Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
J Exp Med ; 192(2): 259-70, 2000 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-10899912

RESUMEN

The dominant lymphocytes in human and murine implantation sites are transient, pregnancy-associated uterine natural killer (uNK) cells. These cells are a major source of interferon (IFN)-gamma. Implantation sites in mice lacking uNK cells (alymphoid recombinase activating gene [RAG]-2(-/)- common cytokine receptor chain gamma [gamma(c)](-/)-) or IFN-gamma signaling (IFN-gamma(-/)- or IFN-gammaRalpha(-/)-) fail to initiate normal pregnancy-induced modification of decidual arteries and display hypocellularity or necrosis of decidua. To investigate the functions of uNK cell-derived IFN-gamma during pregnancy, RAG-2(-/)-gamma(c)(-/)- females were engrafted with bone marrow from IFN-gamma(-/)- mice, IFN-gamma signal-disrupted mice (IFN-gammaRalpha(-/)- or signal transducer and activator of transcription [Stat]-1(-/)-), or from mice able to establish normal uNK cells (severe combined immunodeficient [SCID] or C57BL/6). Mated recipients were analyzed at midgestation. All grafts established uNK cells. Grafts from IFN-gamma(-/)- mice did not reverse host vascular or decidual pathology. Grafts from all other donors promoted modification of decidual arteries and decidual cellularity. Grafts from IFN-gammaRalpha(-/)- or Stat-1(-/)- mice overproduced uNK cells, all of which were immature. Grafts from IFN-gamma(-/)-, SCID, or C57BL/6 mice produced normal, mature uNK cells. Administration of murine recombinant IFN-gamma to pregnant RAG-2(-/)-gamma(c)(-/)- mice initiated decidual vessel modification and promoted decidual cellularity in the absence of uNK cells. These in vivo findings strongly suggest that uNK cell-derived IFN-gamma modifies the expression of genes in the uterine vasculature and stroma, which initiates vessel instability and facilitates pregnancy-induced remodeling of decidual arteries.


Asunto(s)
Decidua/fisiología , Interferón gamma/fisiología , Células Asesinas Naturales/fisiología , Preñez/fisiología , Útero/irrigación sanguínea , Animales , Antígenos CD/fisiología , Proteínas de Unión al ADN/fisiología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones SCID , Embarazo , Receptores del Factor de Necrosis Tumoral/fisiología , Receptores Tipo I de Factores de Necrosis Tumoral , Factor de Necrosis Tumoral alfa/fisiología , Útero/inmunología
2.
J Exp Med ; 190(1): 141-4, 1999 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-10429678

RESUMEN

We have examined the question of whether there is an additional checkpoint in T cell development that regulates T cell receptor (TCR)-beta expression in CD25+44- thymocytes by mechanisms that are independent of the pre-TCR. Our analysis in various mutant mice indicates that all changes in cytoplasmic TCR-beta expression can be accounted for by pre-TCR-dependent signal mediation, putting into question the function of a putative pro-TCR.


Asunto(s)
Complejo CD3/fisiología , Regulación de la Expresión Génica , Glicoproteínas de Membrana/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Animales , Antígenos CD4/análisis , Antígenos CD8/análisis , Ratones , Ratones Endogámicos C57BL , Receptores de Interleucina-2/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo
3.
J Exp Med ; 189(3): 563-74, 1999 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-9927518

RESUMEN

Intracellular signals emanating from cytokine and antigen receptors are integrated during the process of intrathymic development. Still, the relative contributions of cytokine receptor signaling to pre-T cell receptor (TCR) and TCR-mediated differentiation remain undefined. Interleukin (IL)-7 interactions with its cognate receptor complex (IL-7Ralpha coupled to the common cytokine receptor gamma chain, gammac) play a dominant role in early thymopoiesis. However, alpha/beta T cell development in IL-7-, IL-7Ralpha-, and gammac-deficient mice is only partially compromised, suggesting that additional pathways can rescue alpha/beta T lineage cells in these mice. We have investigated the potential interdependence of gammac- and pre-TCR-dependent pathways during intrathymic alpha/beta T cell differentiation. We demonstrate that gammac-dependent cytokines do not appear to be required for normal pre-TCR function, and that the rate-limiting step in alpha/beta T cell development in gammac- mice does not involve TCR-beta chain rearrangements, but rather results from poor maintenance of early thymocytes. Moreover, mice double mutant for both gammac and pre-Talpha show vastly reduced thymic cellularity and a complete arrest of thymocyte differentiation at the CD44(+)CD25(+) cell stage. These observations demonstrate that the pre-TCR provides the gammac-independent signal which allows alpha/beta T cell development in gammac- mice. Thus, a series of overlapping signals derived from cytokine and T cell receptors guide the process of alpha/beta thymocyte development.


Asunto(s)
Células Madre Hematopoyéticas/inmunología , Receptor Cross-Talk , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Citocinas/inmunología , Linfocitos T/inmunología , Timo/inmunología , Animales , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , División Celular , Receptores de Hialuranos , Activación de Linfocitos , Ratones , Ratones Mutantes , Modelos Inmunológicos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Citocinas/genética , Receptores de Interleucina-2 , Timo/citología , Timo/crecimiento & desarrollo
4.
J Exp Med ; 193(12): 1413-24, 2001 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-11413196

RESUMEN

The product of the protooncogene Vav1 participates in multiple signaling pathways and is a critical regulator of antigen-receptor signaling in B and T lymphocytes, but its role during in vivo natural killer (NK) cell differentiation is not known. Here we have studied NK cell development in Vav1-/- mice and found that, in contrast to T and NK-T cells, the absolute numbers of phenotypically mature NK cells were not reduced. Vav1-/- mice produced normal amounts of interferon (IFN)-gamma in response to Listeria monocytogenes and controlled early infection but showed reduced tumor clearance in vivo. In vitro stimulation of surface receptors in Vav1-/- NK cells resulted in normal IFN-gamma production but reduced tumor cell lysis. Vav1 was found to control activation of extracellular signal-regulated kinases and exocytosis of cytotoxic granules. In contrast, conjugate formation appeared to be only mildly affected, and calcium mobilization was normal in Vav1-/- NK cells. These results highlight fundamental differences between proximal signaling events in T and NK cells and suggest a functional dichotomy for Vav1 in NK cells: a role in cytotoxicity but not for IFN-gamma production.


Asunto(s)
Proteínas de Ciclo Celular , Células Asesinas Naturales/inmunología , Proteínas Proto-Oncogénicas/fisiología , Transducción de Señal , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Calcio/metabolismo , Diferenciación Celular , Células Cultivadas , Citotoxicidad Inmunológica , Exocitosis , Interferón gamma/biosíntesis , Listeriosis/inmunología , Ratones , Ratones Noqueados , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias Experimentales/inmunología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas c-vav , Receptores Inmunológicos/metabolismo , Linfocitos T/inmunología
5.
J Exp Med ; 192(5): 659-70, 2000 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-10974032

RESUMEN

The cellular receptor for murine thymic stromal lymphopoietin (TSLP) was detected in a variety of murine, but not human myelomonocytic cell lines by radioligand binding. cDNA clones encoding the receptor were isolated from a murine T helper cell cDNA library. TSLP receptor (TSLPR) is a member of the hematopoietin receptor family. Transfection of TSLPR cDNA resulted in only low affinity binding. Cotransfection of the interleukin 7 (IL-7)Ralpha chain cDNA resulted in conversion to high affinity binding. TSLP did not activate cells from IL-7Ralpha(-/)- mice, but did activate cells from gammac(-/)- mice. Thus, the functional TSLPR requires the IL-7Ralpha chain, but not the gammac chain for signaling.


Asunto(s)
Hematopoyesis/efectos de los fármacos , Linfocitos/efectos de los fármacos , Receptores de Citocinas/fisiología , Receptores de Interleucina-7/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Línea Celular , Mapeo Cromosómico , Clonación Molecular , Citocinas/farmacología , Humanos , Interleucina-7/farmacología , Linfocitos/fisiología , Masculino , Ratones , Datos de Secuencia Molecular , ARN Mensajero/análisis , Receptores de Citocinas/química , Receptores de Citocinas/genética , Receptores de Interleucina-7/química , Proteínas Recombinantes/química , Transducción de Señal
6.
Curr Top Microbiol Immunol ; 324: 109-24, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-18481456

RESUMEN

The study of human hematopoiesis is conditioned by access to nondiseased human tissue samples that harbor the cellular substrates for this developmental process. Technical and ethical concerns limit the availability to tissues derived from the fetal and newborn periods, while adult samples are generally restricted to peripheral blood. Access to a small animal model that faithfully recapitulates the process of human hematopoiesis would provide an important tool. Natural killer (NK) cells comprise between 10% and 15% of human peripheral blood lymphocytes and appear conserved in several species. NK cells are implicated in the recognition of pathogen-infected cells and in the clearance of certain tumor cells. In this chapter, we discuss NK cell developmental pathways and the use of humanized murine models for the study of human hematopoiesis and, in particular, human NK cell development.


Asunto(s)
Diferenciación Celular , Hematopoyesis/fisiología , Células Asesinas Naturales/fisiología , Modelos Animales , Animales , Humanos , Ratones , Ratones SCID
7.
Transplant Proc ; 40(2): 624-30, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18374147

RESUMEN

Myoblast transfer therapy (MTT) is a strategy that has been proposed to treat some striated muscle pathologies. However, the first therapeutic trials using this technique were unsuccessful due to the limited migration and early cell death of the injected myoblasts. Various strategies have been considered to increase myoblast survival in the host muscle after MTT. Overexpression of heat shock proteins (HSPs) in mouse myoblasts has been shown to improve cell resistance against apoptosis in vitro and in vivo. Our objective was to determine whether heat shock (HS) treatment increased the survival of human myoblasts leading to better participation of the injected cells in muscle regeneration. For this study, HS-treated human myoblasts were injected into the tibialis anterior (TA) muscles of immunodeficient RAG-/- gammaC-/- mice. TA muscles were excised at 24 hour and at 1 month after injection. Our results showed that HS treatment increased the expression of the hsp70 protein and protected the cells from apoptosis in vitro. HS treatment dramatically increased the number of human fibers present at 1 month after injection when compared with nontreated cells. Interestingly, HS treatment decreased apoptosis at 24 hour after human myoblast injection, but no differences were observed concerning proliferation, suggesting that the increased fiber formation among the HS-treated group was probably due to decreased cell death. These data suggested that HS treatment might be used in the clinical context to improve the success of MTT.


Asunto(s)
Supervivencia de Injerto/fisiología , Mioblastos/trasplante , Trasplante Heterólogo/fisiología , Animales , Apoptosis , Células Cultivadas , Regulación de la Expresión Génica , Marcadores Genéticos , Proteínas HSP70 de Choque Térmico/genética , Calor , Humanos , Ratones , Ratones Noqueados , Ratones SCID , Músculo Esquelético/citología , Músculo Esquelético/fisiología , Enfermedades Musculares/cirugía , Mioblastos/citología , Mioblastos/fisiología , Resultado del Tratamiento
8.
Curr Biol ; 11(5): R175-7, 2001 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-11267886
9.
Curr Opin Immunol ; 10(2): 196-207, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9602309

RESUMEN

The early phases of T-cell development require both cell-cell interactions and soluble factors provided by stromal cells within the thymic microenvironment. Still, the precise nature of the signals delivered in vivo by cytokines (resulting in survival, proliferation or differentiation) remains unclear. Recent studies using mice deficient in cytokines or in their receptors have helped to identify essential signaling pathways required for the development of intrathymic precursors to mature alpha beta and gamma delta T cells. In addition, cytokine requirements for the development of natural killer cells were revealed in such mutants. The results obtained demonstrate that the development of all classes of lymphocytes (natural killer, gamma delta T cells and alpha beta T cells) is cytokine dependent, but the specific requirements differ for each lineage.


Asunto(s)
Proteínas Tirosina Quinasas Receptoras/fisiología , Receptores de Citocinas/fisiología , Linfocitos T/citología , Animales , Antígenos CD/fisiología , Diferenciación Celular/fisiología , Hematopoyesis/fisiología , Humanos , Interleucina-7/fisiología , Ratones , Proteínas Proto-Oncogénicas c-kit/fisiología , Receptores de Interleucina/fisiología , Receptores de Interleucina-7 , Transducción de Señal/fisiología , Factor de Células Madre/fisiología , Células del Estroma/fisiología , Linfocitos T/química , Timo/citología
10.
Curr Opin Immunol ; 12(2): 159-65, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10712942
11.
Mucosal Immunol ; 10(3): 602-612, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-27624779

RESUMEN

The human pathogen enteropathogenic Escherichia coli (EPEC), as well as the mouse pathogen Citrobacter rodentium, colonize the gut mucosa via attaching and effacing lesion formation and cause diarrheal diseases. EPEC and C. rodentium type III secretion system (T3SS) effectors repress innate immune responses and infiltration of immune cells. Inflammatory caspases such as caspase-1 and caspase-4/11 are crucial mediators of host defense and inflammation in the gut via their ability to process cytokines such as interleukin (IL)-1ß and IL-18. Here we report that the effector NleF binds the catalytic domain of caspase-4 and inhibits its proteolytic activity. Following infection of intestinal epithelial cells (IECs) EPEC inhibited caspase-4 and IL-18 processing in an NleF-dependent manner. Depletion of caspase-4 in IECs prevented the secretion of mature IL-18 in response to infection with EPECΔnleF. NleF-dependent inhibition of caspase-11 in colons of mice prevented IL-18 secretion and neutrophil influx at early stages of C. rodentium infection. Neither wild-type C. rodentium nor C. rodentiumΔnleF triggered neutrophil infiltration or IL-18 secretion in Cas11 or Casp1/11-deficient mice. Thus, IECs have a key role in modulating early innate immune responses in the gut via a caspase-4/11-IL-18 axis, which is targeted by virulence factors encoded by enteric pathogens.


Asunto(s)
Caspasas Iniciadoras/metabolismo , Caspasas/metabolismo , Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Escherichia coli Enteropatógena/inmunología , Infecciones por Escherichia coli/inmunología , Proteínas de Escherichia coli/metabolismo , Evasión Inmune , Mucosa Intestinal/inmunología , Factores de Virulencia/metabolismo , Animales , Caspasas/genética , Caspasas Iniciadoras/genética , Células Cultivadas , Citrobacter rodentium/patogenicidad , Escherichia coli Enteropatógena/patogenicidad , Proteínas de Escherichia coli/genética , Femenino , Humanos , Inmunidad Innata , Interleucina-18/metabolismo , Mucosa Intestinal/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Interferente Pequeño/genética , Factores de Virulencia/genética
12.
Cancer Res ; 61(4): 1717-26, 2001 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-11245488

RESUMEN

We undertook a series of systematic studies to address the role of fibroblast growth factor/fibroblast growth factor receptor (FGF/FGFR) activity in tumor growth and angiogenesis. We expressed dominant-negative FGFR2 (FGFR2-DN) or FGFR1 (FGFR1-DN) in glioma C6 cells by using constitutive or tetracycline-regulated expression systems. Anchorage-dependent or independent growth was inhibited in FGFR-DN-expressing cells. Tumor development after xenografting FGFR-DN-expressing cells in immunodeficient mice or after transplantation in rat brain was strongly inhibited. Quantification of microvessels demonstrated a significant decrease in vessel density in tumors derived from FGFR-DN-expressing cells. Furthermore, in a rabbit corneal assay, the angiogenic response after implantation of FGFR-DN-expressing cells was decreased. In tumors expressing FGFR-DN, vascular endothelial growth factor expression was strongly inhibited as compared with control tumor. These results indicate that inhibition of FGF activity may constitute a dominant therapeutic strategy in the treatment of FGF-producing cerebral malignancies and may disrupt both angiogenesis-dependent and -independent signals required for glioma growth and invasion.


Asunto(s)
Neoplasias Encefálicas/irrigación sanguínea , Neoplasias Encefálicas/patología , Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Glioma/irrigación sanguínea , Glioma/patología , Neovascularización Patológica/patología , Receptores de Factores de Crecimiento de Fibroblastos/antagonistas & inhibidores , Animales , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , División Celular/fisiología , Factores de Crecimiento Endotelial/biosíntesis , Factores de Crecimiento Endotelial/genética , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Factor 2 de Crecimiento de Fibroblastos/genética , Factor 4 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/biosíntesis , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/fisiología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Glioma/genética , Glioma/metabolismo , Linfocinas/biosíntesis , Linfocinas/genética , Masculino , Ratones , Neovascularización Patológica/metabolismo , Fenotipo , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/genética , Conejos , Ratas , Ratas Sprague-Dawley , Proteínas Tirosina Quinasas Receptoras/biosíntesis , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/biosíntesis , Receptores de Factores de Crecimiento de Fibroblastos/genética , Receptores de Factores de Crecimiento de Fibroblastos/fisiología , Tetraciclina/farmacología , Transfección , Factor A de Crecimiento Endotelial Vascular , Factores de Crecimiento Endotelial Vascular
13.
Hum Gene Ther ; 12(7): 823-31, 2001 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-11339898

RESUMEN

Design of efficient transplantation strategies for myoblast-based gene therapies in humans requires animal models in which xenografts are tolerated for long periods of time. In addition, such recipients should be able to withstand pretransplantation manipulations for enhancement of graft growth. Here we report that a newly developed immunodeficient mouse carrying two known mutations (the recombinase activating gene 2, RAG2, and the common cytokine receptor gamma, gammac) is a candidate fulfilling these requirements. Skeletal muscles from RAG2(-/-)/gammac(-/-) double mutant mice recover normally after myotoxin application or cryolesion, procedures commonly used to induce regeneration and improve transplantation efficiency. Well-differentiated donor-derived muscle tissue could be detected up to 9 weeks after transplantation of human myoblasts into RAG2(-/-)/gammac(-/-) muscles. These results suggest that the RAG2(-/-)/gammac(-/-) mouse model will provide new opportunities for human muscle research.


Asunto(s)
Trasplante de Células , Terapia Genética/métodos , Modelos Animales , Músculo Esquelético/citología , Músculo Esquelético/inmunología , Tolerancia al Trasplante , Animales , Diferenciación Celular , División Celular/efectos de los fármacos , Proteínas Cardiotóxicas de Elápidos/farmacología , Proteínas de Unión al ADN/genética , Distrofina/análisis , Eliminación de Gen , Humanos , Inmunohistoquímica , Subunidad gamma Común de Receptores de Interleucina , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Microscopía Fluorescente , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Proteínas Nucleares , Receptores de Interleucina-7/genética , Regeneración/efectos de los fármacos , Tolerancia al Trasplante/efectos de los fármacos , Tolerancia al Trasplante/genética , Tolerancia al Trasplante/inmunología , Trasplante Heterólogo
14.
Hum Gene Ther ; 14(12): 1169-79, 2003 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-12908968

RESUMEN

The limited success of human myoblast transplantation has been related to immune rejection, poor survival, and limited spread of injected myoblasts after transplantation. An important issue that has received little attention, but is nevertheless of fundamental importance in myoblast transplantation protocols, is the proliferative capacity of human satellite cells. Previous studies from our laboratory have demonstrated that the maximum number of divisions that a population of satellite cells can make decreases with age during the first two decades of life then stabilizes in adulthood. These observations indicate that when satellite cells are used as vectors in myoblast transplantation protocols it is important to consider donor age and the number of divisions that the cells have made prior to transplantation as limiting factors in obtaining an optimal number of donor derived muscle fibers. In this study, myoblasts derived from donors of different ages (newborn, 17 years old, and 71 years old) were isolated and amplified in culture. Their potential to participate in in vivo muscle regeneration in RAG2(-/-)/gamma(c)/C5 triple immunodeficient hosts after implantation was evaluated at 4 and 8 weeks postimplantation. Our results demonstrate that prolonged amplification in culture and the approach to replicative senescence are both important factors that may condition the success of myoblast transplantation protocols.


Asunto(s)
Senescencia Celular , Células Satélite del Músculo Esquelético/trasplante , Adolescente , Anciano , Animales , División Celular , Células Cultivadas , Senescencia Celular/fisiología , Femenino , Técnica del Anticuerpo Fluorescente , Rechazo de Injerto/inmunología , Humanos , Recién Nacido , Masculino , Ratones , Fibras Musculares Esqueléticas/citología , Proteínas Musculares/análisis , Proteínas Musculares/inmunología , Músculo Esquelético/química , Músculo Esquelético/citología , Regeneración
15.
J Interferon Cytokine Res ; 19(5): 533-41, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10386866

RESUMEN

Gene transduction into immature human hematopoietic cells collected from umbilical cord blood, bone marrow, or mobilized peripheral blood cells could be useful for the treatment of genetic and acquired disorders of the hematopoietic system. Immunodeficient mouse models have been used frequently as recipients to assay the growth and differentiation of human hematopoietic stem/progenitor cells. Indeed, high levels of human cell engraftment were first reported in human/murine chimeras using NOD/SCID mice, which now are considered as the standard for these types of experiments. However, NOD/SCID mice have some clear disadvantages (including spontaneous tumor formation) that limit their general use. We have developed a new immunodeficient mouse model by combining recombinase activating gene-2 (RAG2) and common cytokine receptor gamma chain (gamma c) mutations. The RAG2-/-/gamma c- double mutant mice are completely alymphoid (T-, B-, NK-), show no spontaneous tumor formation, and exhibit normal hematopoietic parameters. Interestingly, human cord blood cell engraftment in RAG2-/-/gamma c- mice was greatly enhanced by the exogenous administration of human cytokines interleukin-(IL-3) granulocyte-macrophage colony-stimulating factor, (GM-CSF), and erythropoietin in contrast to the NOD/SCID model. This unique feature of the RAG2-/-/gamma c- mouse model should be particularly well suited for assessing the role of different cytokines in human lymphopoiesis and stem/progenitor cell function in vivo.


Asunto(s)
Citocinas/farmacología , Trasplante de Células Madre Hematopoyéticas , Fragmentos de Péptidos/genética , Receptores de Citocinas/genética , Inmunodeficiencia Combinada Grave/genética , Animales , Antígenos CD34/sangre , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Humanos , Interleucina-3/farmacología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mutación , Proteínas Nucleares , Fenotipo , Receptores de Citocinas/química
16.
Int Rev Immunol ; 17(1-4): 103-20, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9914945

RESUMEN

Over the past decades, cytokines and their receptors have been shown to play a decisive role in the differentiation of both innate and adaptive immunity. The essential roles of cytokine/receptor pathways in vivo, however, have remained elusive and poorly defined. In some cases, primary immunodeficiency syndromes have provided the natural models in which the role of cytokines and their receptors in the development and function of the immune system have been elucidated. Animal models of cytokine/receptor deficiencies generated through gene targeting have also played a decisive factor in identifying the true biological roles of cytokine/receptor pathways. The end result of these approaches has been an enormous advance in our understanding of the cytokine control of normal and pathological human conditions, as well as the advent of new diagnostic tools and novel therapies.


Asunto(s)
Citocinas/deficiencia , Citocinas/genética , Síndromes de Inmunodeficiencia/genética , Receptores de Citocinas/deficiencia , Receptores de Citocinas/genética , Animales , Humanos , Síndromes de Inmunodeficiencia/metabolismo
17.
Placenta ; 21(7): 693-702, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-10985973

RESUMEN

Implantation sites from three strains of immunodeficient mice [tgepsilon26, IL-2Rbeta nullxp56(lck)null and IL-2Rgamma null, now known as common cytokine chain gamma (gamma(c)) null], which lack uterine natural killer (uNK) cells, are histologically abnormal. The related anomalies (found from day 10 of gestation) include the absence of aggregation of lymphocytes in the mesometrial triangle, acellularity of the mesometrial decidua, decidual arteries with relatively thick walls and reduced lumen diameters, unusual prominence of the endothelium in the major decidual vessels, and an overall reduction in placental size. In this study we have characterized implantation sites in a new mouse strain (gammac(-)/RAG2(-)) that is deficient in all lymphoid lineages. We have compared implantation sites in tgepsilon26 to gammac(-)/RAG2(-)at the ultrastructural level in order to determine the earliest-time point at which implantation sites differed from those in immunocompetent mice, and the cell types affected. Implantation sites from both the uNK cell-deficient mice resemble those from random-bred, immunocompetent mice on days 6 and 7 of gestation. On day 8 of gestation, decidual cells on the mesometrial sides of implantation sites in both tgepsilon26 and gammac(-)/RAG2(-)revealed pleotrophic morphology and degeneration. In some vessels, endothelial cells were distorted or displaced from their supporting cells. Progressive changes, suggestive of loss of function of both the mesometrial decidua and endothelial cells, were seen to day 14 of gestation, the latest time-point analysed. In contrast to tgepsilon26 mice, homozygously-mated gammac(-)/RAG2(-)had normal litter sizes, with birthweights and weaning weights similar to congenic C57Bl/6J controls, and no significant perinatal loss. In both strains, the newly-documented endothelial cell lesions predict detrimental alterations to vasomotor function of the uterine vasculature. These studies add strength to the hypothesis that uNK cells may have specialized physiological, rather than classically immune, functions in the pregnant mammalian uterus.


Asunto(s)
Implantación del Embrión , Síndromes de Inmunodeficiencia/patología , Células Asesinas Naturales/fisiología , Útero/ultraestructura , Animales , Decidua/ultraestructura , Femenino , Muerte Fetal/inmunología , Edad Gestacional , Síndromes de Inmunodeficiencia/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Microscopía Electrónica , Embarazo
18.
Placenta ; 21 Suppl A: S77-80, 2000.
Artículo en Inglés | MEDLINE | ID: mdl-10831128

RESUMEN

Mice genetically deficient in the natural killer (NK) cell lineage lack uterine (uNK cells) and demonstrate morphometrically-quantifiable histopathology within their implantation sites. Two particular mouse strains, tg(epsilon),26 and RAG-2 null x gamma(c) null, have been used successfully as transplant recipients to address questions relating to the biology of uNK cells. uNK cells did not differentiate within decidualized uterine graft segments from normal mice, which were anastomosed orthotopically into immunodeficient hosts. uNK cells did appear in similar grafts placed into immunocompetent hosts, indicating that uNK cells or their progenitors must home to the uterus. This was confirmed by splenocyte transplantation into pregnant uNK cell deficient recipients. Only splenocytes from pregnant donors, not those from non-pregnant donors, homed to the uterus. Homing in this in vivo assay was independent of the CC-chemokine receptors, CCR-2 and CCR-5. Longer-term bone marrow cell reconstitution of neonatal or virgin adult uNK cell-deficient mice has identified a functional role for uNK cells in modification of the decidual arterioles which is mediated by IFN-gamma. By utilizing mutant and gene-ablated mice as donors for tissue or haematopoietic cell transplants to uNK cell deficient mice, it should be possible to fully characterize the in vivo regulation and functions of these pregnancy-specific uterine lymphocytes.


Asunto(s)
Células Asesinas Naturales/inmunología , Preñez/inmunología , Útero/inmunología , Animales , Movimiento Celular , Implantación del Embrión/inmunología , Femenino , Ratones , Ratones Mutantes , Embarazo , Bazo/citología , Bazo/inmunología , Útero/citología , Útero/trasplante
19.
Mucosal Immunol ; 5(5): 555-66, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22569301

RESUMEN

Intestinal immune cells are important in host defense, yet the determinants for human lymphoid homeostasis in the intestines are poorly understood. In contrast, lymphoid homeostasis has been studied extensively in mice, where the requirement for a functional common γ-chain molecule has been established. We hypothesized that humanized mice could offer insights into human intestinal lymphoid homeostasis if generated in a strain with an intact mouse common γ-chain molecule. To address this hypothesis, we used three mouse strains (non-obese diabetic (NOD)/severe-combined immunodeficient (SCID) (N/S); NOD/SCID γ-chain(-/-) (NSG); and Rag2(-/-) γ-chain(-/-) (DKO)) and two humanization techniques (bone marrow liver thymus (BLT) and human CD34(+) cell bone marrow transplant of newborn mice (hu)) to generate four common types of humanized mice: N/S-BLT, NSG-BLT, NSG-hu, and DKO-hu mice. The highest levels of intestinal human T cells throughout the small and large intestines were observed in N/S-BLT mice, which have an intact common γ-chain molecule. Furthermore, the small intestine lamina propria T-cell populations of N/S-BLT mice exhibit a human intestine-specific surface phenotype. Thus, the extensive intestinal immune reconstitution of N/S-BLT mice was both quantitatively and qualitatively better when compared with the other models tested such that N/S-BLT mice are well suited for the analysis of human intestinal lymphocyte trafficking and human-specific diseases affecting the intestines.


Asunto(s)
Células de la Médula Ósea/inmunología , Subunidad gamma Común de Receptores de Interleucina/metabolismo , Intestinos/inmunología , Linfocitos T/inmunología , Quimera por Trasplante , Animales , Animales Recién Nacidos , Antígenos CD34/metabolismo , Trasplante de Médula Ósea , Proteínas de Unión al ADN , Modelos Animales de Enfermedad , Homeostasis , Humanos , Subunidad gamma Común de Receptores de Interleucina/genética , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID
20.
Blood ; 95(3): 984-91, 2000 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-10648413

RESUMEN

Fetal liver kinase ligands (flk2L/flt3L) and stem cell factor (SCF) have been shown to promote natural killer (NK) cell differentiation from hematopoietic stem cell (HSC) precursors in vitro. However, the contribution of signaling through the receptors for these growth factors for in vivo NK cell development remains ill-defined. We have analyzed the role of the SCF receptor c-kit in NK cell differentiation by reconstituting NK-deficient mice with fetal liver (FL) HSCs of c-kit(-/-) (W/W) mice. Although c-kit(-/-)NK cells were generated in W/W chimeras, they were reduced in number, contained a lower percentage of CD45R (B220)(+) cells, and were poorly cytolytic. In vitro experiments showed that generation of NK cells from FL precursors was reduced in the absence of c-kit signaling and that SCF promoted the survival of peripheral c-kit(+) NK cells. We conclude that c-kit/SCF interactions in vivo are dispensable for the commitment of HSC to the NK lineage, but they provide essential signals for generating normal numbers of fully mature NK cells.


Asunto(s)
Células Asesinas Naturales/citología , Proteínas Proto-Oncogénicas c-kit/fisiología , Factor de Células Madre/fisiología , Animales , Citotoxicidad Celular Dependiente de Anticuerpos , Subgrupos de Linfocitos B/citología , Diferenciación Celular , Supervivencia Celular/efectos de los fármacos , Células Cultivadas , Cruzamientos Genéticos , Citotoxicidad Inmunológica , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Trasplante de Tejido Fetal , Células Asesinas Activadas por Linfocinas/citología , Células Asesinas Activadas por Linfocinas/inmunología , Células Asesinas Activadas por Linfocinas/metabolismo , Células Asesinas Naturales/metabolismo , Hígado/citología , Hígado/embriología , Trasplante de Hígado , Recuento de Linfocitos , Proteínas de la Membrana/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Proteínas Proto-Oncogénicas c-kit/genética , Quimera por Radiación , Factor de Células Madre/deficiencia , Factor de Células Madre/genética , Factor de Células Madre/farmacología , Subgrupos de Linfocitos T/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA