Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Vis Exp ; (184)2022 06 29.
Artigo em Inglês | MEDLINE | ID: mdl-35848826

RESUMO

The human skin xenograft model, in which human donor skin is transplanted onto an immunodeficient mouse host, is an important option for translational research in skin immunology. Murine and human skin differ substantially in anatomy and immune cell composition. Therefore, traditional mouse models have limitations for dermatological research and drug discovery. However, successful xenotransplants are technically challenging and require optimal specimen and mouse graft site preparation for graft and host survival. The present protocol provides an optimized technique for transplanting human skin onto mice and discusses necessary considerations for downstream experimental aims. This report describes the appropriate preparation of a human donor skin sample, assembly of a surgical setup, mouse and surgical site preparation, skin transplantation, and post-surgical monitoring. Adherence to these methods allows for maintenance of xenografts for over 6 weeks post-surgery. The techniques outlined below allow maximum grafting efficiency due to the development of engineering controls, sterile technique, and pre- and post-surgical conditioning. Appropriate performance of the xenograft model results in long-lived human skin graft samples for experimental characterization of human skin and preclinical testing of compounds in vivo.


Assuntos
Transplante de Pele , Pele , Animais , Modelos Animais de Doenças , Xenoenxertos , Humanos , Imunidade , Camundongos , Transplante de Pele/métodos , Transplante Heterólogo
2.
Sci Immunol ; 7(69): eabi4613, 2022 03 18.
Artigo em Inglês | MEDLINE | ID: mdl-35302863

RESUMO

Transforming growth factor-ß1 (TGF-ß1) is inextricably linked to regulatory T cell (Treg) biology. However, precisely untangling the role for TGF-ß1 in Treg differentiation and function is complicated by the pleiotropic and context-dependent activity of this cytokine and the multifaceted biology of Tregs. Among CD4+ T cells, Tregs are the major producers of latent TGF-ß1 and are uniquely able to activate this cytokine via expression of cell surface docking receptor glycoprotein A repetitions predominant (GARP) and αv integrins. Although a preponderance of evidence indicates no essential roles for Treg-derived TGF-ß1 in Treg immunosuppression, TGF-ß1 signaling is crucial for Treg development in the thymus and periphery. Furthermore, active TGF-ß1 instructs the differentiation of other T cell subsets, including TH17 cells. Here, we will review TGF-ß1 signaling in Treg development and function and discuss knowledge gaps, future research, and the TGF-ß1/Treg axis in the context of cancer immunotherapy and fibrosis.


Assuntos
Linfócitos T Reguladores , Fator de Crescimento Transformador beta1 , Biologia , Tolerância Imunológica , Proteínas de Membrana/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta1/metabolismo
3.
J Immunol ; 207(7): 1763-1775, 2021 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-34470859

RESUMO

Regulatory T cells (Tregs) reside in nonlymphoid tissues where they carry out unique functions. The molecular mechanisms responsible for Treg accumulation and maintenance in these tissues are relatively unknown. Using an unbiased discovery approach, we identified LAYN (layilin), a C-type lectin-like receptor, to be preferentially and highly expressed on a subset of activated Tregs in healthy and diseased human skin. Expression of layilin on Tregs was induced by TCR-mediated activation in the presence of IL-2 or TGF-ß. Mice with a conditional deletion of layilin in Tregs had reduced accumulation of these cells in tumors. However, these animals somewhat paradoxically had enhanced immune regulation in the tumor microenvironment, resulting in increased tumor growth. Mechanistically, layilin expression on Tregs had a minimal effect on their activation and suppressive capacity in vitro. However, expression of this molecule resulted in a cumulative anchoring effect on Treg dynamic motility in vivo. Taken together, our results suggest a model whereby layilin facilitates Treg adhesion in skin and, in doing so, limits their suppressive capacity. These findings uncover a unique mechanism whereby reduced Treg motility acts to limit immune regulation in nonlymphoid organs and may help guide strategies to exploit this phenomenon for therapeutic benefit.


Assuntos
Proteínas de Transporte/metabolismo , Glicoproteínas de Membrana/metabolismo , Receptores de Retorno de Linfócitos/metabolismo , Pele/imunologia , Linfócitos T Reguladores/imunologia , Animais , Proteínas de Transporte/genética , Movimento Celular , Células Cultivadas , Humanos , Tolerância Imunológica , Ativação Linfocitária , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Imunológicos , Especificidade de Órgãos , Receptores de Retorno de Linfócitos/genética , Fator de Crescimento Transformador beta/metabolismo
4.
J Exp Med ; 217(9)2020 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-32539073

RESUMO

Tumor-infiltrating CD8+ T cells mediate antitumor immune responses. However, the mechanisms by which T cells remain poised to kill cancer cells despite expressing high levels of inhibitory receptors are unknown. Here, we report that layilin, a C-type lectin domain-containing membrane glycoprotein, is selectively expressed on highly activated, clonally expanded, but phenotypically exhausted CD8+ T cells in human melanoma. Lineage-specific deletion of layilin on murine CD8+ T cells reduced their accumulation in tumors and increased tumor growth in vivo. Congruently, gene editing of LAYN in human CD8+ T cells reduced direct tumor cell killing ex vivo. On a molecular level, layilin colocalized with integrin αLß2 (LFA-1) on T cells, and cross-linking layilin promoted the activated state of this integrin. Accordingly, LAYN deletion resulted in attenuated LFA-1-dependent cellular adhesion. Collectively, our results identify layilin as part of a molecular pathway in which exhausted or "dysfunctional" CD8+ T cells enhance cellular adhesiveness to maintain their cytotoxic potential.


Assuntos
Proteínas de Transporte/metabolismo , Imunidade , Integrinas/metabolismo , Lectinas Tipo C/metabolismo , Glicoproteínas de Membrana/metabolismo , Neoplasias/imunologia , Animais , Linfócitos T CD8-Positivos/imunologia , Adesão Celular , Proliferação de Células , Células Clonais , Citocinas/biossíntese , Citotoxicidade Imunológica , Edição de Genes , Humanos , Ativação Linfocitária/imunologia , Antígeno-1 Associado à Função Linfocitária/metabolismo , Linfócitos do Interstício Tumoral/imunologia , Melanoma/imunologia , Melanoma/patologia , Camundongos Endogâmicos C57BL , Metástase Neoplásica , Neoplasias/patologia , Ligação Proteica , Talina/metabolismo
5.
J Immunother Cancer ; 7(1): 355, 2019 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-31856922

RESUMO

Cytokines of the common γ-chain receptor family such as IL-15 are vital with respect to activating immune cells, sustaining healthy immune functions, and augmenting the anti-tumor activity of effector cells, making them ideal candidates for cancer immunotherapy. IL-15, either in its soluble form (IL-15sol) or complexed with IL-15Rα (IL-15Rc), has been shown to exhibit potent anti-tumor activities in various experimental cancer studies. Here we describe the impact of intraperitoneal IL-15 in a cancer cell-delivered IL-15 immunotherapy approach using the 70Z/3-L leukemia mouse model. Whereas both forms of IL-15 led to significantly improved survival rates compared to the parent cell line, there were striking differences in the extent of the improved survival: mice receiving cancer cells secreting IL-15sol showed significantly longer survival and protective long-term immunity compared to those producing IL-15Rc. Interestingly, injection of leukemia cells secreting IL-15sol lead to heightened expansion of CD4+ and CD8+ T-cell populations in the peritoneum compared to IL-15Rc. Cell-secreted IL-15Rc resulted in an influx and/or expansion of NK1.1+ cells in the peritoneum which was much less pronounced in the IL-15sol model. Furthermore, IL-15Rc but not IL-15sol lead to T-cell exhaustion and disease progression. To our knowledge, this is the first study detailing a significantly different biological effect of cell-delivered IL-15sol versus IL-15Rc in a mouse cancer immunotherapy study.


Assuntos
Imunomodulação , Imunoterapia , Interleucina-15/metabolismo , Leucemia/etiologia , Leucemia/metabolismo , Receptores de Interleucina-15/metabolismo , Animais , Linhagem Celular Tumoral , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Expressão Gênica , Humanos , Interleucina-15/sangue , Interleucina-15/genética , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Leucemia/patologia , Leucemia/terapia , Melanoma Experimental , Camundongos , Ligação Proteica , Receptores de Interleucina-15/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Transdução Genética , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Cell Rep ; 27(8): 2304-2312.e6, 2019 05 21.
Artigo em Inglês | MEDLINE | ID: mdl-31116977

RESUMO

Mechanisms that govern transcriptional regulation of inflammation in atherosclerosis remain largely unknown. Here, we identify the nuclear transcription factor c-Myb as an important mediator of atherosclerotic disease in mice. Atherosclerosis-prone animals fed a diet high in cholesterol exhibit increased levels of c-Myb in the bone marrow. Use of mice that either harbor a c-Myb hypomorphic allele or where c-Myb has been preferentially deleted in B cell lineages revealed that c-Myb potentiates atherosclerosis directly through its effects on B lymphocytes. Reduced c-Myb activity prevents the expansion of atherogenic B2 cells yet associates with increased numbers of IgM-producing antibody-secreting cells (IgM-ASCs) and elevated levels of atheroprotective oxidized low-density lipoprotein (OxLDL)-specific IgM antibodies. Transcriptional profiling revealed that c-Myb has a limited effect on B cell function but is integral in maintaining B cell progenitor populations in the bone marrow. Thus, targeted disruption of c-Myb beneficially modulates the complex biology of B cells in cardiovascular disease.


Assuntos
Células Produtoras de Anticorpos/imunologia , Aterosclerose/genética , Aterosclerose/imunologia , Imunoglobulina M/metabolismo , Proteínas Proto-Oncogênicas c-myb/genética , Proteínas Proto-Oncogênicas c-myb/imunologia , Animais , Células Produtoras de Anticorpos/metabolismo , Aterosclerose/patologia , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Genes myb , Masculino , Camundongos
7.
J Leukoc Biol ; 105(3): 507-518, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30576006

RESUMO

B cell development is regulated by stromal cells (SCs) that form a supportive microenvironment. These SCs along with other cell types produce cytokines, chemokines, and adhesion molecules that guide B cell commitment and differentiation. BM, spleen (Sp), and the gut lamina propria (LP) constitute distinctive anatomical compartments that support B cell differentiation. In order to characterize and compare the signals necessary to generate IgA+ B cells, we developed an in vitro system to co-culture gut LP, BM, or Sp-derived SCs with B lineage cells. Using this co-culture system, we found that gut LP SCs promote IgA+ B cell accumulation through the production of soluble stimulatory factors. In contrast to gut LP SCs, BM and splenic SCs were found to impair IgA+ B cell accumulation in vitro. Taken together, these observations provide new insights into how SCs derived from different anatomical locations shape IgA+ B cell responses.


Assuntos
Linfócitos B/metabolismo , Imunoglobulina A/metabolismo , Células Estromais/metabolismo , Animais , Fator Ativador de Células B/metabolismo , Diferenciação Celular , Linhagem Celular , Feminino , Mucosa Intestinal/citologia , Camundongos Endogâmicos C57BL , Solubilidade , Células Estromais/citologia
8.
PLoS One ; 12(9): e0185509, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28957409

RESUMO

Immature B cells are the first B cell progenitors to express a fully formed B cell receptor and are therefore subject to extensive selection processes that act to mitigate the emergence of autoreactive clones. While it is well appreciated that most B cell generation in the bone marrow is highly dependent on access to molecules present in the local milieu, the existence of extrinsically provided factors that modulate immature B cell biology is ambiguous. Nonetheless, a population of CD49b+CD90lo cells has demonstrated in vitro potential to promote immature B cell survival. Using a mouse basophil reporter strain we confirmed the identity of these CD49b+CD90lo supportive cells as basophils. However, analysis of bone marrow B cell populations following lineage specific basophil depletion demonstrates that basophils do not have a significant role in vivo in modulating immature B cell biology during steady-state conditions.


Assuntos
Basófilos/citologia , Células da Medula Óssea/citologia , Células Precursoras de Linfócitos B/citologia , Animais , Basófilos/metabolismo , Células da Medula Óssea/metabolismo , Linhagem da Célula , Sobrevivência Celular , Técnicas de Cocultura , Citoproteção , Feminino , Proteínas de Homeodomínio/metabolismo , Cadeias lambda de Imunoglobulina/metabolismo , Integrina alfa2/metabolismo , Contagem de Linfócitos , Camundongos , Células Precursoras de Linfócitos B/metabolismo , Antígenos Thy-1/metabolismo
9.
Eur J Immunol ; 46(12): 2835-2841, 2016 12.
Artigo em Inglês | MEDLINE | ID: mdl-27618761

RESUMO

Growing cancers are known to modify immune responses through suppressive mechanisms manifested within the local tumor microenvironment. Accumulating evidence indicates that secreted tumor products can also influence on distant immunological compartments, including myelopoiesis in the bone marrow. However, it is unknown if a similar effect can occur to regulate B-cell lymphopoiesis in breast cancer. Examining the MMTV-PyMT murine model of breast cancer, we show a complete block in bone marrow B-cell lymphopoiesis, which is dependent on tumor burden. We also observed an increase in the total number of splenic B cells and an elevated frequency of marginal zone B cells. By using in vitro assays of B-cell lymphopoiesis, we show that tumor-secreted molecules directly inhibit B-cell progenitor proliferation and favor maturation. These data demonstrate a profound sensitivity of B-cell lymphopoiesis to the accumulation of ectopically produced molecules during tumor growth in PyMT.


Assuntos
Linfócitos B/fisiologia , Células da Medula Óssea/fisiologia , Neoplasias da Mama/imunologia , Evasão Tumoral , Microambiente Tumoral , Animais , Antígenos Virais de Tumores/genética , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Humanos , Ativação Linfocitária , Linfopoese , Vírus do Tumor Mamário do Camundongo/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Carga Tumoral
10.
Blood ; 126(10): 1184-92, 2015 Sep 03.
Artigo em Inglês | MEDLINE | ID: mdl-26170030

RESUMO

Systemic inflammation perturbs the bone marrow environment by evicting resident B cells and favoring granulopoiesis over lymphopoiesis. Despite these conditions, a subset of marrow B cell remains to become activated and produce potent acute immunoglobulin M (IgM) responses. This discrepancy is currently unresolved and a complete characterization of early perturbations in the B-cell niche has not been undertaken. Here, we show that within a few hours of challenging mice with adjuvant or cecal puncture, B cells accumulate in the bone marrow redistributed away from sinusoid vessels. This response correlates with enhanced sensitivity to CXC chemokine ligand 12 (CXCL12) but not CXCL13 or CC chemokine ligand 21. Concurrently, a number of B-cell survival and differentiation factors are elevated to produce a transiently supportive milieu. Disrupting homing dynamics with a CXC chemokine receptor 4 inhibitor reduced the formation of IgM-secreting cells. These data highlight the rapidity with which peripheral inflammation modifies the marrow compartment, and demonstrate that such modifications regulate acute IgM production within this organ. Furthermore, our study indicates that conversion to a state of emergency granulopoiesis is temporally delayed, allowing B cells opportunity to respond to antigen.


Assuntos
Linfócitos B/imunologia , Células da Medula Óssea/imunologia , Inflamação/imunologia , Ativação Linfocitária/imunologia , Linfopoese/imunologia , Transferência Adotiva , Animais , Medula Óssea/imunologia , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
12.
Cancer Immunol Res ; 2(11): 1113-24, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25154710

RESUMO

Inciting the cellular arm of adaptive immunity has been the fundamental goal of cancer immunotherapy strategies, specifically focusing on inducing tumor antigen-specific responses by CD8(+) cytotoxic T lymphocytes (CTL). However, there is an emerging appreciation that the cytotoxic function of CD4(+) T cells can be effective in a clinical setting. Harnessing this potential will require an understanding of how such cells arise. In this study, we use an IL12-transduced variant of the 70Z/3 leukemia cell line in a B6D2F1 (BDF1) murine model system to reveal a novel cascade of cells and soluble factors that activate anticancer CD4(+) killer cells. We show that natural killer T cells play a pivotal role by activating dendritic cells in a contact-dependent manner; soluble products of this interaction, including MCP-1, propagate the activation signal, culminating in the development of CD4(+) CTLs that directly mediate an antileukemia response while also orchestrating a multipronged attack by other effector cells. A more complete picture of the conditions that induce such a robust response will allow us to capitalize on CD4(+) T-cell plasticity for maximum therapeutic effect.


Assuntos
Linfócitos T CD4-Positivos/imunologia , Comunicação Celular/imunologia , Imunidade Inata , Imunoterapia/métodos , Leucemia/imunologia , Ativação Linfocitária/imunologia , Animais , Linhagem Celular Tumoral , Técnicas de Cocultura , Células Dendríticas/imunologia , Feminino , Citometria de Fluxo , Imunidade Inata/imunologia , Camundongos , Células T Matadoras Naturais/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Baço/citologia , Baço/imunologia , Linfócitos T Citotóxicos/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA