Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 132
Filtrar
1.
Biochem Biophys Res Commun ; 581: 12-19, 2021 12 03.
Artigo em Inglês | MEDLINE | ID: mdl-34653673

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Sistemas CRISPR-Cas , Neoplasias do Colo/diagnóstico por imagem , Efeito Fundador , Edição de Genes/métodos , Genoma , Camundongos Transgênicos/genética , Animais , Antígenos CD8/genética , Antígenos CD8/metabolismo , Linfócitos T CD8-Positivos/imunologia , Proteína 9 Associada à CRISPR/genética , Proteína 9 Associada à CRISPR/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/imunologia , Diagnóstico por Imagem/métodos , Técnicas de Introdução de Genes , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Xenoenxertos , Luciferases/genética , Luciferases/metabolismo , Medições Luminescentes/métodos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Camundongos Transgênicos/imunologia , RNA Guia de Cinetoplastídeos/genética , RNA Guia de Cinetoplastídeos/metabolismo , Zigoto/imunologia , Zigoto/metabolismo
2.
Eur J Immunol ; 51(7): 1698-1714, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33949677

RESUMO

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.


Assuntos
Fator 1 de Modelagem da Cromatina/imunologia , Células T Matadoras Naturais/imunologia , Animais , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Linhagem da Célula/imunologia , Montagem e Desmontagem da Cromatina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos/imunologia , Receptores de Antígenos de Linfócitos T alfa-beta/imunologia , Timócitos/imunologia
3.
Immunohorizons ; 5(3): 147-156, 2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33789929

RESUMO

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.


Assuntos
Galactosilceramidas/imunologia , Camundongos Transgênicos/imunologia , Células T Matadoras Naturais/imunologia , Ubiquitina-Proteína Ligases/genética , Animais , Antígenos CD1d/biossíntese , Diferenciação Celular/imunologia , Citocinas/biossíntese , Galactosilceramidas/metabolismo , Fígado/imunologia , Fígado/metabolismo , Ativação Linfocitária , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Células T Matadoras Naturais/metabolismo , Baço/imunologia , Baço/metabolismo , Timo/imunologia , Timo/metabolismo
4.
Clin Exp Immunol ; 205(2): 169-181, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33864242

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Artrite Reumatoide/imunologia , Fator Ativador de Células B/imunologia , Linfoma/imunologia , Camundongos Transgênicos/imunologia , Inibidores do Fator de Necrose Tumoral/imunologia , Fator de Necrose Tumoral alfa/imunologia , Animais , Doenças Autoimunes/imunologia , Autoimunidade/imunologia , Linfócitos B/imunologia , Linhagem Celular , Camundongos , Camundongos Endogâmicos C57BL , Baço/imunologia
5.
Sci Rep ; 10(1): 9722, 2020 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-32546687

RESUMO

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.


Assuntos
Anticorpos Monoclonais/imunologia , Neoplasias da Próstata/imunologia , Engenharia de Proteínas/métodos , Sequência de Aminoácidos/genética , Animais , Animais Geneticamente Modificados , Anticorpos Monoclonais/farmacologia , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Humanos , Masculino , Camundongos , Camundongos Transgênicos/imunologia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/imunologia , Antígeno Prostático Específico/imunologia
6.
Front Immunol ; 9: 2599, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30581430

RESUMO

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.


Assuntos
Íleus/imunologia , Interleucina-10/imunologia , Intestinos/imunologia , Leucócitos/imunologia , Complicações Pós-Operatórias/imunologia , Animais , Modelos Animais de Doenças , Motilidade Gastrointestinal/imunologia , Humanos , Inflamação/imunologia , Macrófagos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/imunologia , Monócitos/imunologia , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Período Pós-Operatório
7.
Anticancer Res ; 38(8): 4481-4484, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-30061213

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Rejeição de Enxerto/imunologia , Neoplasias/imunologia , Transferência Adotiva/métodos , Animais , Proliferação de Células/fisiologia , Citotoxicidade Imunológica/imunologia , Feminino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/imunologia
8.
Infect Immun ; 86(1)2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-29038126

RESUMO

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.


Assuntos
Infecções por Chlamydia/imunologia , Chlamydia trachomatis/imunologia , Antígeno HLA-DR4/imunologia , Infertilidade/imunologia , Infertilidade/microbiologia , Camundongos Transgênicos/imunologia , Administração Intranasal/métodos , Animais , Anticorpos Antibacterianos/imunologia , Vacinas Bacterianas/imunologia , Linhagem Celular Tumoral , Infecções por Chlamydia/microbiologia , Modelos Animais de Doenças , Feminino , Células HeLa , Humanos , Imunidade Celular/imunologia , Imunidade Humoral/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/microbiologia , Vacinação/métodos , Vagina/imunologia , Vagina/microbiologia
9.
J Immunol ; 199(12): 4165-4179, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-29084838

RESUMO

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.


Assuntos
Apresentação de Antígeno , Linfócitos T CD4-Positivos/imunologia , Antígenos CD40/imunologia , Células Dendríticas/imunologia , Malária/imunologia , Camundongos Transgênicos/imunologia , Parasitemia/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos de Protozoários/imunologia , Antígenos CD40/deficiência , Ligante de CD40/imunologia , Células Cultivadas , Cruzamentos Genéticos , Hibridomas , Ativação Linfocitária , Malária Cerebral/imunologia , Malária Cerebral/prevenção & controle , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/genética , Plasmodium berghei/imunologia , Quimera por Radiação
10.
Immunol Lett ; 180: 66-72, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27832962

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Diabetes Mellitus Experimental/imunologia , Antígeno HLA-A2/imunologia , Transportador 8 de Zinco/imunologia , Animais , Linhagem Celular , Proliferação de Células/fisiologia , Citocinas/imunologia , Dimerização , Humanos , Inflamação/imunologia , Camundongos , Camundongos Transgênicos/imunologia , Peptídeos/imunologia
11.
J Immunol Methods ; 427: 42-50, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26410104

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Imunidade Humoral/imunologia , Malária Falciparum/imunologia , Camundongos Transgênicos/imunologia , Animais , Ensaio de Imunoadsorção Enzimática , Xenoenxertos , Antígenos de Histocompatibilidade Classe II , Humanos , Vacinas Antimaláricas , Camundongos , Proteínas de Protozoários/imunologia
12.
Med Sci (Paris) ; 30(11): 949-51, 2014 Nov.
Artigo em Francês | MEDLINE | ID: mdl-25388574
13.
Clin Exp Immunol ; 178(1): 118-28, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24842626

RESUMO

Mouse monocyte/macrophage major histocompatibility complex (MHC) receptor 1 (MMR1; or MMR2) specific for H-2D(d) (or H-2K(d) ) molecules is expressed on monocytes from non-H-2D(d) (or non-H-2K(d) ), but not those from H-2D(d) (or H-2K(d) ), inbred mice. The MMR1 and/or MMR2 is essential for the rejection of H-2D(d) - and/or H-2K(d) -transgenic mouse skin onto C57BL/6 (H-2D(b) K(b) ) mice. Recently, we found that human leucocyte antigen (HLA)-B44 was the sole ligand of human MMR1 using microbeads that had been conjugated with 80 types of HLA class I molecules covering 94·2% (or 99·4%) and 92·4% (or 96·2%) of HLA-A and B molecules of Native Americans (or Japanese), respectively. In the present study, we also explored the ligand specificity of human MMR2 using microbeads. Microbeads coated with HLA-A32, HLA-B13 or HLA-B62 antigens bound specifically to human embryonic kidney (HEK)293T or EL-4 cells expressing human MMR2 and to the solubilized MMR2-green fluorescent protein (GFP) fusion protein; and MMR2(+) monocytes from a volunteer bound HLA-B62 molecules with a Kd of 8·7 × 10(-9) M, implying a three times down-regulation of MMR2 expression by the ligand expression. H-2K(d) (or H-2D(d) ) transgene into C57BL/6 mice down-regulated not only MMR2 (or MMR1) but also MMR1 (or MMR2) expression, leading to further down-regulation of MMR expression. In fact, monocytes from two (i.e. MMR1(+) /MMR2(+) and MMR1(-) /MMR2(-) ) volunteers bound seven to nine types of microbeads among 80, indicating ≤ 10 types of MMR expression on monocytes. The physiological role of constitutive MMRs on monocytes possibly towards allogeneic (e.g. fetal) cells in the blood appears to be distinct from that of inducible MMRs on macrophages toward allografts in tissue.


Assuntos
Regulação para Baixo/imunologia , Antígenos de Histocompatibilidade Classe I/imunologia , Macrófagos/imunologia , Monócitos/imunologia , Animais , Células HEK293 , Humanos , Ligantes , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/imunologia , Receptores Imunológicos/imunologia
14.
Gut ; 63(1): 43-53, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23161496

RESUMO

OBJECTIVE: Eosinophilic oesophagitis (EoE) is a chronic inflammatory condition of the oesophagus with limited treatment options. No previous transgenic model has specifically targeted the oesophageal mucosa to induce oesophageal eosinophilia. DESIGN: We developed a mouse model that closely resembles EoE by utilising oxazolone haptenation in mice with transgenic overexpression of an eosinophil poietic and survival factor (interleukin (IL)-5) in resident squamous oesophageal epithelia. RESULTS: Overexpression of IL-5 in the healthy oesophagus was achieved in transgenic mice (L2-IL5) using the squamous epithelial promoter Epstein-Barr virus ED-L2. Oxazolone-challenged L2-IL5 mice developed dose-dependent pan-oesophageal eosinophilia, including eosinophil microabscess formation and degranulation as well as basal cell hyperplasia. Moreover, oesophagi expressed increased IL-13 and the eosinophil agonist chemokine eotaxin-1. Treatment of these mice with corticosteroids significantly reduced eosinophilia and epithelial inflammation. CONCLUSIONS: L2-IL5 mice provide a novel experimental model that can potentially be used in preclinical testing of EoE-related therapeutics and mechanistic studies identifying pathogenetic features associated with mucosal eosinophilia.


Assuntos
Modelos Animais de Doenças , Esofagite Eosinofílica/etiologia , Interleucina-5/metabolismo , Camundongos Transgênicos , Animais , Anti-Inflamatórios/uso terapêutico , Biomarcadores/metabolismo , Dexametasona/uso terapêutico , Esofagite Eosinofílica/tratamento farmacológico , Esofagite Eosinofílica/metabolismo , Epitélio , Herpesvirus Humano 4 , Interleucina-5/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/genética , Camundongos Transgênicos/imunologia , Camundongos Transgênicos/metabolismo , Oxazolona , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima , Proteínas Virais Reguladoras e Acessórias
15.
Eur J Immunol ; 44(1): 296-306, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24127407

RESUMO

Mast cells are abundantly situated at contact sites between the body and its environment, such as the skin and, especially during certain immune responses, at mucosal surfaces. They mediate allergic reactions and degrade toxins as well as venoms. However, their roles during innate and adaptive immune responses remain controversial and it is likely that major functions remain to be discovered. Recent developments in mast cell-specific conditional gene targeting in the mouse promise to enhance our understanding of these fascinating cells. To complete the genetic toolbox to study mast cell development, homeostasis and function, it is imperative to inducibly manipulate their gene expression. Here, we report the generation of a novel knock-in mouse line expressing a tamoxifen-inducible version of the Cre recombinase from within the endogenous c-Kit locus. We demonstrate highly efficient and specific inducible expression of a fluorescent reporter protein in mast cells both in vivo and in vitro. Furthermore, induction of diphtheria toxin A expression allowed selective and efficient ablation of mast cells at various anatomical locations, while other hematopoietic cells remain unaffected. This novel mouse strain will hence be very valuable to study mast cell homeostasis and how specific genes influence their functions in physiology and pathology.


Assuntos
Toxina Diftérica/metabolismo , Marcação de Genes/métodos , Integrases/metabolismo , Mastócitos/imunologia , Camundongos Transgênicos/imunologia , Fragmentos de Peptídeos/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Morte Celular/genética , Toxina Diftérica/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Introdução de Genes , Loci Gênicos/genética , Integrases/genética , Mastócitos/efeitos dos fármacos , Mastócitos/patologia , Camundongos , Especificidade de Órgãos , Fragmentos de Peptídeos/genética , Proteínas Proto-Oncogênicas c-kit/genética , Tamoxifeno/administração & dosagem , Transgenes/genética
16.
Neoplasia ; 15(1): 85-94, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23359264

RESUMO

Chemotherapy enhances the antitumor adaptive immune T cell response, but the immunosuppressive tumor environment often dominates, resulting in cancer relapse. Antigen-presenting cells such as tumor-associated macrophages (TAMs) and tumor dendritic cells (TuDCs) are the main protagonists of tumor-infiltrating lymphocyte (TIL) immunosuppression. TAMs have been widely investigated and are associated with poor prognosis, but the immunosuppressive activity of TuDCs is less well understood. We performed two-photon imaging of the tumor tissue to examine the spatiotemporal interactions between TILs and TuDCs after chemotherapy. In a strongly immunosuppressive murine tumor model, cyclophosphamide-mediated chemotherapy transiently enhanced the antitumor activity of adoptively transferred ovalbumin-specific CD8(+) T cell receptor transgenic T cells (OTI) but barely affected TuDC compartment within the tumor. Time lapse imaging of living tumor tissue showed that TuDCs are organized as a mesh with dynamic interconnections. Once infiltrated into the tumor parenchyma, OTI T cells make antigen-specific and long-lasting contacts with TuDCs. Extensive analysis of TIL infiltration on histologic section revealed that after chemotherapy the majority of OTI T cells interact with TuDCs and that infiltration is restricted to TuDC-rich areas. We propose that the TuDC network exerts antigen-dependent unproductive retention that trap T cells and limit their antitumor effectiveness.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Dendríticas/imunologia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos T Citotóxicos/imunologia , Animais , Antígenos/imunologia , Feminino , Tolerância Imunológica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/imunologia , Receptores de Antígenos de Linfócitos T/imunologia
17.
Virology ; 435(1): 14-28, 2013 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-23217612

RESUMO

Work with human specific viruses will greatly benefit from the use of an in vivo system that provides human target cells and tissues in a physiological setting. In this regard humanized mice (hu-Mice) have played an important role in our understanding of viral pathogenesis and testing of therapeutic strategies. Limitations with earlier versions of hu-Mice that lacked a functioning human immune system are currently being overcome. The new generation hu-Mouse models are capable of multilineage human hematopoiesis and generate T cells, B cells, macrophages and dendritic cells required for an adaptive human immune response. Now any human specific pathogen that can infect humanized mice can be studied in the context of ongoing infection and immune responses. Two leading humanized mouse models are currently employed: the hu-HSC model is created by transplantation of human hematopoietic stem cells (HSC), whereas the BLT mouse model is prepared by transplantation of human fetal liver, thymus and HSC. A number of human specific viruses such as HIV-1, dengue, EBV and HCV are being studied intensively in these systems. Both models permit infection by mucosal routes with viruses such as HIV-1 thus allowing transmission prevention studies. Cellular and humoral immune responses are seen in both the models. While there is efficient antigen specific IgM production, IgG responses are suboptimal due to inefficient immunoglobulin class switching. With the maturation of T cells occurring in the autologous human thymus, BLT mice permit human HLA restricted T cell responses in contrast to hu-HSC mice. However, the strength of the immune responses needs further improvement in both models to reach the levels seen in humans. The scope of hu-Mice use is further broadened by transplantation of additional tissues like human liver thus permitting immunopathogenesis studies on hepatotropic viruses such as HCV. Numerous studies that encompass antivirals, gene therapy, viral evolution, and the generation of human monoclonal antibodies have been conducted with promising results in these mice. For further improvement of the new hu-Mouse models, ongoing work is focused on generating new strains of immunodeficient mice transgenic for human HLA molecules to strengthen immune responses and human cytokines and growth factors to improve human cell reconstitution and their homeostatic maintenance.


Assuntos
Camundongos Transgênicos/virologia , Linfócitos T/virologia , Viroses/virologia , Vírus/imunologia , Animais , Modelos Animais de Doenças , Efeito Fundador , Antígenos HLA/genética , Antígenos HLA/imunologia , Transplante de Células-Tronco Hematopoéticas , Humanos , Switching de Imunoglobulina , Imunoglobulinas , Transplante de Fígado/imunologia , Camundongos , Camundongos Transgênicos/imunologia , Linfócitos T/imunologia , Timo/imunologia , Timo/virologia , Transplante Heterólogo , Viroses/imunologia
18.
J Immunol ; 189(11): 5223-9, 2012 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-23089398

RESUMO

Engraftment of human hematopoietic stem cells into immunodeficient mice that lack T cells, B cells, and NK cells results in reconstitution of human blood lineage cells, especially B cells, in the recipient mice. However, these humanized mice do not make any significant level of IgG Ab in response to Ag stimulation. In this study, we show that in humanized mice, B cells are immature, and there is a complete deficiency of CD209(+) (DC-SIGN) human dendritic cells. These defects can be corrected by expression of human GM-CSF and IL-4 in humanized mice. As a result, these cytokine-treated humanized mice produced significant levels of Ag-specific IgG after immunization, including the production of neutralizing Abs specific for H5N1 avian influenza virus. A significant level of Ag-specific CD4 T cell response was also induced. Thus, we have identified defects in humanized mice and devised approaches to correct these defects such that the platform can be used for studying Ab responses and to generate novel human Abs against virulent pathogens and other clinically relevant targets.


Assuntos
Anticorpos Antivirais/imunologia , Linfócitos B/imunologia , Células Dendríticas/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Hospedeiro Imunocomprometido , Virus da Influenza A Subtipo H5N1/imunologia , Interleucina-4/imunologia , Linfócitos T/imunologia , Animais , Anticorpos Antivirais/biossíntese , Antígenos Virais/imunologia , Linfócitos B/citologia , Diferenciação Celular , Células Dendríticas/citologia , Feto , Efeito Fundador , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Humanos , Imunoglobulina G/biossíntese , Imunoglobulina G/imunologia , Interleucina-4/genética , Fígado/citologia , Fígado/imunologia , Camundongos , Camundongos Transgênicos/imunologia , Infecções por Orthomyxoviridae/genética , Infecções por Orthomyxoviridae/imunologia , Plasmídeos , Análise de Célula Única , Linfócitos T/citologia , Transfecção
19.
Immunol Lett ; 146(1-2): 1-7, 2012 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-22507217

RESUMO

The development of chimeric mice bearing the components of a human immune system to serve as animal models for "human" diseases has been the goal of many years of research. In this review we will discuss the latest advances in this field, their remaining shortcomings and challenges to the generation of new models of mouse/human chimeras.


Assuntos
Quimera/imunologia , Linfócitos/imunologia , Camundongos Transgênicos/imunologia , Modelos Animais , Animais , Quimera/genética , Modelos Animais de Doenças , Feto , Previsões , Efeito Fundador , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/imunologia , Humanos , Sistema Imunitário , Linfócitos/citologia , Camundongos , Camundongos Transgênicos/genética , Transplante Heterólogo
20.
J Exp Med ; 208(3): 617-29, 2011 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-21357741

RESUMO

Little is know about the nature of peripheral B cell tolerance or how it may vary in distinct lineages. Although autoantibody transgenic studies indicate that anergy and apoptosis are involved, some studies claim that receptor editing occurs. To model peripheral B cell tolerance in a normal, polyclonal immune system, we generated transgenic mice expressing an Igκ-light chain-reactive superantigen targeted to the plasma membrane of hepatocytes (pAlb mice). In contrast to mice expressing κ superantigen ubiquitously, in which κ cells edit efficiently to λ, in pAlb mice, κ B cells underwent clonal deletion. Their κ cells failed to populate lymph nodes, and the remaining splenic κ cells were anergic, arrested at a semi-mature stage without undergoing receptor editing. In the liver, κ cells recognized superantigen, down-regulated surface Ig, and expressed active caspase 3, suggesting ongoing apoptosis at the site of B cell receptor ligand expression. Some, apparently mature, κ B1 and follicular B cells persisted in the peritoneum. BAFF (B cell-activating factor belonging to the tumor necrosis factor family) overexpression rescued splenic κ B cell maturation and allowed κ cells to populate lymph nodes. Our model facilitates analysis of tissue-specific autoimmunity, tolerance, and apoptosis in a polyclonal B cell population. The results suggest that deletion, not editing, is the major irreversible pathway of tolerance induction among peripheral B cells.


Assuntos
Linfócitos B/imunologia , Proteínas da Membrana Bacteriana Externa/imunologia , Deleção Clonal/imunologia , Fígado/imunologia , Porinas/imunologia , Receptores Opioides kappa/imunologia , Receptores Virais/imunologia , Superantígenos/imunologia , Animais , Apoptose/imunologia , Autoimunidade/imunologia , Hepatócitos/imunologia , Cadeias kappa de Imunoglobulina/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA