOD/SCID mice, presumably due to their higher levels of NK cell activity. The objective of these studies was to deplete NK cells both by genetic manipulation of the hosts and by antibody depletion of cell populations that may regulate engraftment with human STRC. METHODS: C57BL/6-SCID mice and immunodeficient NOD mice genetically deleted in NK cell activity were injected intravenously with human cord blood cells to quantify STRC engraftment. Cohorts of these mice were also treated with anti-NK1.1 or anti-CD122 (IL-2r beta-chain) antibodies. RESULTS: Human STRC fail to engraft in C57BL/6-SCID mice treated with anti-NK1.1 or with anti-CD122 antibody that targets mouse NK and myeloid cells. NOD/SCID mice, NOD-Rag1(null) mice, and NOD-Rag1(null)Pfp(null) mice that are genetically deleted in NK cell cytotoxic activity support only low levels of STRC engraftment. In contrast, STRC engraft at high levels in all three strains of immunodeficient NOD mice treated with anti-CD122 antibody. CONCLUSION: Injection of anti-CD122 antibody leads to high levels of STRC engraftment in immunodeficient NOD mice, but not in C57BL/6-SCID mice. These data document that depletion of NK cells is required, and that additional murine host innate immune factors, presumably myeloid cells, are important in regulating human STRC engraftment.


Asunto(s)
Trasplante de Células Madre de Sangre del Cordón Umbilical/métodos , Supervivencia de Injerto , Células Asesinas Naturales , Receptores de Interleucina-2/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/fisiología , Depleción Linfocítica , Ratones , Ratones Endogámicos NOD , Ratones SCID , Modelos Animales , Células Mieloides/inmunología , Factores de Tiempo , Trasplante Heterólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA