Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
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
2.
Blood ; 115(6): 1267-76, 2010 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-19846887

RESUMEN

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


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Cardiomiopatías/genética , Modelos Animales de Enfermedad , Errores Innatos del Metabolismo Lipídico/genética , Lipoproteínas/fisiología , Mutación/genética , Fitosteroles/metabolismo , Sitoesteroles/metabolismo , Trombocitopenia/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/genética , Animales , Tiempo de Sangría , Cardiomiopatías/patología , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Cruzamientos Genéticos , Femenino , Feto/citología , Feto/metabolismo , Errores Innatos del Metabolismo Lipídico/patología , Lipoproteínas/genética , Hígado/citología , Hígado/metabolismo , Masculino , Megacariocitos/citología , Megacariocitos/metabolismo , Ratones , Ratones Endogámicos A , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Trombocitopenia/patología
3.
Clin Immunol ; 135(1): 84-98, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20096637

RESUMEN

"Humanized" mouse models created by engraftment of immunodeficient mice with human hematolymphoid cells or tissues are an emerging technology with broad appeal across multiple biomedical disciplines. However, investigators wishing to utilize humanized mice with engrafted functional human immune systems are faced with a myriad of variables to consider. In this study, we analyze HSC engraftment methodologies using three immunodeficient mouse strains harboring the IL2rgamma(null) mutation; NOD-scid IL2rgamma(null), NOD-Rag1(null) IL2rgamma(null), and BALB/c-Rag1(null) IL2rgamma(null) mice. Strategies compared engraftment of human HSC derived from umbilical cord blood following intravenous injection into adult mice and intracardiac and intrahepatic injection into newborn mice. We observed that newborn recipients exhibited enhanced engraftment as compared to adult recipients. Irrespective of the protocol or age of recipient, both immunodeficient NOD strains support enhanced hematopoietic cell engraftment as compared to the BALB/c strain. Our data define key parameters for establishing humanized mouse models to study human immunity.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas/métodos , Células Madre Hematopoyéticas/inmunología , Subunidad gamma Común de Receptores de Interleucina/inmunología , Animales , Animales Recién Nacidos , Citometría de Flujo , Histocitoquímica , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Modelos Animales , Organismos Libres de Patógenos Específicos , Estadísticas no Paramétricas
4.
Ann N Y Acad Sci ; 1103: 77-89, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17332083

RESUMEN

There are many rodent models of autoimmune diabetes that have been used to study the pathogenesis of human type 1 diabetes (T1D), including the non-obese diabetic (NOD) mouse, the biobreeding (BB) rat, and the transgenic mouse models. However, mice and rats are not humans, and these rodent models do not completely recapitulate the autoimmune pathogenesis of the human disease. In addition, many of the reagents, tools, and therapeutics proposed for use in humans may be species specific and cannot be investigated in rodents. Researchers have used nonhuman primates to more closely mimic the human immune system and, to study species-specific therapeutics, but these studies are associated with additional ethical and economic constraints and, to date, no model of autoimmune diabetes in this species has been described. New animal models are needed that will permit the in vivo investigation of human immune systems and analyses of the pathogenesis of human T1D without putting individuals at risk. To fill this need, we are developing humanized mouse models for the in vivo study of T1D. These models are based on our newly generated stock of NOD-scid IL2rgamma(null) mice, which engraft at higher levels with human hematolymphoid cells and exhibit enhanced function of the engrafted human immune systems compared with previous humanized mouse models. Overall, development of these new generations of humanized mice should facilitate in vivo studies of the human immune system as well as permit the investigation of the pathogenesis and effector phases of human T1D.


Asunto(s)
Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Reproducibilidad de los Resultados
5.
Invest Ophthalmol Vis Sci ; 46(11): 4097-106, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16249486

RESUMEN

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


Asunto(s)
Quemaduras Químicas/metabolismo , Enfermedades de la Córnea/metabolismo , Quemaduras Oculares/inducido químicamente , Cicatrización de Heridas/fisiología , Animales , Trasplante de Médula Ósea , Quemaduras Químicas/inmunología , Quemaduras Químicas/patología , Quemaduras Químicas/cirugía , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Colagenasas/genética , Colagenasas/metabolismo , Enfermedades de la Córnea/inmunología , Enfermedades de la Córnea/patología , Enfermedades de la Córnea/cirugía , Modelos Animales de Enfermedad , Epitelio Corneal/fisiología , Citometría de Flujo , Regulación de la Expresión Génica/fisiología , Masculino , Metaloproteinasa 1 de la Matriz , Metaloproteinasas de la Matriz/genética , Metaloproteinasas de la Matriz/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos MRL lpr , Neutrófilos/fisiología , Análisis de Secuencia por Matrices de Oligonucleótidos , ARN Mensajero/metabolismo , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Hidróxido de Sodio , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/genética , Proteínas Supresoras de la Señalización de Citocinas/metabolismo
6.
Exp Biol Med (Maywood) ; 230(9): 659-67, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16179734

RESUMEN

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


Asunto(s)
Anemia/genética , Autoinmunidad/genética , Mutación , Proteínas/genética , Psoriasis/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Cartilla de ADN , Expresión Génica , Ligamiento Genético , Humanos , Linfocitos/inmunología , Ratones , Ratones Endogámicos , Datos de Secuencia Molecular , Fenotipo , Homología de Secuencia de Aminoácido
7.
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
8.
Cancer Genet ; 205(5): 232-41, 2012 May.
Artículo en Inglés | MEDLINE | ID: mdl-22682622

RESUMEN

Although researchers have yet to establish a link between muscular dystrophy (MD) and sarcomas in human patients, literature suggests that the MD genes dystrophin and dysferlin act as tumor suppressor genes in mouse models of MD. For instance, dystrophin-deficient mdx and dysferlin-deficient A/J mice, models of human Duchenne MD and limb-girdle MD type 2B, respectively, develop mixed sarcomas with variable penetrance and latency. To further establish the correlation between MD and sarcoma development, and to test whether a combined deletion of dystrophin and dysferlin exacerbates MD and augments the incidence of sarcomas, we generated dystrophin and dysferlin double mutant mice (STOCK-Dysf(prmd)Dmd(mdx-5Cv)). Not surprisingly, the double mutant mice develop severe MD symptoms and, moreover, develop rhabdomyosarcoma (RMS) at an average age of 12 months, with an incidence of >90%. Histological and immunohistochemical analyses, using a panel of antibodies against skeletal muscle cell proteins, electron microscopy, cytogenetics, and molecular analysis reveal that the double mutant mice develop RMS. The present finding bolsters the correlation between MD and sarcomas, and provides a model not only to examine the cellular origins but also to identify mechanisms and signal transduction pathways triggering development of RMS.


Asunto(s)
Modelos Animales de Enfermedad , Distrofina/genética , Proteínas de la Membrana/genética , Rabdomiosarcoma/genética , Animales , Células Cultivadas , Disferlina , Femenino , Marcadores Genéticos , Masculino , Ratones , Ratones Endogámicos C57BL , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Distrofia Muscular Animal/genética , Mutación , Cariotipificación Espectral
9.
Methods Mol Biol ; 602: 105-17, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20012395

RESUMEN

Immunodeficient mice have been used as recipients of human peripheral blood mononuclear cells (PBMC) for in vivo analyses of human xeno-graft-versus-host disease (GVHD). This xeno-GVHD model system in many ways mimics the human disease. The model system is established by intravenous or intraperitoneal injection of human PBMC or spleen cells into unconditioned or irradiated immunodeficient recipient mice. Recently, the development of several stocks of immunodeficient Prkdc ( scid ) (scid) and recombination activating 1 or 2 gene (Rag1 or Rag2) knockout mice bearing a targeted mutation in the gene encoding the IL2 receptor gamma chain (IL2rgamma) have been reported. The addition of the mutated IL2rgamma gene onto an immunodeficient mouse stock facilitates heightened engraftment with human PBMC. Stocks of mice with mutations in the IL2rgamma gene have been studied in several laboratories on NOD-scid, NOD-Rag1 ( null ), BALB/c-Rag1 ( null ), BALB/c-Rag2 ( null ), and Stock-H2(d)-Rag2 ( null ) strain backgrounds. Parameters to induce human xeno-GVHD in H2(d)-Rag2 ( null ) IL2rgamma ( null ) mice have been published, but variability in the frequency of disease and kinetics of GVHD were observed. The availability of the NOD-scid IL2rgamma ( null ) stock that engrafts more readily with human PBMC than does the Stock-H2(d)-Rag2 ( null ) IL2rgamma ( null ) stock should lead to a more reproducible humanized mouse model of GVHD and for the use in drug evaluation and validation. Furthermore, GVHD in human PBMC-engrafted scid mice has been postulated to result predominately from a human anti-mouse major histocompatibility complex (MHC) class II reactivity. Our recent development of NOD-scid IL2rgamma ( null ) beta2m ( null ) and NOD-scid IL2rgamma ( null ) Ab ( null ) stocks of mice now make it possible to investigate directly the role of host MHC class I and class II in the pathogenesis of GVHD in humanized mice using NOD-scid IL2rgamma ( null ) stocks that engraft at high levels with human PBMC and are deficient in murine MHC class I, class II, or both classes of MHC molecules.


Asunto(s)
Modelos Animales de Enfermedad , Genes MHC Clase II , Genes MHC Clase I , Enfermedad Injerto contra Huésped/inmunología , Complejo Mayor de Histocompatibilidad , Ratones Noqueados , Receptores de Interleucina-2/genética , Animales , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Femenino , Enfermedad Injerto contra Huésped/genética , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Complejo Mayor de Histocompatibilidad/genética , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Fenotipo , Receptores de Interleucina-2/inmunología , Trasplante Heterólogo/efectos adversos , Trasplante Heterólogo/inmunología , Microglobulina beta-2/genética , Microglobulina beta-2/metabolismo
10.
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
11.
J Immunol ; 174(10): 6477-89, 2005 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15879151

RESUMEN

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


Asunto(s)
Movilización de Célula Madre Hematopoyética , Linfopoyesis/genética , Linfopoyesis/inmunología , Mielopoyesis/genética , Mielopoyesis/inmunología , Trasplante de Células Madre de Sangre Periférica/métodos , Receptores de Interleucina-7/deficiencia , Receptores de Interleucina-7/genética , Envejecimiento/genética , Envejecimiento/inmunología , Animales , Recuento de Células Sanguíneas , Citotoxicidad Inmunológica/genética , Células Dendríticas/citología , Femenino , Citometría de Flujo , Movilización de Célula Madre Hematopoyética/métodos , Humanos , Inmunoglobulinas/sangre , Inmunofenotipificación , Subunidad gamma Común de Receptores de Interleucina , Células Asesinas Naturales/inmunología , Longevidad/genética , Longevidad/inmunología , Activación de Linfocitos/genética , Linfoma/genética , Linfoma/inmunología , Linfoma/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Tolerancia a Radiación/genética , Tolerancia a Radiación/inmunología , Receptores de Interleucina-2/deficiencia , Receptores de Interleucina-2/genética , Receptores de Interleucina-2/fisiología , Receptores de Interleucina-7/fisiología , Bazo/citología , Bazo/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología
12.
Clin Immunol ; 112(3): 273-83, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15308121

RESUMEN

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


Asunto(s)
Genes RAG-1/genética , Rechazo de Injerto/inmunología , Antígeno HLA-A2/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Islotes Pancreáticos/inmunología , Linfocitos/inmunología , Inmunodeficiencia Combinada Grave/inmunología , Animales , Glucemia/análisis , Diabetes Mellitus/sangre , Diabetes Mellitus/inducido químicamente , Diabetes Mellitus/inmunología , Diabetes Mellitus/cirugía , Eliminación de Gen , Enfermedad Injerto contra Huésped/inmunología , Humanos , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones Transgénicos , Perforina , Proteínas Citotóxicas Formadoras de Poros , Bazo/inmunología , Trasplante Homólogo/inmunología
13.
Hum Mol Genet ; 12(1): 61-9, 2003 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-12490533

RESUMEN

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


Asunto(s)
Ictiosis/genética , Mutación , Anomalía de Pelger-Huët/genética , Receptores Citoplasmáticos y Nucleares/genética , Animales , Modelos Animales de Enfermedad , Genotipo , Humanos , Ratones , Microscopía Electrónica , Anomalía de Pelger-Huët/patología , Fenotipo , Análisis de Secuencia de ADN , Receptor de Lamina B
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA