Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
Add more filters










Publication year range
1.
Mol Immunol ; 113: 2-5, 2019 09.
Article in English | MEDLINE | ID: mdl-29126597

ABSTRACT

Major histocompatibility complex (MHC) class I molecules present peptide antigens to MHC class I-restricted CD8+ T lymphocytes. The peptides loaded onto MHC class I molecules are typically derived from cytosolic antigens, which includes both self and foreign proteins. In addition to this classical MHC class I antigen presentation pathway, some cell types, especially dendritic cells can present antigens from exogenous sources to MHC class I-restricted CD8+ T cells, in a process called cross-presentation. A variety of cellular processes, including endocytosis, vesicle trafficking, and autophagy, play critical roles in these antigen presentation pathways. In this review article, we discuss the role of autophagy, an intracellular degradation system that delivers cytoplasmic constituents to lysosomes, in MHC class I-restricted antigen presentation. A mechanistic understanding of the role of autophagy-related proteins in MHC class I restricted antigen presentation may guide future efforts in manipulating autophagy to prevent or treat human disease.


Subject(s)
Antigen Presentation/immunology , Autophagy/immunology , Histocompatibility Antigens Class I/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Humans , Lysosomes/immunology
2.
Proc Natl Acad Sci U S A ; 114(31): E6371-E6380, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28716903

ABSTRACT

The class III PI3K Vacuolar protein sorting 34 (Vps34) plays a role in both canonical and noncanonical autophagy, key processes that control the presentation of antigens by dendritic cells (DCs) to naive T lymphocytes. We generated DC-specific Vps34-deficient mice to assess the contribution of Vps34 to DC functions. We found that DCs from these animals have a partially activated phenotype, spontaneously produce cytokines, and exhibit enhanced activity of the classic MHC class I and class II antigen-presentation pathways. Surprisingly, these animals displayed a defect in the homeostatic maintenance of splenic CD8α+ DCs and in the capacity of these cells to cross-present cell corpse-associated antigens to MHC class I-restricted T cells, a property that was associated with defective expression of the T-cell Ig mucin (TIM)-4 receptor. Importantly, mice deficient in the Vps34-associated protein Rubicon, which is critical for a noncanonical form of autophagy called "Light-chain 3 (LC3)-associated phagocytosis" (LAP), lacked such defects. Finally, consistent with their defect in the cross-presentation of apoptotic cells, DC-specific Vps34-deficient animals developed increased metastases in response to challenge with B16 melanoma cells. Collectively, our studies have revealed a critical role of Vps34 in the regulation of CD8α+ DC homeostasis and in the capacity of these cells to process and present antigens associated with apoptotic cells to MHC class I-restricted T cells. Our findings also have important implications for the development of small-molecule inhibitors of Vps34 for therapeutic purposes.


Subject(s)
Antigen Presentation/immunology , Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Class III Phosphatidylinositol 3-Kinases/genetics , Cross-Priming/immunology , Dendritic Cells/immunology , Animals , Antigen Presentation/genetics , Autophagy/genetics , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/metabolism , CD8 Antigens/metabolism , Cells, Cultured , Class III Phosphatidylinositol 3-Kinases/metabolism , Cross-Priming/genetics , Cytokines/immunology , Endocytosis/physiology , Histocompatibility Antigens Class I/immunology , Melanoma, Experimental/pathology , Membrane Proteins/biosynthesis , Mice , Mice, Knockout , Phagocytosis/physiology
3.
Proc Natl Acad Sci U S A ; 113(32): E4662-70, 2016 08 09.
Article in English | MEDLINE | ID: mdl-27462110

ABSTRACT

Tregs are essential for maintaining peripheral tolerance, and thus targeting these cells may aid in the treatment of autoimmunity and cancer by enhancing or reducing suppressive functions, respectively. Before these cells can be harnessed for therapeutic purposes, it is necessary to understand how they maintain tolerance under physiologically relevant conditions. We now report that transcription factor Kruppel-like factor 2 (KLF2) controls naive Treg migration patterns via regulation of homeostatic and inflammatory homing receptors, and that in its absence KLF2-deficient Tregs are unable to migrate efficiently to secondary lymphoid organs (SLOs). Diminished Treg trafficking to SLOs is sufficient to initiate autoimmunity, indicating that SLOs are a primary site for maintaining peripheral tolerance under homeostatic conditions. Disease severity correlates with impaired Treg recruitment to SLOs and, conversely, promotion of Tregs into these tissues can ameliorate autoimmunity. Moreover, stabilizing KLF2 expression within the Treg compartment enhances peripheral tolerance by diverting these suppressive cells from tertiary tissues into SLOs. Taken together, these results demonstrate that peripheral tolerance is enhanced or diminished through modulation of Treg trafficking to SLOs, a process that can be controlled by adjusting KLF2 protein levels.


Subject(s)
Immune Tolerance , Kruppel-Like Transcription Factors/physiology , T-Lymphocytes, Regulatory/physiology , Animals , Autoimmunity , Cell Movement , Lymphoid Tissue/immunology , Mice , Receptors, Lymphocyte Homing/physiology
4.
Front Immunol ; 6: 226, 2015.
Article in English | MEDLINE | ID: mdl-26029211

ABSTRACT

Invariant natural killer T (iNKT) cells become activated during a wide variety of infections. This includes organisms lacking cognate CD1d-binding glycolipid antigens recognized by the semi-invariant T cell receptor of iNKT cells. Additional studies have shown that iNKT cells also become activated in vivo in response to microbial products such as bacterial lipopolysaccharide, a potent inducer of cytokine production in antigen-presenting cells (APCs). Other studies have shown that iNKT cells are highly responsive to stimulation by cytokines such as interleukin-12. These findings have led to the concept that microbial pathogens can activate iNKT cells either directly via glycolipids or indirectly by inducing cytokine production in APCs. iNKT cells activated in this manner produce multiple cytokines that can influence the outcome of infection, usually in favor of the host, although potent iNKT cell activation may contribute to an uncontrolled cytokine storm and sepsis. One aspect of the response of iNKT cells to microbial pathogens is that it is short-lived and followed by an extended time period of unresponsiveness to reactivation. This refractory period may represent a means to avoid chronic activation and cytokine production by iNKT cells, thus protecting the host against some of the negative effects of iNKT cell activation, but potentially putting the host at risk for secondary infections. These effects of microbial pathogens and their products on iNKT cells are not only important for understanding the role of these cells in immune responses against infections but also for the development of iNKT cell-based therapies.

5.
Immunology ; 146(1): 1-10, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26032048

ABSTRACT

Multiple sclerosis (MS) is a chronic inflammatory disease that causes demyelination of neurons in the central nervous system. Traditional therapies for MS have involved anti-inflammatory and immunosuppressive drugs with significant side effects that often only provide short-term relief. A more desirable outcome of immunotherapy would be to protect against disease before its clinical manifestation or to halt disease after its initiation. One attractive approach to accomplish this goal would be to restore tolerance by targeting immunoregulatory cell networks. Although much of the work in this area has focused on CD4(+) Foxp3(+) regulatory T cells, other studies have investigated natural killer T (NKT) cells, a subset of T cells that recognizes glycolipid antigens in the context of the CD1d glycoprotein. Studies with human MS patients have revealed alterations in the numbers and functions of NKT cells, which have been partially supported by studies with the experimental autoimmune encephalomyelitis model of MS. Additional studies have shown that activation of NKT cells with synthetic lipid antigens can, at least under certain experimental conditions, protect mice against the development of MS-like disease. Although mechanisms of this protection remain to be fully investigated, current evidence suggests that it involves interactions with other immunoregulatory cell types such as regulatory T cells and immunosuppressive myeloid cells. These studies have provided a strong foundation for the rational design of NKT-cell-based immunotherapies for MS that induce tolerance while sparing overall immune function. Nevertheless, additional pre-clinical and clinical studies will be required to bring this goal to fruition.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/immunology , Immunotherapy/methods , Lymphocyte Activation/immunology , Multiple Sclerosis/immunology , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/immunology , Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/therapy , Galactosylceramides/immunology , Galactosylceramides/therapeutic use , Humans , Mice , Mice, Inbred C57BL , Multiple Sclerosis/therapy
6.
J Infect Dis ; 211(3): 383-93, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25149763

ABSTRACT

Viruses that cause systemic disease often spread through the bloodstream to infect target tissues. Although viremia is an important step in the pathogenesis of many viruses, how viremia is established is not well understood. Reovirus has been used to dissect mechanisms of viral pathogenesis and is being evaluated in clinical trials as an oncolytic agent. After peroral entry into mice, reovirus replicates within the gastrointestinal tract and disseminates systemically via hematogenous or neural routes. Junctional adhesion molecule-A (JAM-A) is a tight junction protein that serves as a receptor for reovirus. JAM-A is required for establishment of viremia and viral spread to sites of secondary replication. JAM-A also is expressed on the surface of circulating hematopoietic cells. To determine contributions of endothelial and hematopoietic JAM-A to reovirus dissemination and pathogenesis, we generated strains of mice with altered JAM-A expression in these cell types and assessed bloodstream spread of reovirus strain type 1 Lang (T1L), which disseminates solely by hematogenous routes. We found that endothelial JAM-A but not hematopoietic JAM-A facilitates reovirus T1L bloodstream entry and egress. Understanding how viruses establish viremia may aid in development of inhibitors of this critical step in viral pathogenesis and foster engineering of improved oncolytic viral vectors.


Subject(s)
Cell Adhesion Molecules/metabolism , Endothelial Cells/metabolism , Receptors, Cell Surface/metabolism , Reoviridae/metabolism , Tight Junctions/metabolism , Viremia/metabolism , Animals , Cells, Cultured , Endothelial Cells/virology , Fibroblasts/metabolism , Fibroblasts/virology , Male , Mice , Mice, Inbred C57BL , Receptors, Virus/metabolism , Tight Junctions/virology , Viremia/virology
7.
Proc Natl Acad Sci U S A ; 111(43): E4638-47, 2014 Oct 28.
Article in English | MEDLINE | ID: mdl-25313053

ABSTRACT

Lipid accumulation in obesity triggers a low-grade inflammation that results from an imbalance between pro- and anti-inflammatory components of the immune system and acts as the major underlying mechanism for the development of obesity-associated diseases, notably insulin resistance and type 2 diabetes. Innate-like B cells are a subgroup of B cells that respond to innate signals and modulate inflammatory responses through production of immunomodulatory mediators such as the anti-inflammatory cytokine IL-10. In this study, we examined innate-like B cells in visceral white adipose tissue (VAT) and the relationship of these cells with their counterparts in the peritoneal cavity and spleen during diet-induced obesity (DIO) in mice. We show that a considerable number of innate-like B cells bearing a surface phenotype distinct from the recently identified "adipose natural regulatory B cells" populate VAT of lean animals, and that spleen represents a source for the recruitment of these cells in VAT during DIO. However, demand for these cells in the expanding VAT outpaces their recruitment during DIO, and the obese environment in VAT further impairs their function. We further show that removal of splenic precursors of innate-like B cells through splenectomy exacerbates, whereas supplementation of these cells via adoptive transfer ameliorates, DIO-associated insulin resistance. Additional adoptive transfer experiments pointed toward a dominant role of IL-10 in mediating the protective effects of innate-like B cells against DIO-induced insulin resistance. These findings identify spleen-supplied innate-like B cells in VAT as previously unrecognized players and therapeutic targets for obesity-associated diseases.


Subject(s)
Adipose Tissue, White/cytology , B-Lymphocytes/cytology , Insulin Resistance , Obesity/immunology , Obesity/prevention & control , Spleen/cytology , Spleen/immunology , Animals , CD5 Antigens/metabolism , Diet, High-Fat , Immunity, Innate , Interleukin-10/biosynthesis , Lymphocyte Count , Male , Mice, Inbred C57BL , Peritoneal Cavity/pathology , Phenotype , Splenectomy
8.
Immunity ; 41(3): 451-464, 2014 Sep 18.
Article in English | MEDLINE | ID: mdl-25220211

ABSTRACT

Innate immune responses are critical for mucosal immunity. Here we describe an innate lymphocyte population, iCD8α cells, characterized by expression of CD8α homodimers. iCD8α cells exhibit innate functional characteristics such as the capacity to engulf and kill bacteria. Development of iCD8α cells depends on expression of interleukin-2 receptor γ chain (IL-2Rγc), IL-15, and the major histocompatibility complex (MHC) class Ib protein H2-T3, also known as the thymus leukemia antigen or TL. While lineage tracking experiments indicated that iCD8α cells have a lymphoid origin, their development was independent of the transcriptional suppressor Id2, suggesting that these cells do not belong to the family of innate lymphoid cells. Finally, we identified cells with a similar phenotype in humans, which were profoundly depleted in newborns with necrotizing enterocolitis. These findings suggest a critical role of iCD8α cells in immune responses associated with the intestinal epithelium.


Subject(s)
Antigen Presentation/immunology , CD8 Antigens/biosynthesis , Immunity, Mucosal/immunology , Intestinal Mucosa/cytology , Lymphocytes/immunology , Animals , Citrobacter rodentium/immunology , Cytochalasin D/pharmacology , Enterocolitis, Necrotizing , Helicobacter pylori/immunology , Histocompatibility Antigens Class I/biosynthesis , Humans , Inhibitor of Differentiation Protein 2/genetics , Interleukin Receptor Common gamma Subunit/biosynthesis , Interleukin-15/biosynthesis , Interleukin-2/biosynthesis , Interleukin-7/biosynthesis , Intestinal Mucosa/immunology , Lymphocyte Activation/immunology , Lymphocytes/classification , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/drug effects , Phagocytosis/immunology
9.
Proc Natl Acad Sci U S A ; 111(26): 9579-84, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24979767

ABSTRACT

Regulatory T cells (Tregs) are a specialized subset of CD4(+) T cells that maintain self-tolerance by functionally suppressing autoreactive lymphocytes. The Treg compartment is composed of thymus-derived Tregs (tTregs) and peripheral Tregs (pTregs) that are generated in secondary lymphoid organs after exposure to antigen and specific cytokines, such as TGF-ß. With regard to this latter lineage, pTregs [and their ex vivo generated counterparts, induced Tregs (iTregs)] offer particular therapeutic potential because these cells can be raised against specific antigens to limit autoimmunity. We now report that transcription factor Krüppel-like factor 2 (KLF2) is necessary for the generation of iTregs but not tTregs. Moreover, drugs that limit KLF2 proteolysis during T-cell activation enhance iTreg development. To the authors' knowledge, this study identifies the first transcription factor to distinguish between i/pTreg and tTreg ontogeny and demonstrates that KLF2 is a therapeutic target for the production of regulatory T cells.


Subject(s)
Autoimmunity/immunology , Cell Differentiation/immunology , Kruppel-Like Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/genetics , Chromatin Immunoprecipitation , DNA Primers/genetics , Flow Cytometry , Kruppel-Like Transcription Factors/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
10.
PLoS One ; 8(7): e67821, 2013.
Article in English | MEDLINE | ID: mdl-23844100

ABSTRACT

In vitro CD4(+) T cell differentiation systems have made important contributions to understanding the mechanisms underlying the differentiation of naive CD4(+) T cells into effector cells with distinct biological functions. Mature CD4(+) T cells expressing CD8αα homodimers are primarily found in the intestinal mucosa of men and mice, and to a lesser extent in other tissues such as peripheral blood. Although CD4(+)CD8α(+) T cells are easily identified, very little is known about their development and immunological functions. It has been reported, however, that CD4(+)CD8α(+) T cells possess regulatory properties. In this report, we present a novel in vitro differentiation system where CD4(+) T cells are stimulated to become CD4(+)CD8α(+) T cells in the presence of TGF-ß, IL-7 and IFN-γ, resulting in cells with very similar features as CD4(+)CD8α(+) intraepithelial lymphocytes. This novel in vitro differentiation culture should provide a powerful and tractable tool for dissecting the differentiation and biological functions of CD4(+)CD8α(+) T cells.


Subject(s)
CD4 Antigens/metabolism , CD8 Antigens/metabolism , Interferon-gamma/pharmacology , Interleukin-7/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/pharmacology , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, CD/metabolism , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , CTLA-4 Antigen/genetics , CTLA-4 Antigen/metabolism , Cell Differentiation/drug effects , Cell Lineage/genetics , Core Binding Factor Alpha 3 Subunit/genetics , Core Binding Factor Alpha 3 Subunit/metabolism , Cytokines/biosynthesis , Immunophenotyping , Interferon-gamma/genetics , Intestinal Mucosa/immunology , Mice , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Programmed Cell Death 1 Receptor , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Vitamin D/pharmacology , Lymphocyte Activation Gene 3 Protein
11.
J Immunol ; 190(10): 5086-101, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23596309

ABSTRACT

Autophagy plays a critical role in multiple aspects of the immune system, including the development and function of T lymphocytes. In mammalian cells, the class III PI3K vacuolar protein sorting (Vps)34 is thought to play a critical role in autophagy. However, recent studies have cast doubt on the role of Vps34 in autophagy, at least in certain cell types. To study the effects of Vps34 on autophagy in T lymphocytes, we generated mice that selectively lack Vps34 in the T cell lineage. Vps34 ablation in T cells caused profound defects in autophagic flux, resulting in accumulation of cellular organelles and apoptosis. These animals exhibited normal intrathymic development of conventional T cells, but they were profoundly impaired in the intrathymic development of invariant NKT cells. In peripheral organs, T cell-specific ablation of Vps34 had a profound impact on T cell homeostasis and function. Furthermore, aged animals developed an inflammatory wasting syndrome characterized by weight loss, intestinal inflammation, and anemia. Consistent with this phenotype, Vps34 was required for the peripheral maintenance and function of CD4(+)Foxp3(+) regulatory T cells. Collectively, our study reveals a critical role for Vps34 in autophagy and for the peripheral homeostasis and function of T lymphocytes.


Subject(s)
Autophagy/genetics , Class III Phosphatidylinositol 3-Kinases/genetics , T-Lymphocytes, Regulatory/immunology , Wasting Syndrome/genetics , Wasting Syndrome/immunology , Adoptive Transfer , Aging , Animals , Apoptosis/genetics , Colitis/immunology , Forkhead Transcription Factors/metabolism , Inflammation , Interleukin-2/biosynthesis , Mice , Mice, Knockout , Natural Killer T-Cells/immunology , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
12.
J Immunol ; 190(5): 1948-60, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23345328

ABSTRACT

Invariant NKT (iNKT) cells are a subset of T lymphocytes that recognize glycolipid Ags presented by the MHC class I-related protein CD1d. Activation of iNKT cells with glycolipid Ags, such as the marine sponge-derived reagent α-galactosylceramide (α-GalCer), results in the rapid production of a variety of cytokines and activation of many other immune cell types. These immunomodulatory properties of iNKT cells have been exploited for the development of immunotherapies against a variety of autoimmune and inflammatory diseases, but mechanisms by which activated iNKT cells confer disease protection have remained incompletely understood. In this study, we demonstrate that glycolipid-activated iNKT cells cooperate with myeloid-derived suppressor cells (MDSCs) in protecting mice against the development of experimental autoimmune encephalomyelitis (EAE) in mice, an animal model for multiple sclerosis. We show that α-GalCer induced the expansion and immunosuppressive activities of MDSCs in the spleen of mice induced for development of EAE. Disease protection in these animals also correlated with recruitment of MDSCs to the CNS. Depletion of MDSCs abrogated the protective effects of α-GalCer against EAE and, conversely, adoptive transfer of MDSCs from α-GalCer-treated mice ameliorated passive EAE induced in recipient animals. The cytokines GM-CSF, IL-4, and IFN-γ, produced by activated iNKT cells, and inducible NO synthase, arginase-1, and IL-10 produced by MDSCs, contributed to these effects. Our findings have revealed cooperative immunosuppressive interactions between iNKT cells and MDSCs that might be exploited for the development of improved immunotherapies for multiple sclerosis and other autoimmune and inflammatory diseases.


Subject(s)
Autoimmunity/drug effects , Central Nervous System/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Galactosylceramides/pharmacology , Myeloid Cells/immunology , Natural Killer T-Cells/immunology , Adoptive Transfer , Animals , Antigen Presentation , Cell Communication/immunology , Cell Movement/drug effects , Cell Movement/immunology , Cell Proliferation , Central Nervous System/drug effects , Central Nervous System/pathology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Interleukin-4/biosynthesis , Interleukin-4/immunology , Mice , Mice, Transgenic , Myeloid Cells/drug effects , Myeloid Cells/pathology , Myeloid Cells/transplantation , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/pathology , Nitric Oxide Synthase Type II/immunology , Spleen/drug effects , Spleen/immunology , Spleen/pathology , Transplantation Chimera , Whole-Body Irradiation
13.
Trends Immunol ; 34(2): 50-8, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23017731

ABSTRACT

Invariant natural killer T (iNKT) cells are a subset of innate-like lymphocytes that recognize glycolipid antigens bound by the major histocompatibility complex (MHC)-class-I-related protein CD1d. iNKT cells are activated early during a variety of infections and inflammatory diseases and contribute to the subsequent development of adaptive immune responses. Consequently, iNKT cells play a critical role in the development and resolution of inflammatory diseases and represent attractive targets for the development of immunotherapies. Recent studies have provided important insight into the mechanisms by which iNKT cells become activated in response to diverse inflammatory stimuli. These new findings should be instrumental to promote the immunomodulatory properties of iNKT cells for treatment of inflammatory diseases.


Subject(s)
Inflammation/immunology , Inflammation/therapy , Lymphocyte Activation , Natural Killer T-Cells/immunology , Animals , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , Humans , Immunotherapy/methods , Infections/immunology , Infections/therapy , Mice
14.
Proc Natl Acad Sci U S A ; 109(19): E1143-52, 2012 May 08.
Article in English | MEDLINE | ID: mdl-22493234

ABSTRACT

Obesity triggers a low-grade systemic inflammation, which plays an important role in the development of obesity-associated metabolic diseases. In searching for links between lipid accumulation and chronic inflammation, we examined invariant natural killer T (iNKT) cells, a subset of T lymphocytes that react with lipids and regulate inflammatory responses. We show that iNKT cells respond to dietary lipid excess and become activated before or at the time of tissue recruitment of inflammatory leukocytes, and that these cells progressively increase proinflammatory cytokine production in obese mice. Such iNKT cells skew other leukocytes toward proinflammatory cytokine production and induce an imbalanced proinflammatory cytokine environment in multiple tissues. Further, iNKT cell deficiency ameliorates tissue inflammation and provides protection against obesity-induced insulin resistance and hepatic steatosis. Conversely, chronic iNKT cell stimulation using a canonical iNKT cell agonist exacerbates tissue inflammation and obesity-associated metabolic disease. These findings place iNKT cells into the complex network linking lipid excess to inflammation in obesity and suggest new therapeutic avenues for obesity-associated metabolic disorders.


Subject(s)
Fatty Liver/immunology , Galactosylceramides/physiology , Inflammation/immunology , Insulin Resistance/immunology , Natural Killer T-Cells/immunology , Obesity/immunology , Adipose Tissue, White/immunology , Adipose Tissue, White/metabolism , Adipose Tissue, White/pathology , Animals , Antigens, CD1d/genetics , Antigens, CD1d/immunology , Antigens, CD1d/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cells, Cultured , Cytokines/immunology , Cytokines/metabolism , Dietary Fats/administration & dosage , Dietary Fats/immunology , Fatty Liver/genetics , Female , Flow Cytometry , Galactosylceramides/administration & dosage , Galactosylceramides/immunology , Inflammation/genetics , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Insulin Resistance/genetics , Lipids/administration & dosage , Lipids/immunology , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Natural Killer T-Cells/metabolism , Obesity/genetics
15.
J Immunol ; 187(8): 4051-60, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21900182

ABSTRACT

The intestinal epithelium is comprised of a monolayer of intestinal epithelial cells (IEC), which provide, among other functions, a physical barrier between the high Ag content of the intestinal lumen and the sterile environment beyond the epithelium. IEC express a nonclassical MHC class I molecule known as the thymus leukemia (TL) Ag. TL is known to interact with CD8αα-expressing cells, which are abundant in the intestinal intraepithelial lymphocyte compartment. In this report, we provide evidence indicating that expression of TL by IEC modulates the cytokine profile of CD4(+) T cells favoring IL-17 production. We show in an adoptive transfer model of colitis that donor-derived cells become more pathogenic when TL is expressed on IEC in recipient animals. Moreover, TL(+)IEC promote development of IL-17-mediated responses capable of protecting mice from Citrobacter rodentium infection. We also show that modulation of IL-17-mediated responses by TL(+)IEC is controlled by the expression of CD8α on CD4(+) T cells. Overall, our results provide evidence for an important interaction between IEC and CD4(+) T cells via TL, which modulates mucosal immune responses.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Epithelial Cells/immunology , Immunity, Mucosal/immunology , Intestinal Mucosa/immunology , Membrane Glycoproteins/immunology , Adoptive Transfer , Animals , CD4-Positive T-Lymphocytes/metabolism , Cell Separation , Coculture Techniques , Colitis/immunology , Colitis/metabolism , Flow Cytometry , Intestinal Mucosa/metabolism , Lymphocyte Activation/immunology , Membrane Glycoproteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction
16.
Diabetes ; 60(11): 2922-7, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21896930

ABSTRACT

OBJECTIVE: Islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP), now known as G6PC2, is a major target of autoreactive T cells implicated in the pathogenesis of type 1 diabetes in both mice and humans. This study aimed to determine whether suppression of G6p2 gene expression might therefore prevent or delay disease progression. RESEARCH DESIGN AND METHODS: G6pc2(-/-) mice were generated on the NOD/ShiLtJ genetic background, and glycemia was monitored weekly up to 35 weeks of age to determine the onset and incidence of diabetes. The antigen specificity of CD8(+) T cells infiltrating islets from NOD/ShiLtJ G6pc2(+/+) and G6pc2(-/-) mice at 12 weeks was determined in parallel. RESULTS: The absence of G6pc2 did not affect the time of onset, incidence, or sex bias of type 1 diabetes in NOD/ShiLtJ mice. Insulitis was prominent in both groups, but whereas NOD/ShiLtJ G6pc2(+/+) islets contained CD8(+) T cells reactive to the G6pc2 NRP peptide, G6pc2 NRP-reactive T cells were absent in NOD/ShiLtJ G6pc2(-/-) islets. CONCLUSIONS: These results demonstrate that G6pc2 is an important driver for the selection and expansion of islet-reactive CD8(+) T cells infiltrating NOD/ShiLtJ islets. However, autoreactivity to G6pc2 is not essential for the emergence of autoimmune diabetes. The results remain consistent with previous studies indicating that insulin may be the primary autoimmune target, at least in NOD/ShiLtJ mice.


Subject(s)
Catalytic Domain , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/therapy , Disease Progression , Gene Deletion , Glucose-6-Phosphatase/genetics , Islets of Langerhans/metabolism , Proteins/genetics , Animals , Autoantibodies/analysis , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/metabolism , Diabetes Mellitus, Type 1/pathology , Female , Glucose-6-Phosphatase/chemistry , Humans , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Male , Mice , Mice, 129 Strain , Mice, Inbred NOD , Mice, Knockout , Mice, Transgenic , Proteins/chemistry , Sex Characteristics
17.
Cell Tissue Res ; 343(1): 43-55, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20734065

ABSTRACT

Cells of the innate immune system interact with pathogens via conserved pattern-recognition receptors, whereas cells of the adaptive immune system recognize pathogens through diverse, antigen-specific receptors that are generated by somatic DNA rearrangement. Invariant natural killer T (iNKT) cells are a subset of lymphocytes that bridge the innate and adaptive immune systems. Although iNKT cells express T cell receptors that are generated by somatic DNA rearrangement, these receptors are semi-invariant and interact with a limited set of lipid and glycolipid antigens, thus resembling the pattern-recognition receptors of the innate immune system. Functionally, iNKT cells most closely resemble cells of the innate immune system, as they rapidly elicit their effector functions following activation, and fail to develop immunological memory. iNKT cells can become activated in response to a variety of stimuli and participate in the regulation of various immune responses. Activated iNKT cells produce several cytokines with the capacity to jump-start and modulate an adaptive immune response. A variety of glycolipid antigens that can differentially elicit distinct effector functions in iNKT cells have been identified. These reagents have been employed to test the hypothesis that iNKT cells can be harnessed for therapeutic purposes in human diseases. Here, we review the innate-like properties and functions of iNKT cells and discuss their interactions with other cell types of the immune system.


Subject(s)
Adaptive Immunity/immunology , Immunity, Innate/immunology , Natural Killer T-Cells/immunology , Animals , Antigens/immunology , Humans , Immunotherapy , Lymphocyte Activation/immunology , Natural Killer T-Cells/cytology
18.
Immunotherapy ; 3(1): 59-75, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21174558

ABSTRACT

Invariant NK T (iNKT) cells are a subset of T lymphocytes that recognize glycolipid antigens bound with the antigen-presenting molecule CD1d. iNKT cells have potent immunoregulatory activities that can promote or suppress immune responses during different pathological conditions. These immunoregulatory properties can be harnessed for therapeutic purposes with cognate glycolipid antigens, such as the marine sponge-derived glycosphingolipid α-galactosylceramide. Preclinical studies have shown substantial promise for iNKT cell-based treatments of infections, cancer and autoimmune and inflammatory diseases. Translation of these preclinical studies to the clinic, while faced with some obstacles, has already had some initial success. In this article, we review the immunodulatory activities of iNKT cells and the potential for developing iNKT cell-based prophylactic and curative therapies of human disease.


Subject(s)
Antigens/immunology , Autoimmune Diseases/therapy , Glycolipids/immunology , Immunotherapy/methods , Infections/therapy , Natural Killer T-Cells/immunology , Neoplasms/therapy , Animals , Antigen Presentation , Autoimmune Diseases/immunology , Clinical Trials as Topic , Galactosylceramides/immunology , Humans , Infections/immunology , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Treatment Outcome
19.
Immunity ; 33(2): 254-65, 2010 Aug 27.
Article in English | MEDLINE | ID: mdl-20691614

ABSTRACT

Follicular (FO) and marginal zone (MZ) B cells are maintained in distinct locations within the spleen, but the genetic basis for this separation is still enigmatic. We now report that B cell sequestration requires lineage-specific regulation of migratory receptors by the transcription factor Klf2. Moreover, using gene-targeted mice we show that altered splenic B cell migration confers a significant in vivo gain-of-function phenotype to FO B cells, including the ability to quickly respond to MZ-associated antigens and pathogens in a T cell-dependent manner. This work demonstrates that in wild-type animals, naive FO B cells are actively removed from the MZ, thus restricting their capacity to respond to blood-borne pathogens.


Subject(s)
B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Movement , Immunity, Humoral , Spleen/cytology , Spleen/immunology , Animals , Antigens, CD19/genetics , Antigens, CD19/immunology , Antigens, T-Independent/genetics , Antigens, T-Independent/immunology , Bone Marrow/immunology , Cell Differentiation , Cells, Cultured , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/immunology , Mice , Mice, Knockout , Receptors, CCR/immunology
20.
J Immunol ; 183(3): 2016-26, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19587017

ABSTRACT

IL-4 contributes to immunopathology induced in mice by primary respiratory syncytial virus (RSV) infection. However, the cellular source of IL-4 in RSV infection is unknown. We identified CD3(-)CD49b(+) cells as the predominant source of IL-4 in the lungs of RSV-infected BALB/c mice. We ruled out T cells, NK cells, NKT cells, mast cells, and eosinophils as IL-4 expressors in RSV infection by flow cytometry. Using IL4 GFP reporter mice (4get) mice, we identified the IL-4-expressing cells in RSV infection as basophils (CD3(-)CD49b(+)FcepsilonRI(+)c-kit(-)). Because STAT1(-/-) mice have an enhanced Th2-type response to RSV infection, we also sought to determine the cellular source and role of IL-4 in RSV-infected STAT1(-/-) mice. RSV infection resulted in significantly more IL-4-expressing CD3(-)CD49b(+) cells in the lungs of STAT1(-/-) mice than in BALB/c mice. CD49b(+)IL-4(+) cells sorted from the lungs of RSV-infected STAT1(-/-) mice and stained with Wright-Giemsa had basophil characteristics. As in wild-type BALB/c mice, IL-4 contributed to lung histopathology in RSV-infected STAT1(-/-) mice. Depletion of basophils in RSV-infected STAT1(-/-) mice reduced lung IL-4 expression. Thus, we show for the first time that a respiratory virus (RSV) induced basophil accumulation in vivo. Basophils were the primary source of IL-4 in the lung in RSV infection, and STAT1 was a negative regulator of virus-induced basophil IL-4 expression.


Subject(s)
Basophils/virology , Gene Expression Regulation , Interleukin-4/genetics , Lung/metabolism , Respiratory Syncytial Virus Infections/immunology , STAT1 Transcription Factor/physiology , Animals , Basophils/metabolism , Basophils/pathology , Flow Cytometry , Immunophenotyping , Lung/pathology , Mice , Mice, Inbred BALB C
SELECTION OF CITATIONS
SEARCH DETAIL
...