Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 306
Filtrar
1.
Biochem Biophys Res Commun ; 581: 12-19, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34653673

RESUMEN

CD8+ T cells play a critical role during adaptive immune response, which often change locations and expand or contract in numbers under different states. In the past, many attempts to develop CD8+T cells that express luciferase in vivo have involved the use of viral transduction, which has drawbacks of hardly tracked via detection of luciferase signal in untouched natural states. Here, we generate a transgenic mouse model via CRISPR-mediated genome editing, C57BL/6-CD8aem(IRES-AkaLuci-2A-EGFP) knock-in mice(CD8a-Aka mice), as a novel tool for non-invasive imaging of CD8+ T cells, which expressed a highly sensitive luciferase-Akaluciferase. Our study offers a convenient and robust tool for understanding fundamental CD8+ T cell biology in experimental applications and preclinical translational studies.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Sistemas CRISPR-Cas , Neoplasias del Colon/diagnóstico por imagen , Efecto Fundador , Edición Génica/métodos , Genoma , Ratones Transgénicos/genética , Animales , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/metabolismo , Línea Celular Tumoral , Neoplasias del Colon/genética , Neoplasias del Colon/inmunología , Diagnóstico por Imagen/métodos , Técnicas de Sustitución del Gen , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Xenoinjertos , Luciferasas/genética , Luciferasas/metabolismo , Mediciones Luminiscentes/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Ratones Transgénicos/inmunología , ARN Guía de Kinetoplastida/genética , ARN Guía de Kinetoplastida/metabolismo , Cigoto/inmunología , Cigoto/metabolismo
2.
Eur J Immunol ; 51(7): 1698-1714, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33949677

RESUMEN

CD4+ CD8+ double-positive thymocytes give rise to both conventional TCRαß+ T cells and invariant natural killer T cells (iNKT cells), but these two kinds of cells display different characteristics. The molecular mechanism underlying iNKT cell lineage development and function acquisition remain to be elucidated. We show that the loss of chromatin assembly factor 1B (CHAF1b) maintains the normal development of conventional TCRαß+ T cells but severely impairs early development of iNKT cells. This dysregulation is accompanied by the impairment in chromatin activation and gene transcription at Vα14-Jα18 locus. Notably, ectopic expression of a Vα14-Jα18 TCR rescues Chaf1b-deficient iNKT cell developmental defects. Moreover, cytokine secretion and antitumor activity are substantially maintained in Vα14-Jα18 TCR transgene-rescued Chaf1b-deficient iNKT cells. Our study identifies CHAF1b as a critical factor that controls the early development but not function acquisition of iNKT cells via lineage- and stage-specific regulation.


Asunto(s)
Factor 1 de Ensamblaje de la Cromatina/inmunología , Células T Asesinas Naturales/inmunología , Animales , Diferenciación Celular/inmunología , Línea Celular Tumoral , Linaje de la Célula/inmunología , Ensamble y Desensamble de Cromatina/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Timocitos/inmunología
3.
Immunohorizons ; 5(3): 147-156, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33789929

RESUMEN

NKT cells are primed lymphocytes that rapidly secrete cytokines and can directly kill cancerous cells. Given the critical role NKT cells play in cancer immune surveillance, we sought to investigate the effect of mutations in Brca1, specifically a conditional deletion of exon 11, on type I invariant NKT cell development. We observed a significant reduction in invariant NKT cells in both primary lymphoid and peripheral organs in Brca1 mutant mice compared with wild-type C57BL/6. However, the original Brca1 mutant strain was on a mixed background containing FVB/N. We determined that strain differences, rather than mutations in Brca1, led to the observed loss in NKT cells. Importantly, we found that whereas FVB/N mice lack Vß8, there was a striking increase in the total number of thymic type I CD1d-α-galactosylceramide tetramer positive NKT cells and skewing of the NKT cell population to NKT2 compared with C57BL/6 mice. Collectively, our data demonstrate the profound effect genetics can have on NKT cell subset differentiation.


Asunto(s)
Galactosilceramidas/inmunología , Ratones Transgénicos/inmunología , Células T Asesinas Naturales/inmunología , Ubiquitina-Proteína Ligasas/genética , Animales , Antígenos CD1d/biosíntesis , Diferenciación Celular/inmunología , Citocinas/biosíntesis , Galactosilceramidas/metabolismo , Hígado/inmunología , Hígado/metabolismo , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mutación , Células T Asesinas Naturales/metabolismo , Bazo/inmunología , Bazo/metabolismo , Timo/inmunología , Timo/metabolismo
4.
Clin Exp Immunol ; 205(2): 169-181, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-33864242

RESUMEN

The impact of treatment on the risk of lymphoma in patients with rheumatoid arthritis (RA) is unclear. Here, we aimed to assess if the risk of lymphoma differs according to the type of tumor necrosis factor inhibitor (TNFi), comparing monoclonal anti-TNF antibodies to the soluble TNF receptor. We used B cell activating factor belonging to the TNF family (BAFF)-transgenic (Tg) mice as a model of autoimmunity-associated lymphoma. Six-month-old BAFF-Tg mice were treated with TNFi for 12 months. Histological examination of the spleen, assessment of the cellular composition of the spleen by flow cytometry and assessment of B cell clonality were performed at euthanasia. Crude mortality and incidence of lymphoma were significantly higher in mice treated with monoclonal anti-TNF antibodies compared to both controls and mice treated with the soluble TNF receptor, even at a high dose. Flow cytometry analysis revealed decreased splenic macrophage infiltration in mice treated with monoclonal anti-TNF antibodies. Overall, this study demonstrates, for the first time, that a very prolonged treatment with monoclonal anti-TNF antibodies increase the risk of lymphoma in B cell-driven autoimmunity. These data suggest a closer monitoring for lymphoma development in patients suffering from B cell-driven autoimmune disease with long-term exposure to monoclonal anti-TNF antibodies.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Artritis Reumatoide/inmunología , Factor Activador de Células B/inmunología , Linfoma/inmunología , Ratones Transgénicos/inmunología , Inhibidores del Factor de Necrosis Tumoral/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Autoinmunidad/inmunología , Linfocitos B/inmunología , Línea Celular , Ratones , Ratones Endogámicos C57BL , Bazo/inmunología
5.
Sci Rep ; 10(1): 9722, 2020 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-32546687

RESUMEN

Accelerated timelines necessitate the discovery of fully human antibodies as biotherapeutics using transgenic animals with a notion that such mAbs bypass humanization. A transgenic animal derived mAb (PCa75) targeted against a prostate cancer antigen had several 'unusual residues' (rare somatic hypermutations, rSHM, with positional frequency of <1%) that resulted in compromised biophysical properties (Tm = 61 °C and intrinsic stability ΔGu = 24.3 kJ/mol) and a sub-optimal immunogenicity profile. In our quest for quality medicine, we pursued antibody engineering strategies to enhance the stability of PCa75. PCa62, an engineered variant of PCa75, retained function while significantly improving the drug-like attributes of the molecule (Tm = 75 °C and intrinsic stability ΔGu = 63.5 kJ/mol). rSHM is rather prevalent, 18 out the 21 approved transgenic animal-derived antibodies have at least one 'unusual residue'. Thus, engineering of rSHM remains critical to enhance the stability and minimize immunogenicity risk of biotherapeutics.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Neoplasias de la Próstata/inmunología , Ingeniería de Proteínas/métodos , Secuencia de Aminoácidos/genética , Animales , Animales Modificados Genéticamente , Anticuerpos Monoclonales/farmacología , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/inmunología , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/inmunología , Humanos , Masculino , Ratones , Ratones Transgénicos/inmunología , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/inmunología , Antígeno Prostático Específico/inmunología
6.
J Leukoc Biol ; 107(6): 993-1007, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32068302

RESUMEN

Cutting-edge questions in αß T cell biology were addressed by investigating a range of different genetically modified mouse models. In comparison, the γδ T cell field lacks behind on the availability of such models. Nevertheless, transgenic mouse models proved useful for the investigation of γδ T cell biology and their stepwise development in the thymus. In general, animal models and especially mouse models give access to a wide range of opportunities of modulating γδ T cells, which is unachievable in human beings. Because of their complex biology and specific tissue tropism, it is especially challenging to investigate γδ T cells in in vitro experiments since they might not reliably reflect their behavior and phenotype under physiologic conditions. This review aims to provide a comprehensive historical overview about how different transgenic mouse models contributed in regards of the understanding of γδ T cell biology, whereby a special focus is set on studies including the elusive role of the γδTCR. Furthermore, evolutionary and translational remarks are discussed under the aspect of future implications for the field. The ultimate full understanding of γδ T cells will pave the way for their usage as a powerful new tool in immunotherapy.


Asunto(s)
Linaje de la Célula/inmunología , Efecto Fundador , Ratones Transgénicos/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/genética , Linfocitos T/inmunología , Animales , Diferenciación Celular , Linaje de la Célula/genética , Movimiento Celular , Expresión Génica , Humanos , Inmunoterapia/métodos , Ratones , Ratones Transgénicos/genética , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/inmunología , Transducción de Señal , Especificidad de la Especie , Linfocitos T/clasificación , Linfocitos T/citología , Timo/citología , Timo/inmunología
8.
Front Immunol ; 9: 2599, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30581430

RESUMEN

Objective: Postoperative ileus (POI) is an inflammation-mediated complication of abdominal surgery, characterized by intestinal dysmotility and leukocyte infiltration into the muscularis externa (ME). Previous studies indicated that interleukin (IL)-10 is crucial for the resolution of a variety of inflammation-driven diseases. Herein, we investigated how IL-10 affects the postoperative ME inflammation and found an unforeseen role of IL-10 in POI. Design: POI was induced by a standardized intestinal manipulation (IM) in C57BL/6 and multiple transgenic mouse strain including C-C motif chemokine receptor 2-/-, IL-10-/-, and LysMcre/IL-10fl/fl mice. Leukocyte infiltration, gene and protein expression of cytokines, chemokines, and macrophage differentiation markers as well as intestinal motility were analyzed. IL-10 serum levels in surgical patients were determined by ELISA. Results: IL-10 serum levels were increased in patient after abdominal surgery. In mice, a complete or leucocyte-restricted IL-10 deficiency ameliorated POI and reduced the postoperative ME neutrophil infiltration. Infiltrating monocytes were identified as main IL-10 producers and undergo IL-10-dependent M2 polarization. Interestingly, M2 polarization is not crucial to POI development as abrogation of monocyte infiltration did not prevent POI due to a compensation of the IL-10 loss by resident macrophages and neutrophils. Organ culture studies demonstrated that IL-10 deficiency impeded neutrophil migration toward the surgically traumatized ME. This mechanism is mediated by reduction of neutrophil attracting chemokines. Conclusion: Monocyte-derived macrophages are the major IL-10 source during POI. An IL-10 deficiency decreases the postoperative expression of neutrophil-recruiting chemokines, consequently reduces the neutrophil extravasation into the postsurgical bowel wall, and finally protects mice from POI.


Asunto(s)
Ileus/inmunología , Interleucina-10/inmunología , Intestinos/inmunología , Leucocitos/inmunología , Complicaciones Posoperatorias/inmunología , Animales , Modelos Animales de Enfermedad , Motilidad Gastrointestinal/inmunología , Humanos , Inflamación/inmunología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/inmunología , Monocitos/inmunología , Infiltración Neutrófila/inmunología , Neutrófilos/inmunología , Periodo Posoperatorio
9.
Anticancer Res ; 38(8): 4481-4484, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30061213

RESUMEN

BACKGROUND: The aim of this study was to analyze the spatial distribution and proliferation of adoptively transferred CD8+ T-lymphocytes sensitized against allogeneic tumors. MATERIALS AND METHODS: Transgenic ß-actin-luc mice that express luciferase were sensitized against allogeneic SL2 lymphoma. CD8+ T-lymphocytes from these mice were transferred to lymphocyte-deficient, recombination activating gene-deficient (Rag-/-) mice bearing SL2 tumors and were tracked using bioluminescence imaging. RESULTS: Two out of six Rag-/- mice rejected their tumors. There were no apparent differences in spatial distribution and proliferative intensity of adoptively-transferred CD8+ T-lymphocytes between the two Rag-/- mice that rejected allogeneic SL2 tumors and the four Rag-/- mice that did not. CONCLUSION: The pattern of distribution in the mouse body and proliferative intensity of CD8+ T-lymphocytes do not seem to be decisive factors influencing allogeneic tumor rejection.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Rechazo de Injerto/inmunología , Neoplasias/inmunología , Traslado Adoptivo/métodos , Animales , Proliferación Celular/fisiología , Citotoxicidad Inmunológica/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/inmunología
10.
Infect Immun ; 86(1)2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-29038126

RESUMEN

Chlamydia trachomatis is the leading cause of infection-induced infertility in women. Attempts to control this epidemic with screening programs and antibiotic therapy have failed. Currently, a vaccine to prevent C. trachomatis infections is not available. In order to develop an animal model for evaluating vaccine antigens that can be applied to humans, we used C. trachomatis serovar D (strain UW-3/Cx) to induce infertility in mice whose major histocompatibility complex class II antigen was replaced with the human leukocyte antigen DR4 (HLA-DR4). Transcervical inoculation of medroxyprogesterone-treated HLA-DR4 transgenic mice with 5 × 105C. trachomatis D inclusion forming units (IFU) induced a significant reduction in fertility, with a mean number of embryos/mouse of 4.4 ± 1.3 compared to 7.8 ± 0.5 for the uninfected control mice (P < 0.05). A similar fertility reduction was elicited in the wild-type (WT) C57BL/6 mice (4.3 ± 1.4 embryos/mouse) compared to the levels of the WT controls (9.1 ± 0.4 embryos/mouse) (P < 0.05). Following infection, WT mice mounted more robust humoral and cellular immune responses than HLA-DR4 mice. As determined by vaginal shedding, HLA-DR4 mice were more susceptible to a transcervical C. trachomatis D infection than WT mice. To assess if HLA-DR4 transgenic and WT mice could be protected by vaccination, 104 IFU of C. trachomatis D was delivered intranasally, and mice were challenged transcervically 6 weeks later with 5 × 105 IFU of C. trachomatis D. As determined by severity and length of vaginal shedding, WT C57BL/6 and HLA-DR4 mice were significantly protected by vaccination. The advantages and limitations of the HLA-DR4 transgenic mouse model for evaluating human C. trachomatis vaccine antigens are discussed.


Asunto(s)
Infecciones por Chlamydia/inmunología , Chlamydia trachomatis/inmunología , Antígeno HLA-DR4/inmunología , Infertilidad/inmunología , Infertilidad/microbiología , Ratones Transgénicos/inmunología , Administración Intranasal/métodos , Animales , Anticuerpos Antibacterianos/inmunología , Vacunas Bacterianas/inmunología , Línea Celular Tumoral , Infecciones por Chlamydia/microbiología , Modelos Animales de Enfermedad , Femenino , Células HeLa , Humanos , Inmunidad Celular/inmunología , Inmunidad Humoral/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/microbiología , Vacunación/métodos , Vagina/inmunología , Vagina/microbiología
11.
J Immunol ; 199(12): 4165-4179, 2017 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-29084838

RESUMEN

We describe an MHC class II (I-Ab)-restricted TCR transgenic mouse line that produces CD4+ T cells specific for Plasmodium species. This line, termed PbT-II, was derived from a CD4+ T cell hybridoma generated to blood-stage Plasmodium berghei ANKA (PbA). PbT-II cells responded to all Plasmodium species and stages tested so far, including rodent (PbA, P. berghei NK65, Plasmodium chabaudi AS, and Plasmodium yoelii 17XNL) and human (Plasmodium falciparum) blood-stage parasites as well as irradiated PbA sporozoites. PbT-II cells can provide help for generation of Ab to P. chabaudi infection and can control this otherwise lethal infection in CD40L-deficient mice. PbT-II cells can also provide help for development of CD8+ T cell-mediated experimental cerebral malaria (ECM) during PbA infection. Using PbT-II CD4+ T cells and the previously described PbT-I CD8+ T cells, we determined the dendritic cell (DC) subsets responsible for immunity to PbA blood-stage infection. CD8+ DC (a subset of XCR1+ DC) were the major APC responsible for activation of both T cell subsets, although other DC also contributed to CD4+ T cell responses. Depletion of CD8+ DC at the beginning of infection prevented ECM development and impaired both Th1 and follicular Th cell responses; in contrast, late depletion did not affect ECM. This study describes a novel and versatile tool for examining CD4+ T cell immunity during malaria and provides evidence that CD4+ T cell help, acting via CD40L signaling, can promote immunity or pathology to blood-stage malaria largely through Ag presentation by CD8+ DC.


Asunto(s)
Presentación de Antígeno , Linfocitos T CD4-Positivos/inmunología , Antígenos CD40/inmunología , Células Dendríticas/inmunología , Malaria/inmunología , Ratones Transgénicos/inmunología , Parasitemia/inmunología , Linfocitos T Citotóxicos/inmunología , Animales , Antígenos de Protozoos/inmunología , Antígenos CD40/deficiencia , Ligando de CD40/inmunología , Células Cultivadas , Cruzamientos Genéticos , Hibridomas , Activación de Linfocitos , Malaria Cerebral/inmunología , Malaria Cerebral/prevención & control , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos/genética , Plasmodium berghei/inmunología , Quimera por Radiación
12.
PLoS One ; 12(3): e0173176, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28257518

RESUMEN

Although the role of T cells in autoimmunity has been explored for many years, the mechanisms leading to the initial priming of an autoimmune T cell response remain enigmatic. The 'hit and run' model suggests that self-antigens released upon cell death can provide the initial signal for a self-sustaining autoimmune response. Using a novel transgenic mouse model where we could induce the release of self-antigens via caspase-dependent apoptosis. We tracked the fate of CD8+ T cells specific for the self-antigen. Our studies demonstrated that antigens released from apoptotic cells were cross-presented by CD11c+ cells in the draining lymph node. This cross-presentation led to proliferation of self-antigen specific T cells, followed by a transient ability to produce IFN-γ, but did not lead to the development of autoimmune diabetes. Using this model we examined the consequences on T cell immunity when apoptosis was combined with dendritic cell maturation signals, an autoimmune susceptible genetic background, and the deletion of Tregs. The results of our study demonstrate that autoimmune diabetes cannot be initiated by the presentation of antigens released from apoptotic cells in vivo even in the presence of factors known to promote autoimmunity.


Asunto(s)
Autoantígenos/administración & dosificación , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Linfocitos T Reguladores/inmunología , Animales , Presentación de Antígeno/genética , Presentación de Antígeno/inmunología , Apoptosis/genética , Autoantígenos/inmunología , Autoinmunidad/genética , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/etiología , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Humanos , Tolerancia Inmunológica , Interferón gamma/genética , Ratones , Ratones Transgénicos/inmunología
13.
Proc Natl Acad Sci U S A ; 114(3): E396-E405, 2017 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-27994144

RESUMEN

Astrocytes are believed to bridge interactions between infiltrating lymphocytes and neurons during brain ischemia, but the mechanisms for this action are poorly understood. Here we found that interleukin-15 (IL-15) is dramatically up-regulated in astrocytes of postmortem brain tissues from patients with ischemic stroke and in a mouse model of transient focal brain ischemia. We generated a glial fibrillary acidic protein (GFAP) promoter-controlled IL-15-expressing transgenic mouse (GFAP-IL-15tg) line and found enlarged brain infarcts, exacerbated neurodeficits after the induction of brain ischemia. In addition, knockdown of IL-15 in astrocytes attenuated ischemic brain injury. Interestingly, the accumulation of CD8+ T and natural killer (NK) cells was augmented in these GFAP-IL-15tg mice after brain ischemia. Of note, depletion of CD8+ T or NK cells attenuated ischemic brain injury in GFAP-IL-15tg mice. Furthermore, knockdown of the IL-15 receptor α or blockade of cell-to-cell contact diminished the activation and effector function of CD8+ T and NK cells in GFAP-IL-15tg mice, suggesting that astrocytic IL-15 is delivered in trans to target cells. Collectively, these findings indicate that astrocytic IL-15 could aggravate postischemic brain damage via propagation of CD8+ T and NK cell-mediated immunity.


Asunto(s)
Astrocitos/metabolismo , Lesiones Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Inmunidad Celular/inmunología , Interleucina-15/inmunología , Interleucina-15/metabolismo , Anciano de 80 o más Años , Animales , Astrocitos/inmunología , Encéfalo/inmunología , Encéfalo/metabolismo , Lesiones Encefálicas/inmunología , Isquemia Encefálica/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Modelos Animales de Enfermedad , Femenino , Proteína Ácida Fibrilar de la Glía/inmunología , Proteína Ácida Fibrilar de la Glía/metabolismo , Humanos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/inmunología , Ratones Transgénicos/metabolismo , Neuroglía/inmunología , Neuroglía/metabolismo , Neuronas/inmunología , Neuronas/metabolismo , Regiones Promotoras Genéticas/inmunología , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/metabolismo , Regulación hacia Arriba/inmunología
14.
Toxicol Pathol ; 45(1): 134-145, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27815489

RESUMEN

Mice and humans branched from a common ancestor approximately 80 million years ago. Despite this, mice are routinely utilized as animal models of human disease and in drug development because they are inexpensive, easy to handle, and relatively straightforward to genetically manipulate. While this has led to breakthroughs in the understanding of genotype-phenotype relationships and in the identification of therapeutic targets, translation of beneficial responses to therapeutics from mice to humans has not always been successful. In a large part, these differences may be attributed to variations in the alignment of protein expression and signaling in the immune systems between mice and humans. Well-established inbred strains of "The Laboratory Mouse" vary in their immune response patterns as a result of genetic mutations and polymorphisms arising from intentional selection for research relevant traits, and even closely related substrains vary in their immune response patterns as a result of genetic mutations and polymorphisms arising from genetic drift. This article reviews some of the differences between the mouse and human immune system and between inbred mouse strains and shares examples of how these differences can impact the usefulness of mouse models of disease.


Asunto(s)
Ratones Endogámicos/inmunología , Ratones Transgénicos/inmunología , Modelos Animales , Polimorfismo Genético , Investigación Biomédica Traslacional , Animales , Ingeniería Genética , Humanos , Inmunidad Innata/genética , Células Asesinas Naturales/inmunología , Ratones Endogámicos/clasificación , Ratones Endogámicos/genética , Ratones Transgénicos/clasificación , Ratones Transgénicos/genética , Especificidad de la Especie
15.
Immunol Lett ; 180: 66-72, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27832962

RESUMEN

Recent studies demonstrated that activated CD8+ T cells contributed to the development of T1D, and Zinc Transporter 8 (ZnT8) has emerged as a target of autoreactive T cells in human T1D in recent years. In the previous work, we identified that ZnT8107-115 peptide as a candidate to generate CD8+ T cells and induce diabetes in mice. In addition, MHC-peptide complexes that interact with autoreactive T cells can induce immune tolerance. In the current study, we constructed ZnT8107-115/HLA-A2 dimers, and utilized them to immunize diabetes mice. The proliferation, cytotoxicity, and inflammatory cytokine of CD8+ T were analyzed, and the incidence and severity of diabetes were detected. We found that ZnT8107-115/HLA-A2 dimers inhibited proliferation, cytotoxicity, and inflammatory cytokine of CD8+ T. Additionally, ZnT8107-115/HLA-A2 dimers ameliorated the incidence and severity of diabetes mice. Our findings suggested that ZnT8107-115/HLA-A2 dimers abrogate pathogenic CD8+ T cells in diabetes, and the strategies represented promising way in T1D and other autoimmune diseases.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Experimental/inmunología , Antígeno HLA-A2/inmunología , Transportador 8 de Zinc/inmunología , Animales , Línea Celular , Proliferación Celular/fisiología , Citocinas/inmunología , Dimerización , Humanos , Inflamación/inmunología , Ratones , Ratones Transgénicos/inmunología , Péptidos/inmunología
16.
Cytokine ; 85: 83-91, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27300756

RESUMEN

IL-21 promotes autoimmune type-1 diabetes (T1D) in NOD mice by facilitating CD4(+) T cell help to CD8(+) T cells. IL-21 also enables autoreactive CD8(+) T cells to respond to weak TCR ligands and induce T1D. Here, we assessed whether IL-21 is essential for T1D induction in a mouse model where the disease can occur independently of CD4 help. In this model, which expresses lymphocytic choriomeningitis virus (LCMV) glycoprotein (GP) antigen under the rat insulin promoter (RIP-GP), LCMV infection activates CD8(+) T cells reactive to the GP-derived GP33 peptide that attack pancreatic islets and cause T1D. We show that IL-21 deficiency in RIP-GP mice did not impair T1D induction by LCMV expressing the wildtype GP33 peptide. Surprisingly, LCMV-L6F, expressing a weak peptide mimic of GP33, induced T1D more efficiently in Il21(-/-)RIP-GP mice than in controls. However, LCMV-C4Y expressing a very weak peptide mimic of GP33 did not induce T1D in Il21(-/-) mice, but T cells from the infected mice caused disease in lymphopenic RIP-GP mice upon adoptive transfer. Using Nur77(GFP) reporter mice, we show that CD8(+) T cells from Il21(-/-) mice expressing the GP33-specific transgenic P14 TCR showed increased reactivity towards low affinity TCR ligands. Collectively, our findings show that IL-21 is not always required for T1D induction by autoreactive CD8(+) T cells, and suggest that IL-21 may play an important role in regulating CD8(+) T cell reactivity towards low affinity TCR ligands.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Interleucinas/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Traslado Adoptivo/métodos , Animales , Antígenos Virales/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/virología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/virología , Insulina/inmunología , Islotes Pancreáticos/inmunología , Islotes Pancreáticos/virología , Ligandos , Activación de Linfocitos/inmunología , Recuento de Linfocitos/métodos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/virología , Virus de la Coriomeningitis Linfocítica/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Transgénicos/inmunología , Regiones Promotoras Genéticas/inmunología , Ratas
17.
J Immunol Methods ; 427: 42-50, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26410104

RESUMEN

In this study, we developed human immune system (HIS) mice that possess functional human CD4+ T cells and B cells, named HIS-CD4/B mice. HIS-CD4/B mice were generated by first introducing HLA class II genes, including DR1 and DR4, along with genes encoding various human cytokines and human B cell activation factor (BAFF) to NSG mice by adeno-associated virus serotype 9 (AAV9) vectors, followed by engrafting human hematopoietic stem cells (HSCs). HIS-CD4/B mice, in which the reconstitution of human CD4+ T and B cells resembles to that of humans, produced a significant level of human IgG against Plasmodium falciparum circumsporozoite (PfCS) protein upon immunization. CD4+ T cells in HIS-CD4/B mice, which possess central and effector memory phenotypes like those in humans, are functional, since PfCS protein-specific human CD4+ T cells secreting IFN-γ and IL-2 were detected in immunized HIS-CD4/B mice. Lastly, PfCS protein-immunized HIS-CD4/B mice were protected from in vivo challenge with transgenic P. berghei sporozoites expressing the PfCS protein. The immune sera collected from protected HIS-CD4/B mice reacted against transgenic P. berghei sporozoites expressing the PfCS protein and also inhibited the parasite invasion into hepatocytes in vitro. Taken together, these studies show that our HIS-CD4/B mice could mount protective human anti-malaria immunity, consisting of human IgG and human CD4+ T cell responses both specific for a human malaria antigen.


Asunto(s)
Modelos Animales de Enfermedad , Inmunidad Humoral/inmunología , Malaria Falciparum/inmunología , Ratones Transgénicos/inmunología , Animales , Ensayo de Inmunoadsorción Enzimática , Xenoinjertos , Antígenos de Histocompatibilidad Clase II , Humanos , Vacunas contra la Malaria , Ratones , Proteínas Protozoarias/inmunología
18.
Biomed Res Int ; 2015: 807146, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25759822

RESUMEN

Immunization against amyloid-beta-peptide (Aß) has been widely investigated as a potential immunotherapeutic approach for Alzheimer's disease (AD). With the aim of developing an active immunogenic vaccine without need of coadjuvant modification for human trials and therefore avoiding such side effects, we designed the Aß 1-42 vaccine (EB101), delivered in a liposomal matrix, that based on our previous studies significantly prevents and reverses the AD neuropathology, clearing Aß plaques while markedly reducing neuronal degeneration, behavioral deficits, and minimizing neuroinflammation in APP/PS1 transgenic mice. Here, the efficacy of our immunogenic vaccine EB101 was compared with the original immunization vaccine cocktail Aß 42 + CFA/IFA (Freund's adjuvant), in order to characterize the effect of sphingosine-1-phosphate (S1P) in the immunotherapeutic response. Quantitative analysis of amyloid burden showed a notable decrease in the neuroinflammation reaction against Aß plaques when S1P was compared with other treatments, suggesting that S1P plays a key role as a neuroprotective agent. Moreover, EB101 immunized mice presented a protective immunogenic reaction resulting in the increase of Aß-specific antibody response and decrease of reactive glia in the affected brain areas, leading to a Th2 immunological reaction.


Asunto(s)
Enfermedad de Alzheimer/inmunología , Precursor de Proteína beta-Amiloide/inmunología , Oligopéptidos/inmunología , Vacunas/inmunología , Péptidos beta-Amiloides/inmunología , Animales , Anticuerpos/inmunología , Formación de Anticuerpos/inmunología , Encéfalo/inmunología , Modelos Animales de Enfermedad , Humanos , Inmunización/métodos , Lisofosfolípidos/inmunología , Ratones , Ratones Transgénicos/inmunología , Fragmentos de Péptidos/inmunología , Placa Amiloide/inmunología , Esfingosina/análogos & derivados , Esfingosina/inmunología , Vacunación/métodos
19.
Pharm Res ; 32(7): 2344-59, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25630815

RESUMEN

PURPOSE: Protein aggregates have been discussed as a potential risk factor related to immunogenicity. Here we developed a novel human IgG transgenic (tg) mouse system expressing a mini-repertoire of human IgG1 antibodies (Abs) for the assessment of immunogenic properties of human mAb preparations. METHODS: Transgenic mice were generated using germline versions of the human Ig heavy chain γ1 (IgH-γ1), and the human Ig light chain (IgL) κ and λ genes. Only the soluble form of human IgH-γ1 was used to avoid expression of the membrane Ig-H chain and concomitant allelic exclusion of endogenous murine Ig genes. IgG1 aggregates were generated by different stress conditions such as process-related, low pH and exposure to artificial light. RESULTS: The expression of human Ig proteins induced immunological tolerance to a broad range of human IgG1 molecules in the tg mice. Immunization with IgG1 aggregates demonstrated that soluble oligomers induced by significant light-exposure and carrying neo-epitopes induced a strong immune response in tg mice. In contrast, Ab aggregates alone and monomers with neo-epitopes were not immunogenic. CONCLUSION: This mouse model is able to recognize immunogenic modifications of human IgG1. While the degree of stress-induced aggregation varies for different mAbs, our findings using a particular mAb (mAb1) demonstrate that non-covalently modified aggregates do not break tolerance, contrary to widely held opinion. The immunogenic potential of soluble aggregates of human IgG strongly depends on the presence of neo-epitopes resulting from harsh stress conditions, i.e. extensive exposure to artificial light.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Inmunoglobulina G/inmunología , Ratones Transgénicos/inmunología , Agregado de Proteínas/inmunología , Animales , Anticuerpos Monoclonales/genética , Formación de Anticuerpos , Secuencia de Bases , Citometría de Flujo , Humanos , Tolerancia Inmunológica , Inmunoglobulina G/genética , Cadenas Pesadas de Inmunoglobulina/genética , Cadenas Pesadas de Inmunoglobulina/inmunología , Cadenas Ligeras de Inmunoglobulina/genética , Cadenas Ligeras de Inmunoglobulina/inmunología , Ratones Transgénicos/genética , Datos de Secuencia Molecular , Agregado de Proteínas/genética , Estrés Psicológico/inmunología , Transgenes
20.
J Gen Virol ; 96(Pt 4): 804-814, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25516543

RESUMEN

Yellow fever virus (YFV)-17D is an empirically developed, highly effective live-attenuated vaccine that has been administered to human beings for almost a century. YFV-17D has stood as a paradigm for a successful viral vaccine, and has been exploited as a potential virus vector for the development of recombinant vaccines against other diseases. In this study, a DNA-launched YFV-17D construct (pBeloBAC-FLYF) was explored as a new modality to the standard vaccine to combine the commendable features of both DNA vaccine and live-attenuated viral vaccine. The DNA-launched YFV-17D construct was characterized extensively both in cell culture and in mice. High titres of YFV-17D were generated upon transfection of the DNA into cells, whereas a mutant with deletion in the capsid-coding region (pBeloBAC-YF/ΔC) was restricted to a single round of infection, with no release of progeny virus. Homologous prime-boost immunization of AAD mice with both pBeloBAC-FLYF and pBeloBAC-YF/ΔC elicited specific dose-dependent cellular immune response against YFV-17D. Vaccination of A129 mice with pBeloBAC-FLYF resulted in the induction of YFV-specific neutralizing antibodies in all vaccinated subjects. These promising results underlined the potential of the DNA-launched YFV both as an alternative to standard YFV-17D vaccination and as a vaccine platform for the development of DNA-based recombinant YFV vaccines.


Asunto(s)
ADN/genética , Vacuna contra la Fiebre Amarilla/genética , Vacuna contra la Fiebre Amarilla/inmunología , Fiebre Amarilla/inmunología , Fiebre Amarilla/virología , Virus de la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/inmunología , Animales , Anticuerpos Neutralizantes/inmunología , Anticuerpos Antivirales/inmunología , Línea Celular , Cricetinae , ADN/inmunología , Femenino , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos/genética , Ratones Transgénicos/inmunología , Vacunas Atenuadas/genética , Vacunas Atenuadas/inmunología , Vacunas de ADN/genética , Vacunas de ADN/inmunología , Vacunas Virales/genética , Vacunas Virales/inmunología , Replicación Viral/genética , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...