Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Front Immunol ; 13: 960329, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36420263

RESUMEN

Graft-versus-host disease (GvHD) is a major complication after allogeneic hematopoietic cell transplantation (HCT). Current strategies to prevent GvHD with immunosuppressive drugs carry significant morbidity and may affect the graft-versus-tumor (GVT) effect. Inflammatory bowel disease (IBD) is an intestinal inflammatory condition that affects more than 2 million people in the United States. Current strategies to prevent colitis with immunosuppressive drugs carry significant morbidity. Recently, Repulsive Guidance Molecule b (RGMb) has been identified as part of a signaling hub with neogenin and BMP receptors in mice and humans. In addition, RGMb binds BMP-2/4 in mice and humans as well as PD-L2 in mice. RGMb is expressed in the gut epithelium and by antigen presenting cells, and we found significantly increased expression in mouse small intestine after total body irradiation HCT conditioning. We hypothesized that RGMb may play a role in GvHD and IBD pathogenesis by contributing to mucosal inflammation. Using major-mismatched HCT mouse models, treatment with an anti-RGMb monoclonal antibody (mAb) that blocks the interaction with BMP-2/4 and neogenin prevented GvHD and improved survival compared to isotype control (75% versus 30% survival at 60 days after transplantation). The GVT effect was retained in tumor models. Using an inflammatory bowel disease dextran sulfate sodium model, treatment with anti-RGMb blocking monoclonal antibody but not isotype control prevented colitis and improved survival compared to control (73% versus 33% at 21 days after treatment) restoring gut homeostasis. Anti-RGMb mAb (9D1) treatment decreased IFN-γ and significantly increased IL-5 and IL-10 in the gut of the treated mice compared to the isotype control treated mice.


Asunto(s)
Colitis , Enfermedad Injerto contra Huésped , Enfermedades Inflamatorias del Intestino , Humanos , Ratones , Animales , Inflamación , Enfermedades Inflamatorias del Intestino/terapia , Colitis/inducido químicamente , Inmunosupresores , Anticuerpos Monoclonales/farmacología , Anticuerpos Monoclonales/uso terapéutico , Moléculas de Adhesión Celular Neuronal
2.
Blood Adv ; 4(17): 4232-4243, 2020 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-32898248

RESUMEN

Graft-versus-host disease (GVHD) is a complication of hematopoietic cell transplantation (HCT) caused by alloreactive T cells. Murine models of HCT are used to understand GVHD and T-cell reconstitution in GVHD target organs, most notably the gastrointestinal (GI) tract where the disease contributes most to patient mortality. T-cell receptor (TCR) repertoire sequencing was used to measure T-cell reconstitution from the same donor graft (C57BL/6 H-2b) in the GI tract of different recipients across a spectrum of matching, from syngeneic (C57BL/6), to minor histocompatibility (MHC) antigen mismatch BALB.B (H-2b), to major MHC mismatched B10.BR (H-2k) and BALB/c (H-2d). Although the donor T-cell pools had highly similar TCR, the TCR repertoire after HCT was very specific to recipients in each experiment independent of geography. A single invariant natural killer T clone was identifiable in every recipient group and was enriched in syngeneic recipients according to clonal count and confirmatory flow cytometry. Using a novel cluster analysis of the TCR repertoire, we could classify recipient groups based only on their CDR3 size distribution or TCR repertoire relatedness. Using a method for evaluating the contribution of common TCR motifs to relatedness, we found that reproducible sets of clones were associated with specific recipient groups within each experiment and that relatedness did not necessarily depend on the most common clones in allogeneic recipients. This finding suggests that TCR reconstitution is highly stochastic and likely does not depend on the evaluation of the most expanded TCR clones in any individual recipient but instead depends on a complex polyclonal architecture.


Asunto(s)
Microbioma Gastrointestinal , Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Animales , Humanos , Ratones , Linfocitos T , Trasplante Homólogo
3.
Blood Adv ; 3(21): 3419-3431, 2019 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-31714958

RESUMEN

Acute graft-versus-host disease (GVHD) is a leading cause of mortality after allogeneic hematopoietic cell transplantation (HCT) mediated by dysregulated T-cell immune reconstitution. Given the role of the T-cell immunoglobulin and mucin 1 (TIM-1) surface protein in many immune processes, including organ transplantation tolerance, we asked if TIM-1 might drive post-transplant inflammation and acute GVHD. TIM-1 binds to phosphatidylserine (PtdSer), and agonism of TIM1 on immune cells is proinflammatory. HCT conditioning results in a significant supply of PtdSer from apoptosis and cellular debris. Using murine models, treatment with an antagonistic anti-TIM-1 monoclonal antibody (mAb) protects against acute GVHD while maintaining graft-versus-tumor effects. In contrast, the addition of exogenous free PtdSer worsened GVHD in a TIM-1-dependent manner. Importantly, TIM-1 blockade did not alter the expansion of donor T cells in vitro or in vivo. Instead, TIM-1 blockade reduces proinflammatory cytokines and promotes anti-inflammatory factors like carbonic anhydrase 1 and serum amyloid A1 in the gut tissue. This is mediated by TIM-1 on donor cells, as HCT of wild-type (WT) bone marrow (BM) and conventional T (Tcon) cells into TIM-1-/- knockout (KO) recipient mice showed little survival advantage compared with WT recipients, whereas WT recipients of TIM-1-/- KO Tcon cells or TIM1-/- KO BM had improved survival, in part due to the expression of TIM-1 on donor invariant natural killer T cells, which drives inflammation. Finally, in a humanized mouse xenograft GVHD model, treatment with anti-human TIM-1 antagonist mAb reduced GVHD disease burden and mortality. This supports TIM-1 as important for GVHD pathogenesis and as a target for the prevention of GVHD.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Enfermedad Injerto contra Huésped/tratamiento farmacológico , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Receptor Celular 1 del Virus de la Hepatitis A/antagonistas & inhibidores , Animales , Anticuerpos Bloqueadores/uso terapéutico , Biomarcadores , Modelos Animales de Enfermedad , Expresión Génica , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/mortalidad , Trasplante de Células Madre Hematopoyéticas/métodos , Receptor Celular 1 del Virus de la Hepatitis A/genética , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Humanos , Reconstitución Inmune , Inmunohistoquímica , Inmunofenotipificación , Mediadores de Inflamación/metabolismo , Recuento de Linfocitos , Ratones , Ratones Noqueados , Índice de Severidad de la Enfermedad , Tasa de Supervivencia , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Homólogo
4.
Blood Adv ; 2(19): 2568-2580, 2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30301812

RESUMEN

Conventional local tumor irradiation (LTI), delivered in small daily doses over several weeks, is used clinically as a palliative, rather than curative, treatment for chemotherapy-resistant diffuse large B-cell lymphoma (DLBCL) for patients who are ineligible for hematopoietic cell transplantation. Our goal was to test the hypothesis that accelerated, but not conventional, LTI would be more curative by inducing T cell-mediated durable remissions. We irradiated subcutaneous A20 and BL3750 lymphoma tumors in mice with a clinically relevant total radiation dose of 30 Gy LTI, delivered in 10 doses of 3 Gy over 4 days (accelerated irradiation) or as 10 doses of 3 Gy over 12 days (conventional irradiation). Compared with conventional LTI, accelerated LTI resulted in more complete and durable tumor remissions. The majority of these mice were resistant to rechallenge with lymphoma cells, demonstrating the induction of memory antitumor immunity. The increased efficacy of accelerated LTI correlated with higher levels of tumor cell necrosis vs apoptosis and expression of "immunogenic cell death" markers, including calreticulin, heat shock protein 70 (Hsp70), and Hsp90. Accelerated LTI-induced remissions were not seen in immunodeficient Rag-2 -/- mice, CD8+ T-cell-depleted mice, or Batf-3 -/- mice lacking CD8α+ and CD103+ dendritic cells. Accelerated, but not conventional, LTI in immunocompetent hosts induced marked increases in tumor-infiltrating CD4+ and CD8+ T cells and MHCII+CD103+CD11c+ dendritic cells and corresponding reductions in exhausted PD-1+Eomes+CD8+ T cells and CD4+CD25+FOXP3+ regulatory T cells. These findings raise the possibility that accelerated LTI can provide effective immune control of human DLBCL.


Asunto(s)
Linfoma de Células B/inmunología , Linfocitos T/inmunología , Animales , Biomarcadores , Reactividad Cruzada/inmunología , Citocinas/metabolismo , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunidad , Inmunofenotipificación , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Linfocitos Infiltrantes de Tumor/patología , Linfoma de Células B/mortalidad , Linfoma de Células B/patología , Linfoma de Células B/radioterapia , Masculino , Ratones , Ratones Noqueados , Radioterapia/métodos , Inducción de Remisión , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
JCI Insight ; 2(20)2017 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-29046484

RESUMEN

Cellular therapies based on permanent genetic modification of conventional T cells have emerged as a promising strategy for cancer. However, it remains unknown if modification of T cell subsets, such as Tregs, could be useful in other settings, such as allograft transplantation. Here, we use a modular system based on a chimeric antigen receptor (CAR) that binds covalently modified mAbs to control Treg activation in vivo. Transient expression of this mAb-directed CAR (mAbCAR) in Tregs permitted Treg targeting to specific tissue sites and mitigated allograft responses, such as graft-versus-host disease. mAbCAR Tregs targeted to MHC class I proteins on allografts prolonged islet allograft survival and also prolonged the survival of secondary skin grafts specifically matched to the original islet allograft. Thus, transient genetic modification to produce mAbCAR T cells led to durable immune modulation, suggesting therapeutic targeting strategies for controlling alloreactivity in settings such as organ or tissue transplantation.


Asunto(s)
Tolerancia Inmunológica/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Linfocitos T/metabolismo , Animales , Trasplante de Médula Ósea , Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/inmunología , Antígenos de Histocompatibilidad Clase I , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante de Órganos , Receptores Quiméricos de Antígenos/genética , Factor de Transcripción STAT5 , Linfocitos T Reguladores/inmunología , Trasplante de Tejidos , Tolerancia al Trasplante/inmunología , Trasplante Homólogo
6.
PLoS One ; 11(11): e0166280, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27855183

RESUMEN

T lymphocytes require signals from self-peptides and cytokines, most notably interleukins 7 and 15 (IL-7, IL-15), for survival. While mouse T cells die rapidly if IL-7 or IL-15 is withdrawn, human T cells can survive prolonged withdrawal of IL-7 and IL-15. Here we show that IL-7 and IL-15 are required to maintain human T cell proliferative capacity through the STAT5 signaling pathway. T cells from humanized mice proliferate better if stimulated in the presence of human IL-7 or IL-15 or if T cells are exposed to human IL-7 or IL-15 in mice. Freshly isolated T cells from human peripheral blood lose proliferative capacity if cultured for 24 hours in the absence of IL-7 or IL-15. We further show that phosphorylation of STAT5 correlates with proliferation and inhibition of STAT5 reduces proliferation. These results reveal a novel role of IL-7 and IL-15 in maintaining human T cell function, provide an explanation for T cell dysfunction in humanized mice, and have significant implications for in vitro studies with human T cells.


Asunto(s)
Interleucina-15/metabolismo , Interleucina-7/metabolismo , Factor de Transcripción STAT5/metabolismo , Transducción de Señal , Linfocitos T/citología , Linfocitos T/metabolismo , Animales , Proliferación Celular , Humanos , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Activación de Linfocitos , Recuento de Linfocitos , Ratones , Fosforilación , Regulación hacia Arriba
7.
Oncogene ; 32(8): 1066-1072, 2013 Feb 21.
Artículo en Inglés | MEDLINE | ID: mdl-22484426

RESUMEN

Although numerous mouse models of B-cell malignancy have been developed via the enforced expression of defined oncogenic lesions, the feasibility of generating lineage-defined human B-cell malignancies using mice reconstituted with modified human hematopoietic stem cells (HSCs) remains unclear. In fact, whether human cells can be transformed as readily as murine cells by simple oncogene combinations is a subject of considerable debate. Here, we describe the development of humanized mouse model of MYC/BCL2-driven 'double-hit' lymphoma. By engrafting human HSCs transduced with the oncogene combination into immunodeficient mice, we generate a fatal B malignancy with complete penetrance. This humanized-MYC/BCL2-model (hMB) accurately recapitulates the histopathological and clinical aspects of steroid-, chemotherapy- and rituximab-resistant human 'double-hit' lymphomas that involve the MYC and BCL2 loci. Notably, this model can serve as a platform for the evaluation of antibody-based therapeutics. As a proof of principle, we used this model to show that the anti-CD52 antibody alemtuzumab effectively eliminates lymphoma cells from the spleen, liver and peripheral blood, but not from the brain. The hMB humanized mouse model underscores the synergy of MYC and BCL2 in 'double-hit' lymphomas in human patients. Additionally, our findings highlight the utility of humanized mouse models in interrogating therapeutic approaches, particularly human-specific monoclonal antibodies.


Asunto(s)
Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Animales , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Linfoma de Células B/genética , Ratones , Penetrancia , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , Análisis de Supervivencia
8.
Nat Commun ; 4: 2043, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23784006

RESUMEN

The B7/CD28 family has profound modulatory effects in immune responses and constitutes an important target for the development of novel therapeutic drugs against human diseases. Here we describe a new CD28 homologue (CD28H) that has unique functions in the regulation of the human immune response and is absent in mice. CD28H is constitutively expressed on all naive T cells. Repetitive antigenic exposure, however, induces a complete loss of CD28H on many T cells, and CD28H negative T cells have a phenotype of terminal differentiation and senescence. After extensive screening in a receptor array, a B7-like molecule, B7 homologue 5 (B7-H5), was identified as a specific ligand for CD28H. B7-H5 is constitutively found in macrophages and could be induced on dendritic cells. The B7-H5/CD28H interaction selectively costimulates human T-cell growth and cytokine production via an AKT-dependent signalling cascade. Our study identifies a novel costimulatory pathway regulating human T-cell responses.


Asunto(s)
Antígenos CD28/metabolismo , Activación de Linfocitos/inmunología , Proteínas de la Membrana/metabolismo , Linfocitos T/inmunología , Secuencia de Aminoácidos , Animales , Antígenos B7 , Antígenos CD28/química , Técnicas de Cocultivo , Epítopos/inmunología , Fluoresceínas/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Genoma Humano/genética , Células HEK293 , Humanos , Ligandos , Activación de Linfocitos/efectos de los fármacos , Proteínas de la Membrana/genética , Ratones , Datos de Secuencia Molecular , Fosforilación/efectos de los fármacos , Fosfotirosina/metabolismo , Unión Proteica/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/inmunología , Succinimidas/metabolismo , Linfocitos T/metabolismo , Vanadatos/farmacología
9.
PLoS One ; 6(4): e18382, 2011 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-21559522

RESUMEN

Increasing demand for human hematopoietic stem cells (HSCs) in clinical and research applications necessitates expansion of HSCs in vitro. Before these cells can be used they must be carefully evaluated to assess their stem cell activity. Here, we expanded cord blood CD34(+) CD133(+) cells in a defined medium containing angiopoietin like 5 and insulin-like growth factor binding protein 2 and evaluated the cells for stem cell activity in NOD-SCID Il2rg(-/-) (NSG) mice by multi-lineage engraftment, long term reconstitution, limiting dilution and serial reconstitution. The phenotype of expanded cells was characterized by flow cytometry during the course of expansion and following engraftment in mice. We show that the SCID repopulating activity resides in the CD34(+) CD133(+) fraction of expanded cells and that CD34(+) CD133(+) cell number correlates with SCID repopulating activity before and after culture. The expanded cells mediate long-term hematopoiesis and serial reconstitution in NSG mice. Furthermore, they efficiently reconstitute not only neonate but also adult NSG recipients, generating human blood cell populations similar to those reported in mice reconstituted with uncultured human HSCs. These findings suggest an expansion of long term HSCs in our culture and show that expression of CD34 and CD133 serves as a marker for HSC activity in human cord blood cell cultures. The ability to expand human HSCs in vitro should facilitate clinical use of HSCs and large-scale construction of humanized mice from the same donor for research applications.


Asunto(s)
Angiopoyetinas/metabolismo , Antígenos CD34/biosíntesis , Antígenos CD/biosíntesis , Glicoproteínas/biosíntesis , Subunidad gamma Común de Receptores de Interleucina/genética , Antígeno AC133 , Proteínas Similares a la Angiopoyetina , Animales , Plaquetas/metabolismo , Sangre Fetal/metabolismo , Citometría de Flujo/métodos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Péptidos
10.
Stem Cells Dev ; 20(8): 1371-81, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21142526

RESUMEN

Clinical and preclinical applications of human hematopoietic stem cells (HSCs) are often limited by scarcity of cells. Expanding human HSCs to increase their numbers while maintaining their stem cell properties has therefore become an important area of research. Here, we report a robust HSC coculture system wherein cord blood CD34(+) CD133(+) cells were cocultured with mesenchymal stem cells engineered to express angiopoietin-like-5 in a defined medium. After 11 days of culture, SCID repopulating cells were expanded ~60-fold by limiting dilution assay in NOD-scid Il2rg(-/-) (NSG) mice. The cultured CD34(+) CD133(+) cells had similar engraftment potential to uncultured CD34(+) CD133(+) cells in competitive repopulation assays and were capable of efficient secondary reconstitution. Further, the expanded cells supported a robust multilineage reconstitution of human blood cells in NSG recipient mice, including a more efficient T-cell reconstitution. These results demonstrate that the expanded CD34(+) CD133(+) cells maintain both short-term and long-term HSC activities. To our knowledge, this ~60-fold expansion of SCID repopulating cells is the best expansion of human HSCs reported to date. Further development of this coculture method for expanding human HSCs for clinical and preclinical applications is therefore warranted.


Asunto(s)
Angiopoyetinas/biosíntesis , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Antígeno AC133 , Proteínas Similares a la Angiopoyetina , Angiopoyetinas/genética , Angiopoyetinas/metabolismo , Animales , Antígenos CD/biosíntesis , Antígenos CD34/biosíntesis , Antígenos CD34/sangre , Técnicas de Cultivo de Célula/métodos , Células Cultivadas , Sangre Fetal/citología , Sangre Fetal/trasplante , Glicoproteínas/biosíntesis , Supervivencia de Injerto/inmunología , Células Madre Hematopoyéticas/inmunología , Humanos , Trasplante de Células Madre Mesenquimatosas , Ratones , Péptidos , Linfocitos T/trasplante , Trasplante Heterólogo/inmunología
11.
J Immunol ; 179(12): 8076-82, 2007 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-18056348

RESUMEN

Lyme disease, caused by the tick-borne spirochete Borrelia burgdorferi (Bb), is a multisystem illness, affecting many organs, such as the heart, the nervous system, and the joints. Months after Bb infection, approximately 60% of patients experience intermittent arthritic attacks, a condition that in some individuals progresses to chronic joint inflammation. Although mice develop acute arthritis in response to Bb infection, the joint inflammation clears after 2 wk, despite continuous infection, only very rarely presenting with chronic Lyme arthritis. Thus, the lack of an animal system has so far prevented the elucidation of this persistent inflammatory process that occurs in humans. In this study, we report that the majority of Bb-infected CD28-/- mice develop chronic Lyme arthritis. Consistent with observations in chronic Lyme arthritis patients, the infected mutant, but not wild-type mice present recurring monoarticular arthritis over an extended time period, as well as anti-outer surface protein A of Bb serum titers. Furthermore, we demonstrate that anti-outer surface protein A Abs develop in these mice only after establishment of chronic Lyme arthritis. Thus, the Bb-infected CD28-/- mice provide a murine model for studying chronic Lyme arthritis.


Asunto(s)
Borrelia burgdorferi , Antígenos CD28/genética , Modelos Animales de Enfermedad , Articulaciones/microbiología , Enfermedad de Lyme/genética , Ratones , Animales , Bacteriemia , Enfermedad Crónica , Ratones Noqueados
12.
Immunology ; 113(2): 234-45, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15379984

RESUMEN

Contact skin immunization of mice with reactive hapten antigen and subsequent airway challenge with the same hapten induces immediate airflow obstruction and subsequent airway hyper-reactivity (AHR) to methacholine challenge, which is dependent on B cells but not on T cells. This responsiveness to airway challenge with antigen is elicited as early as 1 day postimmunization and can be adoptively transferred to naïve recipients via 1-day immune cells. Responses are absent in 1-day immune B-cell-deficient JH(-/-) mice and B-1 B-cell-deficient xid male mice, as well as in recipients of 1-day immune cells depleted of cells with the B-1 cell phenotype (CD19(+) B220(+) CD5(+)). As B-1 cells produce immunoglobulin M (IgM), we sought and found significantly increased numbers of anti-hapten IgM-producing cells in the spleen and lymph nodes of 1-day immune wild-type mice, but not in xid mice. Then, we passively immunized naive mice with anti-hapten IgM monoclonal antibody and, following airway hapten challenge of the recipients, we showed both immediate airflow obstruction and AHR. In addition, AHR was absent in complement C5 and C5a receptor-deficient mice. In summary, this study of the very early elicited phase of a hapten asthma model suggests, for the first time, a role of B-1 cells in producing IgM to activate complement to rapidly mediate asthma airway reactivity only 1 day after immunization.


Asunto(s)
Asma/inmunología , Linfocitos B/inmunología , Hiperreactividad Bronquial/inmunología , Complemento C5a/inmunología , Inmunoglobulina M/inmunología , Animales , Asma/patología , Asma/fisiopatología , Hiperreactividad Bronquial/patología , Líquido del Lavado Bronquioalveolar/citología , Líquido del Lavado Bronquioalveolar/inmunología , Modelos Animales de Enfermedad , Femenino , Haptenos/inmunología , Inmunización/métodos , Pulmón/patología , Masculino , Cloruro de Metacolina/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Pruebas de Función Respiratoria , Sistema Respiratorio
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA