Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Nat Commun ; 13(1): 6418, 2022 10 27.
Article in English | MEDLINE | ID: mdl-36302761

ABSTRACT

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.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Humans , Mice , Animals , Vasoactive Intestinal Peptide/pharmacology , Carcinoma, Pancreatic Ductal/drug therapy , Pancreatic Neoplasms/drug therapy , Receptors, Vasoactive Intestinal Peptide , Signal Transduction , Tumor Microenvironment , Pancreatic Neoplasms
2.
Mol Ther Oncolytics ; 8: 71-81, 2018 Mar 30.
Article in English | MEDLINE | ID: mdl-29888320

ABSTRACT

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.

3.
Cancer Immunol Res ; 5(10): 920-928, 2017 10.
Article in English | MEDLINE | ID: mdl-28874354

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cadherins/metabolism , Immunomodulation , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Signal Transduction , Animals , Antineoplastic Agents, Immunological/pharmacology , Cadherins/antagonists & inhibitors , Cadherins/genetics , Cytotoxicity, Immunologic , Disease Models, Animal , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunologic Memory/drug effects , Interferon-gamma/metabolism , Ligands , Lymphocyte Activation/immunology , Male , Mice , Mice, Knockout , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Xenograft Model Antitumor Assays
4.
Nat Commun ; 7: 12150, 2016 07 06.
Article in English | MEDLINE | ID: mdl-27381735

ABSTRACT

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.


Subject(s)
Cell Differentiation/immunology , Myeloid-Derived Suppressor Cells/immunology , Neoplasms/immunology , Terminology as Topic , Algorithms , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Cytokines/immunology , Cytokines/metabolism , Humans , Models, Immunological , Myeloid-Derived Suppressor Cells/cytology , Myeloid-Derived Suppressor Cells/metabolism , Neoplasms/metabolism , Neoplasms/pathology
5.
J Immunol ; 197(2): 644-54, 2016 07 15.
Article in English | MEDLINE | ID: mdl-27271569

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , T-Lymphocyte Subsets/immunology , Animals , Blotting, Western , CD8-Positive T-Lymphocytes/metabolism , Cytotoxicity, Immunologic/immunology , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/metabolism , Markov Chains , Mice , Polymerase Chain Reaction , T-Lymphocyte Subsets/metabolism , Time-Lapse Imaging
6.
EBioMedicine ; 5: 59-67, 2016 Mar.
Article in English | MEDLINE | ID: mdl-27077112

ABSTRACT

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.


Subject(s)
Breast Neoplasms/immunology , Oncolytic Virotherapy , Oncolytic Viruses/immunology , Urinary Bladder Neoplasms/immunology , Viral Envelope Proteins/immunology , Animals , Breast Neoplasms/therapy , CD8-Positive T-Lymphocytes/immunology , Cattle , Cell Line, Tumor , Disease Models, Animal , Herpesvirus 1, Human/immunology , Humans , Immediate-Early Proteins/genetics , Immediate-Early Proteins/immunology , Immune Evasion/genetics , Mice , Oncolytic Viruses/genetics , Urinary Bladder Neoplasms/therapy , Viral Envelope Proteins/genetics , Viral Envelope Proteins/therapeutic use
7.
J Diabetes Res ; 2016: 9083103, 2016.
Article in English | MEDLINE | ID: mdl-27069933

ABSTRACT

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.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cation Transport Proteins/immunology , Diabetes Mellitus, Type 1/therapy , Dopamine beta-Hydroxylase/immunology , Epitopes, T-Lymphocyte , Genetic Therapy/methods , Immune Tolerance , Insulin-Secreting Cells/immunology , Proinsulin/immunology , Vaccination , Animals , Autoantibodies/immunology , Cation Transport Proteins/genetics , Cells, Cultured , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Disease Models, Animal , Dopamine beta-Hydroxylase/genetics , Female , Humans , Insulin-Secreting Cells/pathology , Lymphocyte Activation , Mice, Inbred NOD , Proinsulin/genetics , Time Factors , Vaccines, Subunit/genetics , Vaccines, Subunit/immunology , Vaccines, Synthetic/genetics , Vaccines, Synthetic/immunology , Zinc Transporter 8
8.
Vaccine ; 33(51): 7393-7400, 2015 Dec 16.
Article in English | MEDLINE | ID: mdl-26403368

ABSTRACT

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.


Subject(s)
Immune Tolerance , Neoplasms/immunology , Neoplasms/pathology , T-Lymphocytes/immunology , Tumor Microenvironment , Humans , Immune Evasion
9.
Immunol Invest ; 41(6-7): 595-613, 2012.
Article in English | MEDLINE | ID: mdl-23017137

ABSTRACT

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.


Subject(s)
Myeloid Progenitor Cells/drug effects , Neoplasms/pathology , Neoplasms/therapy , T-Lymphocytes/drug effects , Amino Acids/metabolism , Animals , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antigens, Neoplasm/genetics , Antigens, Neoplasm/immunology , Cell Communication/drug effects , Cell Communication/genetics , Cell Communication/immunology , Humans , Immune Tolerance , Immunotherapy , Mice , Myeloid Progenitor Cells/immunology , Myeloid Progenitor Cells/pathology , Neoplasms/immunology , Neoplasms/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Escape
10.
J Exp Med ; 209(10): 1723-42, S1, 2012 Sep 24.
Article in English | MEDLINE | ID: mdl-22966001

ABSTRACT

Foxp3 activity is essential for the normal function of the immune system. Two types of regulatory T (T reg) cells express Foxp3, thymus-generated natural T reg (nT reg) cells, and peripherally generated adaptive T reg (iT reg) cells. These cell types have complementary functions. Until now, it has not been possible to distinguish iT reg from nT reg cells in vivo based solely on surface markers. We report here that Neuropilin 1 (Nrp1) is expressed at high levels by most nT reg cells; in contrast, mucosa-generated iT reg and other noninflammatory iT reg cells express low levels of Nrp1. We found that Nrp1 expression is under the control of TGF-ß. By tracing nT reg and iT reg cells, we could establish that some tumors have a very large proportion of infiltrating iT reg cells. iT reg cells obtained from highly inflammatory environments, such as the spinal cords of mice with spontaneous autoimmune encephalomyelitis (EAE) and the lungs of mice with chronic asthma, express Nrp1. In the same animals, iT reg cells in secondary lymphoid organs remain Nrp1(low). We also determined that, in spontaneous EAE, iT reg cells help to establish a chronic phase of the disease.


Subject(s)
Mucous Membrane/immunology , Neuropilin-1/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Thymus Gland/immunology , Animals , Cell Lineage , Cell Membrane/metabolism , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/drug effects , Inflammation/immunology , Inflammation/metabolism , Intestinal Mucosa/metabolism , Intestines/immunology , Intestines/microbiology , Lymphocyte Activation/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Metagenome/immunology , Mice , Mice, Transgenic , Mucous Membrane/metabolism , Neuropilin-1/genetics , Thymus Gland/metabolism , Transforming Growth Factor beta/pharmacology
11.
J Exp Med ; 209(9): 1671-87, 2012 Aug 27.
Article in English | MEDLINE | ID: mdl-22908323

ABSTRACT

The transition of chronic pancreatic fibroinflammatory disease to neoplasia is a primary example of the paradigm linking inflammation to carcinogenesis. However, the cellular and molecular mediators bridging these entities are not well understood. Because TLR4 ligation can exacerbate pancreatic inflammation, we postulated that TLR4 activation drives pancreatic carcinogenesis. In this study, we show that lipopolysaccharide accelerates pancreatic tumorigenesis, whereas TLR4 inhibition is protective. Furthermore, blockade of the MyD88-independent TRIF pathway is protective against pancreatic cancer, whereas blockade of the MyD88-dependent pathway surprisingly exacerbates pancreatic inflammation and malignant progression. The protumorigenic and fibroinflammatory effects of MyD88 inhibition are mediated by dendritic cells (DCs), which induce pancreatic antigen-restricted Th2-deviated CD4(+) T cells and promote the transition from pancreatitis to carcinoma. Our data implicate a primary role for DCs in pancreatic carcinogenesis and illustrate divergent pathways in which blockade of TLR4 signaling via TRIF is protective against pancreatic cancer and, conversely, MyD88 inhibition exacerbates pancreatic inflammation and neoplastic transformation by augmenting the DC-Th2 axis.


Subject(s)
Adenocarcinoma/pathology , Dendritic Cells/metabolism , Myeloid Differentiation Factor 88/metabolism , Pancreatic Neoplasms/pathology , Th2 Cells/pathology , Toll-Like Receptor 4/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Adenocarcinoma/metabolism , Animals , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/pathology , Dendritic Cells/immunology , Humans , Male , Mice , Mice, Mutant Strains , Myeloid Differentiation Factor 88/genetics , Pancreatic Neoplasms/metabolism , Pancreatitis, Chronic/metabolism , Pancreatitis, Chronic/pathology , Th2 Cells/immunology
12.
Gastroenterology ; 143(4): 1061-72, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22705178

ABSTRACT

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.


Subject(s)
Antigens, CD/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Lipids/analysis , Liver/immunology , Liver/metabolism , Adipogenesis , Animals , Antigens, CD1d/metabolism , Apoptosis , B7-1 Antigen/metabolism , B7-2 Antigen/metabolism , CD11b Antigen/metabolism , CD40 Antigens/metabolism , Cells, Cultured , Dendritic Cells/chemistry , Humans , Immune Tolerance , Intercellular Adhesion Molecule-1/metabolism , Killer Cells, Natural/physiology , Leukocyte Common Antigens/metabolism , Lipid Metabolism , Liver/chemistry , Lymphocyte Activation , Mice , Natural Killer T-Cells/physiology , Phenotype , T-Lymphocytes/physiology , T-Lymphocytes, Regulatory/physiology , Tumor Necrosis Factor-alpha/metabolism
13.
PLoS One ; 7(5): e36101, 2012.
Article in English | MEDLINE | ID: mdl-22567129

ABSTRACT

CD8(+) tumor infiltrating T cells (TIL) lack effector-phase functions due to defective proximal TCR-mediated signaling previously shown to result from inactivation of p56(lck) kinase. We identify a novel interacting partner for p56(lck) in nonlytic TIL, Protocadherin-18 ('pcdh18'), and show that pcdh18 is transcribed upon in vitro or in vivo activation of all CD8(+) central memory T cells (CD44(+)CD62L(hi)CD127(+)) coincident with conversion into effector memory cells (CD44(+)CD62L(lo)CD127(+)). Expression of pcdh18 in primary CD8(+) effector cells induces the phenotype of nonlytic TIL: defective proximal TCR signaling, cytokine secretion, and cytolysis, and enhanced AICD. pcdh18 contains a motif (centered at Y842) shared with src kinases (QGQYQP) that is required for the inhibitory phenotype. Thus, pcdh18 is a novel activation marker of CD8(+) memory T cells that can function as an inhibitory signaling receptor and restrict the effector phase.


Subject(s)
CD8-Positive T-Lymphocytes/metabolism , Cadherins/metabolism , Adenocarcinoma/metabolism , Animals , Cadherins/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Cells, Cultured , Colonic Neoplasms/metabolism , Male , Mice
14.
Gastroenterology ; 141(5): 1915-26.e1-14, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21801698

ABSTRACT

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.


Subject(s)
Dendritic Cells/physiology , Pancreas/pathology , Pancreas/physiopathology , Pancreatitis/pathology , Pancreatitis/physiopathology , Tissue Survival/physiology , Acute Disease , Animals , Arginine/adverse effects , Ceruletide/adverse effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dendritic Cells/pathology , Diphtheria Toxin/pharmacology , Disease Models, Animal , Early Growth Response Protein 1/metabolism , Interleukin-6/metabolism , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Pancreatitis/chemically induced , Phenotype , Time Factors
15.
J Immunol ; 185(12): 7133-40, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21127315

ABSTRACT

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.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Neoplasms/immunology , Receptors, Antigen, T-Cell/immunology , Signal Transduction/immunology , Tumor Escape/immunology , Animals , Exocytosis/immunology , Humans
16.
J Immunol ; 185(4): 2200-8, 2010 Aug 15.
Article in English | MEDLINE | ID: mdl-20639479

ABSTRACT

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.


Subject(s)
Antigens/immunology , Endothelial Cells/immunology , Liver Cirrhosis/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carbon Tetrachloride , Cell Proliferation , Chemokines/metabolism , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endothelial Cells/metabolism , Flow Cytometry , Inflammation Mediators/metabolism , Liver/immunology , Liver/pathology , Liver Cirrhosis/chemically induced , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Regulatory/metabolism , Thioacetamide
17.
J Leukoc Biol ; 87(4): 713-25, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20042467

ABSTRACT

The liver is the most common site of adenocarcinoma metastases, even in patients who initially present with early disease. We postulated that immune-suppressive cells in the liver of tumor-bearing hosts inhibit anti-tumor T cells, thereby accelerating the growth of liver metastases. Using models of early preinvasive pancreatic neoplasia and advanced colorectal cancer, aims of this study were to determine immune phenotype, stimulus for recruitment, inhibitory effects, and tumor-enabling function of immune-suppressive cells in the liver of tumor-bearing hosts. We found that in mice with intra-abdominal malignancies, two distinct CD11b(+)Gr1(+) populations with divergent phenotypic and functional properties accumulate in the liver, becoming the dominant hepatic leukocytes. Their expansion is contingent on tumor expression of KC. These cells are distinct from CD11b(+)Gr1(+) populations in other tissues of tumor-bearing hosts in terms of cellular phenotype and cytokine and chemokine profile. Liver CD11b(+)Gr1(+) cells are highly suppressive of T cell activation, proliferation, and cytotoxicity and induce the development of Tregs. Moreover, liver myeloid-derived suppressor cells accelerate the development of hepatic metastases by inactivation of cytotoxic T cells. These findings may explain the propensity of patients with intra-abdominal cancers to develop liver metastases and suggest a promising target for experimental therapeutics.


Subject(s)
Abdominal Neoplasms/immunology , Immune Tolerance , Liver/immunology , Myeloid Cells/immunology , Pancreatic Neoplasms/immunology , Abdominal Neoplasms/metabolism , Abdominal Neoplasms/pathology , Animals , CD11b Antigen/biosynthesis , CD11b Antigen/immunology , Cell Proliferation , Chemokine CXCL1/biosynthesis , Chemokine CXCL1/immunology , Gene Expression Regulation, Neoplastic/immunology , Liver/metabolism , Liver/pathology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Transgenic , Myeloid Cells/metabolism , Myeloid Cells/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/immunology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Cytotoxic/pathology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology
18.
J Clin Invest ; 119(11): 3213-25, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19855130

ABSTRACT

Hepatic fibrosis occurs during most chronic liver diseases and is driven by inflammatory responses to injured tissue. Because DCs are central to modulating liver immunity, we postulated that altered DC function contributes to immunologic changes in hepatic fibrosis and affects the pathologic inflammatory milieu within the fibrotic liver. Using mouse models, we determined the contribution of DCs to altered hepatic immunity in fibrosis and investigated the role of DCs in modulating the inflammatory environment within the fibrotic liver. We found that DC depletion completely abrogated the elevated levels of many inflammatory mediators that are produced in the fibrotic liver. DCs represented approximately 25% of the fibrotic hepatic leukocytes and showed an elevated CD11b+CD8- fraction, a lower B220+ plasmacytoid fraction, and increased expression of MHC II and CD40. Moreover, after liver injury, DCs gained a marked capacity to induce hepatic stellate cells, NK cells, and T cells to mediate inflammation, proliferation, and production of potent immune responses. The proinflammatory and immunogenic effects of fibrotic DCs were contingent on their production of TNF-alpha. Therefore, modulating DC function may be an attractive approach to experimental therapeutics in fibro-inflammatory liver disease.


Subject(s)
Dendritic Cells/immunology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Proliferation , Cytokines/metabolism , Dendritic Cells/pathology , Gene Expression Regulation/immunology , Inflammation/immunology , Inflammation/pathology , Interleukin-6/metabolism , Killer Cells, Natural/metabolism , Liver/cytology , Liver/metabolism , Liver/pathology , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Phenotype
19.
J Leukoc Biol ; 84(3): 679-88, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18566103

ABSTRACT

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.


Subject(s)
Adenocarcinoma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Macrophages/immunology , Myeloid Cells/immunology , T-Lymphocytes, Helper-Inducer/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Apoptosis/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Cell Survival/immunology , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Fas Ligand Protein/physiology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Immunoblotting , Immunoenzyme Techniques , Immunophenotyping , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-13/immunology , Interleukin-13/metabolism , Interleukin-2/immunology , Interleukin-2/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/metabolism , Macrophages/pathology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Myeloid Cells/metabolism , Myeloid Cells/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I/physiology , Receptors, Tumor Necrosis Factor, Type II/physiology , Reverse Transcriptase Polymerase Chain Reaction , Survival Rate , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology
20.
Immunol Rev ; 222: 192-205, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18364003

ABSTRACT

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.


Subject(s)
Antigens, Neoplasm/immunology , Lymphocyte Activation , Neoplasms/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Tumor Escape , Animals , Antigen Presentation , CD3 Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines , Cytotoxicity, Immunologic , Epitopes , Humans , Immune Tolerance , Immunotherapy , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Mice , Myeloid Cells/immunology , Neoplasms/therapy , T-Lymphocytes/pathology , T-Lymphocytes, Regulatory/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...