Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
J Immunol ; 197(2): 644-54, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-27271569

RESUMO

CD8(+) T cells develop increased sensitivity following Ag experience, and differences in sensitivity exist between T cell memory subsets. How differential TCR signaling between memory subsets contributes to sensitivity differences is unclear. We show in mouse effector memory T cells (TEM) that >50% of lymphocyte-specific protein tyrosine kinase (Lck) exists in a constitutively active conformation, compared with <20% in central memory T cells (TCM). Immediately proximal to Lck signaling, we observed enhanced Zap-70 phosphorylation in TEM following TCR ligation compared with TCM Furthermore, we observed superior cytotoxic effector function in TEM compared with TCM, and we provide evidence that this results from a lower probability of TCM reaching threshold signaling owing to the decreased magnitude of TCR-proximal signaling. We provide evidence that the differences in Lck constitutive activity between CD8(+) TCM and TEM are due to differential regulation by SH2 domain-containing phosphatase-1 (Shp-1) and C-terminal Src kinase, and we use modeling of early TCR signaling to reveal the significance of these differences. We show that inhibition of Shp-1 results in increased constitutive Lck activity in TCM to levels similar to TEM, as well as increased cytotoxic effector function in TCM Collectively, this work demonstrates a role for constitutive Lck activity in controlling Ag sensitivity, and it suggests that differential activities of TCR-proximal signaling components may contribute to establishing the divergent effector properties of TCM and TEM. This work also identifies Shp-1 as a potential target to improve the cytotoxic effector functions of TCM for adoptive cell therapy applications.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Memória Imunológica/fisiologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/imunologia , Subpopulações de Linfócitos T/imunologia , Animais , Western Blotting , Linfócitos T CD8-Positivos/metabolismo , Citotoxicidade Imunológica/imunologia , Citometria de Fluxo , Humanos , Ativação Linfocitária/imunologia , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/metabolismo , Cadeias de Markov , Camundongos , Reação em Cadeia da Polimerase , Subpopulações de Linfócitos T/metabolismo , Imagem com Lapso de Tempo
2.
Gastroenterology ; 143(4): 1061-72, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22705178

RESUMO

BACKGROUND & AIMS: Immune cells of the liver must be able to recognize and react to pathogens yet remain tolerant to food molecules and other nonpathogens. Dendritic cells (DCs) are believed to contribute to hepatic tolerance. Lipids have been implicated in dysfunction of DCs in cancer. Therefore, we investigated whether high lipid content in liver DCs affects induction of tolerance. METHODS: Mouse and human hepatic nonparenchymal cells were isolated by mechanical and enzymatic digestion. DCs were purified by fluorescence-activated cell sorting or with immunomagnetic beads. DC lipid content was assessed by flow cytometry, immune fluorescence, and electron microscopy and by measuring intracellular component lipids. DC activation was determined from surface phenotype and cytokine profile. DC function was assessed in T-cell, natural killer (NK) cell, and NKT cell coculture assays as well as in vivo. RESULTS: We observed 2 distinct populations of hepatic DCs in mice and humans based on their lipid content and expression of markers associated with adipogenesis and lipid metabolism. This lipid-based dichotomy in DCs was unique to the liver and specific to DCs compared with other hepatic immune cells. However, rather than mediate tolerance, the liver DC population with high concentrations of lipid was immunogenic in multiple models; they activated T cells, NK cells, and NKT cells. Conversely, liver DCs with low levels of lipid induced regulatory T cells, anergy to cancer, and oral tolerance. The immunogenicity of lipid-rich liver DCs required their secretion of tumor necrosis factor α and was directly related to their high lipid content; blocking DC synthesis of fatty acids or inhibiting adipogenesis (by reducing endoplasmic reticular stress) reduced DC immunogenicity. CONCLUSIONS: Human and mouse hepatic DCs are composed of distinct populations that contain different concentrations of lipid, which regulates immunogenic versus tolerogenic responses in the liver.


Assuntos
Antígenos CD/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Lipídeos/análise , Fígado/imunologia , Fígado/metabolismo , Adipogenia , Animais , Antígenos CD1d/metabolismo , Apoptose , Antígeno B7-1/metabolismo , Antígeno B7-2/metabolismo , Antígeno CD11b/metabolismo , Antígenos CD40/metabolismo , Células Cultivadas , Células Dendríticas/química , Humanos , Tolerância Imunológica , Molécula 1 de Adesão Intercelular/metabolismo , Células Matadoras Naturais/fisiologia , Antígenos Comuns de Leucócito/metabolismo , Metabolismo dos Lipídeos , Fígado/química , Ativação Linfocitária , Camundongos , Células T Matadoras Naturais/fisiologia , Fenótipo , Linfócitos T/fisiologia , Linfócitos T Reguladores/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
3.
Gastroenterology ; 141(5): 1915-26.e1-14, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21801698

RESUMO

BACKGROUND & AIMS: The cellular mediators of acute pancreatitis are incompletely understood. Dendritic cells (DCs) can promote or suppress inflammation, depending on their subtype and context. We investigated the roles of DC in development of acute pancreatitis. METHODS: Acute pancreatitis was induced in CD11c.DTR mice using caerulein or L-arginine; DCs were depleted by administration of diphtheria toxin. Survival was analyzed using Kaplan-Meier method. RESULTS: Numbers of major histocompatibility complex II(+)CD11c(+) DCs increased 100-fold in pancreata of mice with acute pancreatitis to account for nearly 15% of intrapancreatic leukocytes. Intrapancreatic DCs acquired a distinct immune phenotype in mice with acute pancreatitis; they expressed higher levels of major histocompatibility complex II and CD86 and increased production of interleukin-6, membrane cofactor protein-1, and tumor necrosis factor-α. However, rather than inducing an organ-destructive inflammatory process, DCs were required for pancreatic viability; the exocrine pancreas died in mice that were depleted of DCs and challenged with caerulein or L-arginine. All mice with pancreatitis that were depleted of DCs died from acinar cell death within 4 days. Depletion of DCs from mice with pancreatitis resulted in neutrophil infiltration and increased levels of systemic markers of inflammation. However, the organ necrosis associated with depletion of DCs did not require infiltrating neutrophils, activation of nuclear factor-κB, or signaling by mitogen-activated protein kinase or tumor necrosis factor-α. CONCLUSIONS: DCs are required for pancreatic viability in mice with acute pancreatitis and might protect organs against cell stress.


Assuntos
Células Dendríticas/fisiologia , Pâncreas/patologia , Pâncreas/fisiopatologia , Pancreatite/patologia , Pancreatite/fisiopatologia , Sobrevivência de Tecidos/fisiologia , Doença Aguda , Animais , Arginina/efeitos adversos , Ceruletídeo/efeitos adversos , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/metabolismo , Células Dendríticas/patologia , Toxina Diftérica/farmacologia , Modelos Animais de Doenças , Proteína 1 de Resposta de Crescimento Precoce/metabolismo , Interleucina-6/metabolismo , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pancreatite/induzido quimicamente , Fenótipo , Fatores de Tempo
4.
Immunol Invest ; 41(6-7): 595-613, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23017137

RESUMO

Myeloid-Derived Suppressor Cells (MDSC) are immature myeloid cells that are potent inhibitors of immune cell function and which accumulate under conditions of inflammation, especially cancer. MDSC are suggested to promote the growth of cancer by both enhancement of tumor angiogenesis and metastasis and also inhibition of antitumor immune responses. The presence of deficient and/or defective antitumor adaptive and innate immune responses, coincident with accumulation of MDSC in lymphoid organs and tumor parenchyma, supports the notion of a causal relationship. The potent ability of MDSC to inhibit several components and phases of immune response highlights the likelihood that targeting the inhibitory functions of MDSC may maximize the therapeutic potential of antitumor immunotherapy. In order to guide the rational development of immunotherapeutic strategies that incorporate inhibition of MDSC activity and enzymatic functions, thorough understanding of the role of MDSC in antitumor immune responses is required. In this manuscript we review the multifaceted inhibitory functions of MDSC and consider the role of MDSC-induced inhibition of antitumor T cell effector phase. Support for this research is from NIH R01 CA108573.


Assuntos
Células Progenitoras Mieloides/efeitos dos fármacos , Neoplasias/patologia , Neoplasias/terapia , Linfócitos T/efeitos dos fármacos , Aminoácidos/metabolismo , Animais , Anticorpos Monoclonais/farmacologia , Anticorpos Monoclonais/uso terapêutico , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Comunicação Celular/efeitos dos fármacos , Comunicação Celular/genética , Comunicação Celular/imunologia , Humanos , Tolerância Imunológica , Imunoterapia , Camundongos , Células Progenitoras Mieloides/imunologia , Células Progenitoras Mieloides/patologia , Neoplasias/imunologia , Neoplasias/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Linfócitos T/imunologia , Linfócitos T/patologia , Evasão Tumoral
5.
J Immunol ; 185(12): 7133-40, 2010 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-21127315

RESUMO

The presence in cancer tissue of Ag-specific, activated tumor infiltrating CD8(+) T cells proves that tumors express Ags capable of eliciting immune response. Therefore, in general, tumor escape from immune-mediated clearance is not attributable to immunological ignorance. However, tumor-infiltrating lymphocytes are defective in effector phase function, demonstrating tumor-induced immune suppression that likely underlies tumor escape. Since exocytosis of lytic granules is dependent upon TCR-mediated signal transduction, it is a reasonable contention that tumors may induce defective signal transduction in tumor infiltrating T cells. In this review, we consider the biochemical basis for antitumor T cell dysfunction, focusing on the role of inhibitory signaling receptors in restricting TCR-mediated signaling in tumor-infiltrating lymphocytes.


Assuntos
Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Neoplasias/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Transdução de Sinais/imunologia , Evasão Tumoral/imunologia , Animais , Exocitose/imunologia , Humanos
6.
J Immunol ; 185(4): 2200-8, 2010 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-20639479

RESUMO

The normal liver is characterized by immunologic tolerance. Primary mediators of hepatic immune tolerance are liver sinusoidal endothelial cells (LSECs). LSECs block adaptive immunogenic responses to Ag and induce the generation of T regulatory cells. Hepatic fibrosis is characterized by both intense intrahepatic inflammation and altered hepatic immunity. We postulated that, in liver fibrosis, a reversal of LSEC function from tolerogenic to proinflammatory and immunogenic may contribute to both the heightened inflammatory milieu and altered intrahepatic immunity. We found that, after fibrotic liver injury from hepatotoxins, LSECs become highly proinflammatory and secrete an array of cytokines and chemokines. In addition, LSECs gain enhanced capacity to capture Ag and induce T cell proliferation. Similarly, unlike LSECs in normal livers, in fibrosis, LSECs do not veto dendritic cell priming of T cells. Furthermore, whereas in normal livers, LSECs are active in the generation of T regulatory cells, in hepatic fibrosis LSECs induce an immunogenic T cell phenotype capable of enhancing endogenous CTLs and generating potent de novo CTL responses. Moreover, depletion of LSECs from fibrotic liver cultures mitigates the proinflammatory milieu characteristic of hepatic fibrosis. Our findings offer a critical understanding of the role of LSECs in modulating intrahepatic immunity and inflammation in fibro-inflammatory liver disease.


Assuntos
Antígenos/imunologia , Células Endoteliais/imunologia , Cirrose Hepática/imunologia , Linfócitos T Reguladores/imunologia , Animais , Tetracloreto de Carbono , Proliferação de Células , Quimiocinas/metabolismo , Citocinas/metabolismo , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Células Endoteliais/metabolismo , Citometria de Fluxo , Mediadores da Inflamação/metabolismo , Fígado/imunologia , Fígado/patologia , Cirrose Hepática/induzido quimicamente , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Citotóxicos/imunologia , Linfócitos T Citotóxicos/metabolismo , Linfócitos T Reguladores/metabolismo , Tioacetamida
7.
Immunol Rev ; 222: 192-205, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18364003

RESUMO

The immune response to cancer has been long recognized, including both innate and adaptive responses, showing that the immune system can recognize protein products of genetic and epigenetic changes in transformed cells. The accumulation of antigen-specific T cells within the tumor, the draining lymph node, and the circulation, either in newly diagnosed patients or resultant from experimental immunotherapy, proves that tumors produce antigens and that priming occurs. Unfortunately, just as obviously, tumors grow, implying that anti-tumor immune responses are either not sufficiently vigorous to eliminate the cancer or that anti-tumor immunity is suppressed. Both possibilities are supported by current data. In experimental animal models of cancer and also in patients, systemic immunity is usually not dramatically suppressed, because tumor-bearing animals and patients develop T-cell-dependent immune responses to microbes and to either model antigens or experimental cancer vaccines. However, inhibition of specific anti-tumor immunity is common, and several possible explanations of tolerance to tumor antigens or tumor-induced immunesuppression have been proposed. Inhibition of effective anti-tumor immunity results from the tumor or the host response to tumor growth, inhibiting the activation, differentiation, or function of anti-tumor immune cells. As a consequence, anti-tumor T cells cannot respond productively to developmental, targeting, or activation cues. While able to enhance the number and phenotype of anti-tumor T cells, the modest success of immunotherapy has shown the necessity to attempt to reverse tolerance in anti-tumor T cells, and the vanguard of experimental therapy now focuses on vaccination in combination with blockade of immunosuppressive mechanisms. This review discusses several potential mechanisms by which anti-tumor T cells may be inhibited in function.


Assuntos
Antígenos de Neoplasias/imunologia , Ativação Linfocitária , Neoplasias/imunologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Evasão Tumoral , Animais , Apresentação de Antígeno , Complexo CD3/imunologia , Linfócitos T CD8-Positivos/imunologia , Vacinas Anticâncer , Citotoxicidade Imunológica , Epitopos , Humanos , Tolerância Imunológica , Imunoterapia , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Camundongos , Células Mieloides/imunologia , Neoplasias/terapia , Linfócitos T/patologia , Linfócitos T Reguladores/imunologia
8.
Nat Commun ; 13(1): 6418, 2022 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-36302761

RESUMO

A paucity of effector T cells within tumors renders pancreatic ductal adenocarcinoma (PDAC) resistant to immune checkpoint therapies. While several under-development approaches target immune-suppressive cells in the tumor microenvironment, there is less focus on improving T cell function. Here we show that inhibiting vasoactive intestinal peptide receptor (VIP-R) signaling enhances anti-tumor immunity in murine PDAC models. In silico data mining and immunohistochemistry analysis of primary tumors indicate overexpression of the neuropeptide vasoactive intestinal peptide (VIP) in human PDAC tumors. Elevated VIP levels are also present in PDAC patient plasma and supernatants of cultured PDAC cells. Furthermore, T cells up-regulate VIP receptors after activation, identifying the VIP signaling pathway as a potential target to enhance T cell function. In mouse PDAC models, VIP-R antagonist peptides synergize with anti-PD-1 antibody treatment in improving T cell recruitment into the tumors, activation of tumor-antigen-specific T cells, and inhibition of T cell exhaustion. In contrast to the limited single-agent activity of anti-PD1 antibodies or VIP-R antagonist peptides, combining both therapies eliminate tumors in up to 40% of animals. Furthermore, tumor-free mice resist tumor re-challenge, indicating anti-cancer immunological memory generation. VIP-R signaling thus represents a tumor-protective immune-modulatory pathway that is targetable in PDAC.


Assuntos
Carcinoma Ductal Pancreático , Neoplasias Pancreáticas , Humanos , Camundongos , Animais , Peptídeo Intestinal Vasoativo/farmacologia , Carcinoma Ductal Pancreático/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Receptores de Peptídeo Intestinal Vasoativo , Transdução de Sinais , Microambiente Tumoral , Neoplasias Pancreáticas
9.
J Clin Invest ; 116(10): 2587-90, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17016554

RESUMO

Inflammation resultant from tumor growth, infection with certain pathogens, or in some cases, trauma, can result in systemic release of cytokines, especially GM-CSF, that in turn stimulate the abundant production and activation of a population of immature myeloid cells, termed myeloid suppressor cells (MSCs), that have potent immunosuppressive functions. In this issue of the JCI, Gallina and colleagues have illuminated some complex issues concerning the development, activation, and function of MSCs (see the related article beginning on page 2777). They show that activation of MSCs is initiated in response to IFN-gamma, presumably produced in situ by antitumor T cells in the tumor microenvironment. After this triggering event, MSCs express 2 enzymes involved in l-arginine metabolism, Arginase I and iNOS, whose metabolic products include diffusible and highly reactive peroxynitrites, the ultimate biochemical mediators of T cell immune suppression. The multifaceted regulation of this complex suppressive effector system provides several potential therapeutic targets.


Assuntos
Células Mieloides/imunologia , Neoplasias/imunologia , Animais , Arginase/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Imunidade Celular/imunologia , Interferon gama/metabolismo , Interleucina-13/metabolismo , Camundongos , Modelos Imunológicos , Neoplasias/metabolismo , Óxido Nítrico Sintase Tipo II/metabolismo , Receptores de Superfície Celular/metabolismo , Linfócitos T/imunologia , Linfócitos T/metabolismo
10.
J Leukoc Biol ; 84(3): 679-88, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18566103

RESUMO

Mononuclear phagocytes (MPCs) at the tumor site can be divided into subclasses, including monocyte-lineage myeloid-derived suppressor cells (MDSCs) and the immunosuppressive tumor-infiltrating macrophages (TIMs). Cancer growth coincides with the expansion of MDSCs found in the blood, secondary lymphoid organs, and tumor tissue. These MDSCs are thought to mature into macrophages and to promote tumor development by a combination of growth-enhancing properties and suppression of local antitumor immunoresponses. As little is known about either subset of MPCs, we investigated MPCs infiltrating into murine adenocarcinoma MCA38 tumors. We found that these MPCs displayed immunosuppressive characteristics and a MDSC cell-surface phenotype. Over 70% of the MPCs were mature (F4/80(+)Ly6C(-)) macrophages, and the rest were immature (F480(+) Ly6C(+)) monocytes. MPC maturation was inhibited when the cells infiltrated a tumor variant expressing IL-2 and soluble TNF type II receptor (sTNFRII). In addition, the IL-2/sTNFRII MCA38 tumor microenvironment altered the MPC phenotype; these cells did not survive culturing in vitro as a result of Fas-mediated apoptosis and negligible M-CSFR expression. Furthermore, CD4(+) tumor-infiltrating lymphocytes (TILs) in wild-type tumors robustly expressed IL-13, IFN-gamma, and GM-CSF, and CD4(+) TILs in IL-2/sTNFRII-expressing tumors expressed little IL-13. These data suggest that immunotherapy-altered Th cell balance in the tumor microenvironment can affect the differentiation and maturation of MPCs in vivo. Furthermore, as neither the designation MDSC nor TIM can sufficiently describe the status of monocytes/macrophages in this tumor microenvironment, we believe these cells are best designated as MPCs.


Assuntos
Adenocarcinoma/imunologia , Linfócitos do Interstício Tumoral/imunologia , Macrófagos/imunologia , Células Mieloides/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Animais , Apoptose/imunologia , Linfócitos T CD4-Positivos/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Neoplasias do Colo/imunologia , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Proteína Ligante Fas/fisiologia , Citometria de Fluxo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/imunologia , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Immunoblotting , Técnicas Imunoenzimáticas , Imunofenotipagem , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-13/imunologia , Interleucina-13/metabolismo , Interleucina-2/imunologia , Interleucina-2/metabolismo , Linfócitos do Interstício Tumoral/patologia , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Monócitos/patologia , Células Mieloides/metabolismo , Células Mieloides/patologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor de Fator Estimulador de Colônias de Macrófagos/genética , Receptor de Fator Estimulador de Colônias de Macrófagos/metabolismo , Receptores Tipo I de Fatores de Necrose Tumoral/fisiologia , Receptores Tipo II do Fator de Necrose Tumoral/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Taxa de Sobrevida , Linfócitos T Auxiliares-Indutores/metabolismo , Linfócitos T Auxiliares-Indutores/patologia , Linfócitos T Reguladores/imunologia , Células Th1/imunologia
11.
Mol Ther Oncolytics ; 8: 71-81, 2018 Mar 30.
Artigo em Inglês | MEDLINE | ID: mdl-29888320

RESUMO

Through the action of two virus-encoded decapping enzymes (D9 and D10) that remove protective caps from mRNA 5'-termini, Vaccinia virus (VACV) accelerates mRNA decay and limits activation of host defenses. D9- or D10-deficient VACV are markedly attenuated in mice and fail to counter cellular double-stranded RNA-responsive innate immune effectors, including PKR. Here, we capitalize upon this phenotype and demonstrate that VACV deficient in either decapping enzyme are effective oncolytic viruses. Significantly, D9- or D10-deficient VACV displayed anti-tumor activity against syngeneic mouse tumors of different genetic backgrounds and human hepatocellular carcinoma xenografts. Furthermore, D9- and D10-deficient VACV hyperactivated the host anti-viral enzyme PKR in non-tumorigenic cells compared to wild-type virus. This establishes a new genetic platform for oncolytic VACV development that is deficient for a major pathogenesis determinant while retaining viral genes that support robust productive replication like those required for nucleotide metabolism. It further demonstrates how VACV mutants unable to execute a fundamental step in virus-induced mRNA decay can be unexpectedly translated into a powerful anti-tumor therapy.

12.
Int J Oncol ; 30(4): 803-11, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17332918

RESUMO

It is well known that tumor necrosis factor (TNF) can have both contrary and pleiotropic effects in anti-tumor immune response. In the present study, we prepared two different tumor cell-based immunotherapy models: MCA38 adenocarcinoma and GL261 glioma intracranial interleukin-2 (IL-2)-based. Each tumor was transfected to express IL-2 with or without expression of the soluble form of tumor necrosis factor receptor type II (sTNFRII). Although mice in which TNF is blocked survive longer than IL-2 alone (35.2 versus 26 days), the reverse was observed for GL261 glioma. The differential effect on tumor growth implies enhanced TNF sensitivity of GL261 compared to MCA38. This notion is supported by the observation that TNF induces apoptosis in GL261 but not MCA38 tumors. We further examined tumor infiltrating CD11b+F4/80+ macrophages (or tumor-associated macrophages: TAM) for TNF production in vivo and found that TAM express cell surface TNF implying a role in eliminating glioma cells mediated by the cell surface form of TNF.


Assuntos
Neoplasias Encefálicas/terapia , Glioma/terapia , Imunoterapia Adotiva , Macrófagos/imunologia , Macrófagos/transplante , Fator de Necrose Tumoral alfa/metabolismo , Adenocarcinoma/terapia , Animais , Antígeno CD11b/análise , Linhagem Celular Tumoral , Interleucina-2/metabolismo , Macrófagos/química , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Microglia/imunologia
13.
Crit Rev Immunol ; 26(3): 265-90, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16928189

RESUMO

T-cell receptor (TCR) engagement by antigen results in proliferation, differentiation and cytokine secretion. In the CD8+ T-cell subset, TCR triggering also induces granule exocytosis, the directional release of contents of lysosome-like granules toward the target cell presenting the antigen. This process is responsible for immediate death of target cells. The intracellular events required for granule exocytosis are distinct from those of proliferation and cytokine secretion, as the former do not require de novo protein synthesis. Consequently, the key TCR signaling events required for granule exocytosis may be distinct. In this article, we review present knowledge of regulation of granule exocytosis by molecules of the TCR signaling cascade.


Assuntos
Exocitose , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T Citotóxicos/imunologia , Animais , Citotoxicidade Imunológica , Humanos , Linfo-Histiocitose Hemofagocítica/imunologia , Camundongos , Vesículas Secretórias/metabolismo
14.
J Leukoc Biol ; 79(4): 652-62, 2006 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-16415165

RESUMO

Growth of cancer in rodent models and in patients elicits immune responses directed toward various antigens expressed by the transformed cell. Clearly though, as most tumors grow, unmanipulated antitumor immune responses are incapable of eliminating cancer. Over the past approximately 15 years, antitumor immunoglobulin and T cells have been used to identify tumor antigens, which in turn, have served as the basis for therapeutic vaccine trials. However, experimental cancer vaccines, although in some patients result in elimination of large tumor burdens, have a low frequency of long-term cancer remission in most patients, ca. <5%. Therefore, as tumors express antigens that distinguish themselves from nontransformed cells in immunological terms (i.e., elicit immune responses to growth of primary tumor and can target tumor cells in vivo), and tumor vaccines prime unsuccessful antitumor immune responses in patients, it is likely that growth of cancer induces immune tolerance to tumor cells. Although there are several types of T cell tolerance, mature, antigen-specific CD8+ T cells isolated from tumors are lytic-defective, implying that the tumor microenvironment inactivates the antitumor effector phase. The nature of the functional local tolerance to antitumor immune response is the subject of this review.


Assuntos
Antineoplásicos/imunologia , Tolerância Imunológica , Neoplasias/imunologia , Evasão Tumoral/imunologia , Animais , Antineoplásicos/uso terapêutico , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/uso terapêutico , Humanos , Neoplasias/tratamento farmacológico , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Carga Tumoral/imunologia
15.
Cancer Immunol Res ; 5(10): 920-928, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28874354

RESUMO

Cancers are infiltrated with antitumor CD8+ T cells that arise during tumor growth, but are defective in effector phase functions because of the suppressive microenvironment. The reactivation of TILs can result in tumor destruction, showing that lytic dysfunction in CD8+ tumor-infiltrating lymphocytes (TIL) permits tumor growth. Like all memory T cells, TILs express inhibitory signaling receptors (aka checkpoint inhibitor molecules) that downregulate TCR-mediated signal transduction upon TIL interaction with cells expressing cognate ligands, thereby restricting cell activation and preventing the effector phase. Previously, we identified a novel murine CD8+ TIL inhibitory signaling receptor, protocadherin-18, and showed that it interacts with p56lck kinase to abrogate proximal TCR signaling. Here, we show that TILs from mice deleted in protocadherin-18 had enhanced antitumor activity and that coblockade of PD-1 and protocadherin-18 in wild-type mice significantly enhanced TIL effector phase function. These results define an important role for protocadherin-18 in antitumor T-cell activity. Cancer Immunol Res; 5(10); 920-8. ©2017 AACR.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Caderinas/metabolismo , Imunomodulação , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/metabolismo , Transdução de Sinais , Animais , Antineoplásicos Imunológicos/farmacologia , Caderinas/antagonistas & inibidores , Caderinas/genética , Citotoxicidade Imunológica , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Memória Imunológica/efeitos dos fármacos , Interferon gama/metabolismo , Ligantes , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Knockout , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Ensaios Antitumorais Modelo de Xenoenxerto
16.
Cancer Res ; 62(4): 1110-5, 2002 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-11861390

RESUMO

Membrane-bound complement inhibitors protect host cells from inadvertent complement attack, and complement inhibitors are often up-regulated on tumors, possibly representing a selective adaptation by tumors to escape elimination by a host antitumor immune response. Relevant in vivo studies using rodent models of human cancer have been hampered by the fact that human complement inhibitors are not effective against rodent complement. Using nude rats and MCF7 cells expressing different rat complement inhibitors, a model of human breast cancer was established to investigate the role of complement and complement inhibitors in tumor progression. Expression of rat CD59, an inhibitor of the terminal cytolytic membrane attack complex of complement, had no effect on the incidence or growth rate of MCF7 tumors. In contrast, expression of rat Crry, an inhibitor of complement activation, dramatically enhanced the tumorigenicity of MCF7 cells. The expression of rat Crry on MCF7 inhibited the in vivo deposition of complement C3 fragments that serve as opsonins for receptors on phagocytes and natural killer cells. These data provide direct in vivo evidence that an inhibitor of complement activation can facilitate tumor growth by modulating C3 deposition. These data indicate an important role for complement opsonization in promoting cell-mediated antitumor immune function, a conclusion further supported by the demonstration that expression of rat Crry, but not rat CD59, on MCF7 cells inhibits rat cell-mediated cytotoxicity in vitro. Rat complement activation on MCF7 tumors was mediated by tumor-reactive antibodies present in the serum of naïve nude rats, but there was also an IgM response to MCF7 tumors, a situation with similarities to some human cancers. These data support a hypothesis that blocking complement inhibitor function on tumor cells will not only enhance monoclonal antibody-mediated immunotherapy but may also be effective at enhancing a normally ineffective humoral immune response in the absence of administered antitumor antibody.


Assuntos
Neoplasias da Mama/patologia , Proteínas Inativadoras do Complemento/fisiologia , Proteínas do Sistema Complemento/metabolismo , Neoplasias Mamárias Experimentais/patologia , Animais , Antígenos de Superfície , Neoplasias da Mama/imunologia , Neoplasias da Mama/metabolismo , Antígenos CD59/biossíntese , Antígenos CD59/genética , Antígenos CD59/fisiologia , Divisão Celular/fisiologia , Ativação do Complemento/fisiologia , Proteínas Inativadoras do Complemento/biossíntese , Proteínas do Sistema Complemento/fisiologia , Modelos Animais de Doenças , Feminino , Humanos , Neoplasias Mamárias Experimentais/imunologia , Neoplasias Mamárias Experimentais/metabolismo , Ratos , Receptores de Superfície Celular , Receptores de Complemento/biossíntese , Receptores de Complemento/genética , Receptores de Complemento/fisiologia , Transfecção
17.
EBioMedicine ; 5: 59-67, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27077112

RESUMO

Although counteracting innate defenses allows oncolytic viruses (OVs) to better replicate and spread within tumors, CD8(+) T-cells restrict their capacity to trigger systemic anti-tumor immune responses. Herpes simplex virus-1 (HSV-1) evades CD8(+) T-cells by producing ICP47, which limits immune recognition of infected cells by inhibiting the transporter associated with antigen processing (TAP). Surprisingly, removing ICP47 was assumed to benefit OV immuno-therapy, but the impact of inhibiting TAP remains unknown because human HSV-1 ICP47 is not effective in rodents. Here, we engineer an HSV-1 OV to produce bovine herpesvirus UL49.5, which unlike ICP47, antagonizes rodent and human TAP. Significantly, UL49.5-expressing OVs showed superior efficacy treating bladder and breast cancer in murine models that was dependent upon CD8(+) T-cells. Besides injected subcutaneous tumors, UL49.5-OV reduced untreated, contralateral tumor size and metastases. These findings establish TAP inhibitor-armed OVs that evade CD8(+) T-cells as an immunotherapy strategy to elicit potent local and systemic anti-tumor responses.


Assuntos
Neoplasias da Mama/imunologia , Terapia Viral Oncolítica , Vírus Oncolíticos/imunologia , Neoplasias da Bexiga Urinária/imunologia , Proteínas do Envelope Viral/imunologia , Animais , Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/imunologia , Bovinos , Linhagem Celular Tumoral , Modelos Animais de Doenças , Herpesvirus Humano 1/imunologia , Humanos , Proteínas Imediatamente Precoces/genética , Proteínas Imediatamente Precoces/imunologia , Evasão da Resposta Imune/genética , Camundongos , Vírus Oncolíticos/genética , Neoplasias da Bexiga Urinária/terapia , Proteínas do Envelope Viral/genética , Proteínas do Envelope Viral/uso terapêutico
18.
J Diabetes Res ; 2016: 9083103, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27069933

RESUMO

Type 1 diabetes is an autoimmune disease in which insulin-producing pancreatic islet ß cells are the target of self-reactive B and T cells. T cells reactive with epitopes derived from insulin and/or IGRP are critical for the initiation and maintenance of disease, but T cells reactive with other islet antigens likely have an essential role in disease progression. We sought to identify candidate CD8(+) T cell epitopes that are pathogenic in type 1 diabetes. Proteins that elicit autoantibodies in human type 1 diabetes were analyzed by predictive algorithms for candidate epitopes. Using several different tolerizing regimes using synthetic peptides, two new predicted tolerogenic CD8(+) T cell epitopes were identified in the murine homolog of the major human islet autoantigen zinc transporter ZnT8 (aa 158-166 and 282-290) and one in a non-ß cell protein, dopamine ß-hydroxylase (aa 233-241). Tolerizing vaccination of NOD mice with a cDNA plasmid expressing full-length proinsulin prevented diabetes, whereas plasmids encoding ZnT8 and DßH did not. However, tolerizing vaccination of NOD mice with the proinsulin plasmid in combination with plasmids expressing ZnT8 and DßH decreased insulitis and enhanced prevention of disease compared to vaccination with the plasmid encoding proinsulin alone.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Proteínas de Transporte de Cátions/imunologia , Diabetes Mellitus Tipo 1/terapia , Dopamina beta-Hidroxilase/imunologia , Epitopos de Linfócito T , Terapia Genética/métodos , Tolerância Imunológica , Células Secretoras de Insulina/imunologia , Proinsulina/imunologia , Vacinação , Animais , Autoanticorpos/imunologia , Proteínas de Transporte de Cátions/genética , Células Cultivadas , Diabetes Mellitus Tipo 1/genética , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/patologia , Modelos Animais de Doenças , Dopamina beta-Hidroxilase/genética , Feminino , Humanos , Células Secretoras de Insulina/patologia , Ativação Linfocitária , Camundongos Endogâmicos NOD , Proinsulina/genética , Fatores de Tempo , Vacinas de Subunidades Antigênicas/genética , Vacinas de Subunidades Antigênicas/imunologia , Vacinas Sintéticas/genética , Vacinas Sintéticas/imunologia , Transportador 8 de Zinco
19.
Nat Commun ; 7: 12150, 2016 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-27381735

RESUMO

Myeloid-derived suppressor cells (MDSCs) have emerged as major regulators of immune responses in cancer and other pathological conditions. In recent years, ample evidence supports key contributions of MDSC to tumour progression through both immune-mediated mechanisms and those not directly associated with immune suppression. MDSC are the subject of intensive research with >500 papers published in 2015 alone. However, the phenotypic, morphological and functional heterogeneity of these cells generates confusion in investigation and analysis of their roles in inflammatory responses. The purpose of this communication is to suggest characterization standards in the burgeoning field of MDSC research.


Assuntos
Diferenciação Celular/imunologia , Células Supressoras Mieloides/imunologia , Neoplasias/imunologia , Terminologia como Assunto , Algoritmos , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Modelos Imunológicos , Células Supressoras Mieloides/citologia , Células Supressoras Mieloides/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia
20.
Vaccine ; 33(51): 7393-7400, 2015 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-26403368

RESUMO

The immune system recognizes protein antigens expressed in transformed cells evidenced by accumulation of antigen-specific T cells in tumor and tumor draining lymph nodes. However, despite demonstrable immune response, cancers grow progressively suggesting that priming of antitumor immunity is insufficiently vigorous or that antitumor immunity is suppressed, or both. Compared to virus infection, antitumor T cells are low abundance that likely contributes to tumor escape and enhancement of priming is a long-sought goal of experimental vaccination therapy. Furthermore, patient treatment with antigen-specific T cells can in some cases overcome deficient priming and cause tumor regression supporting the notion that low numbers of T cells permits tumor outgrowth. However, tumor-induced suppression of antitumor immune response is now recognized as a significant factor contributing to cancer growth and reversal of the inhibitory influences within the tumor microenvironment is a major research objective. Multiple cell types and factors can inhibit T cell functions in tumors and may be grouped in two general classes: T cell intrinsic and T cell extrinsic. T cell intrinsic factors are exemplified by T cell expression of cell surface inhibitory signaling receptors that, after contact with cells expressing a cognate ligand, inactivate proximal T Cell Receptor-mediated signal transduction therein rendering T cells dysfunctional. T cell extrinsic factors are more diverse in nature and are produced by tumors and various non-tumor cells in the tumor microenvironment. These include proteins secreted by tumor or stromal cells, highly reactive soluble oxygen and nitrogen species, cytokines, chemokines, gangliosides, and toxic metabolites. These factors may restrict T cell entrance into the tumor parenchyma, cause inactivation of effector phase T cell functions, or induce T cell apoptosis ultimately causing diminished cancer elimination. Here, we review the contributions of inhibitory factors to tumor T cell dysfunction leading to tumor escape.


Assuntos
Tolerância Imunológica , Neoplasias/imunologia , Neoplasias/patologia , Linfócitos T/imunologia , Microambiente Tumoral , Humanos , Evasão da Resposta Imune
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA