Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Cancer Immunol Immunother ; 73(4): 64, 2024 Mar 02.
Article in English | MEDLINE | ID: mdl-38430289

ABSTRACT

Pancreatic cancer remains a challenging disease with limited treatment options, resulting in high mortality rates. The predominant approach to managing pancreatic cancer patients continues to be systemic cytotoxic chemotherapy. Despite substantial advancements in immunotherapy strategies for various cancers, their clinical utility in pancreatic cancer has proven less effective and durable. Whether administered as monotherapy, employing immune checkpoint inhibitors, tumor vaccines, chimeric antigen receptors T cells, or in combination with conventional chemoradiotherapy, the clinical outcomes remain underwhelming. Extensive preclinical experiments and clinical trials in the realm of pancreatic cancer have provided valuable insights into the complexities of immunotherapy. Chief among the hurdles are the immunosuppressive tumor microenvironment, limited immunogenicity, and the inherent heterogeneity of pancreatic cancer. In this comprehensive review, we provide an overview and critical analysis of current clinical immunotherapy strategies for pancreatic cancer, emphasizing their endeavors to overcome immunotherapy resistance. Particular focus is placed on strategies aimed at reshaping the immunosuppressive microenvironment and enhancing T cell-mediated tumor cell killing. Ultimately, through deeper elucidation of the underlying pathogenic mechanisms of pancreatic cancer and the refinement of therapeutic approaches, we anticipate breakthroughs that will pave the way for more effective treatments in this challenging disease.


Subject(s)
Antineoplastic Agents , Pancreatic Neoplasms , Humans , Immunotherapy/methods , Pancreatic Neoplasms/pathology , Antineoplastic Agents/therapeutic use , T-Lymphocytes , Treatment Outcome , Tumor Microenvironment
2.
Cancer Biol Med ; 21(4)2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38425216

ABSTRACT

OBJECTIVE: The human cluster of differentiation (CD)300A, a type-I transmembrane protein with immunoreceptor tyrosine-based inhibitory motifs, was investigated as a potential immune checkpoint for human natural killer (NK) cells targeting hematologic malignancies (HMs). METHODS: We implemented a stimulation system involving the CD300A ligand, phosphatidylserine (PS), exposed to the outer surface of malignant cells. Additionally, we utilized CD300A overexpression, a CD300A blocking system, and a xenotransplantation model to evaluate the impact of CD300A on NK cell efficacy against HMs in in vitro and in vivo settings. Furthermore, we explored the association between CD300A and HM progression in patients. RESULTS: Our findings indicated that PS hampers the function of NK cells. Increased CD300A expression inhibited HM lysis by NK cells. CD300A overexpression shortened the survival of HM-xenografted mice by impairing transplanted NK cells. Blocking PS-CD300A signals with antibodies significantly amplified the expression of lysis function-related proteins and effector cytokines in NK cells, thereby augmenting the ability to lyse HMs. Clinically, heightened CD300A expression correlated with shorter survival and an "exhausted" phenotype of intratumoral NK cells in patients with HMs or solid tumors. CONCLUSIONS: These results propose CD300A as a potential target for invigorating NK cell-based treatments against HMs.


Subject(s)
Hematologic Neoplasms , Killer Cells, Natural , Receptors, Immunologic , Humans , Killer Cells, Natural/immunology , Animals , Mice , Receptors, Immunologic/metabolism , Receptors, Immunologic/genetics , Receptors, Immunologic/immunology , Hematologic Neoplasms/immunology , Hematologic Neoplasms/therapy , Xenograft Model Antitumor Assays , Female , Antigens, CD/metabolism , Antigens, CD/immunology , Male , Cell Line, Tumor , Cytotoxicity, Immunologic , Phosphatidylserines/metabolism
4.
Cell Res ; 33(5): 372-388, 2023 05.
Article in English | MEDLINE | ID: mdl-37055591

ABSTRACT

Inflammatory bowel diseases (IBD) are known to have complex, genetically influenced etiologies, involving dysfunctional interactions between the intestinal immune system and the microbiome. Here, we characterized how the RNA transcript from an IBD-associated long non-coding RNA locus ("CARINH-Colitis Associated IRF1 antisense Regulator of Intestinal Homeostasis") protects against IBD. We show that CARINH and its neighboring gene coding for the transcription factor IRF1 together form a feedforward loop in host myeloid cells. The loop activation is sustained by microbial factors, and functions to maintain the intestinal host-commensal homeostasis via the induction of the anti-inflammatory factor IL-18BP and anti-microbial factors called guanylate-binding proteins (GBPs). Extending these mechanistic insights back to humans, we demonstrate that the function of the CARINH/IRF1 loop is conserved between mice and humans. Genetically, the T allele of rs2188962, the most probable causal variant of IBD within the CARINH locus from the human genetics study, impairs the inducible expression of the CARINH/IRF1 loop and thus increases genetic predisposition to IBD. Our study thus illustrates how an IBD-associated lncRNA maintains intestinal homeostasis and protects the host against colitis.


Subject(s)
Colitis , Inflammatory Bowel Diseases , RNA, Long Noncoding , Humans , Animals , Mice , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Intestines , Colitis/metabolism , Intestinal Mucosa/metabolism
5.
Cell Rep ; 41(9): 111741, 2022 11 29.
Article in English | MEDLINE | ID: mdl-36450257

ABSTRACT

Metabolic rewiring is essential for Th17 cells' functional identity to sense and interpret environmental cues. However, the environmental metabolic checkpoints with specific regulation of Th17 cells, manifesting potential therapeutic opportunities to autoimmune diseases, remain largely unknown. Here, by screening more than one hundred compounds derived from intestinal microbes or diet, we found that vitamin B5 (VB5) restrains Th17 cell differentiation as well as related autoimmune diseases such as experimental autoimmune encephalomyelitis and colitis. Mechanistically, VB5 is catabolized into coenzyme A (CoA) in a pantothenate kinase (PANK)-dependent manner, and in turn, CoA binds to pyruvate kinase isoform 2 (PKM2) to impede its phosphorylation and nuclear translocation, thus inhibiting glycolysis and STAT3 phosphorylation. In humans, reduced serum VB5 levels are found in both IBD and MS patients. Collectively, our study demonstrates a role of VB5 in Th17 cell metabolic reprograming, thus providing a potential therapeutic intervention for Th17 cell-associated autoimmune diseases.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental , Pyruvate Kinase , Humans , Animals , Pantothenic Acid , Th17 Cells , Protein Isoforms , Coenzyme A
6.
Front Immunol ; 13: 906122, 2022.
Article in English | MEDLINE | ID: mdl-35757706

ABSTRACT

Food allergy is a growing concern due to its increasing world-wide incidence. Strict avoidance of allergens is a passive treatment strategy. Since the mechanisms responsible for the occurrence and development of food allergy have not yet been fully elucidated, effective individualized treatment options are lacking. In this review, we summarize the pathways through which food antigens enter the intestine and review the proposed mechanisms describing how the intestine acquires and tolerates food antigens. When oral tolerance is not established, food allergy occurs. In addition, we also discuss the contribution of commensal bacteria of the gut in shaping tolerance to food antigens in the intestinal tract. Finally, we propose that elucidating the mechanisms of intestinal uptake and tolerance of food antigens will provide additional clues for potential treatment options for food allergy.


Subject(s)
Allergens , Antigens/metabolism , Food Hypersensitivity , Bacteria , Food , Humans , Immune Tolerance
7.
Cell Mol Immunol ; 17(2): 178-189, 2020 02.
Article in English | MEDLINE | ID: mdl-30874628

ABSTRACT

Liver-resident NK cells are distinct from conventional NK cells and play an important role in the maintenance of liver homeostasis. How liver-resident NK cells participate in autoimmune cholangitis remains unclear. Here, we extensively investigated the impact of NK cells in the pathogenesis of autoimmune cholangitis utilizing the well-established dnTGFßRII cholangitis model, NK cell-deficient (Nfil3-/-) mice, adoptive transfer and in vivo antibody-mediated NK cell depletion. Our data demonstrated that disease progression was associated with a significantly reduced frequency of hepatic NK cells. Depletion of NK cells resulted in exacerbated autoimmune cholangitis in dnTGFßRII mice. We further confirmed that the DX5-CD11chi liver-resident NK cell subset colocalized with CD4+ T cells and inhibited CD4+ T cell proliferation. Gene expression microarray analysis demonstrated that liver-resident NK cells had a distinct gene expression pattern consisting of the increased expression of genes involved in negative regulatory functions in the context of the inflammatory microenvironment.


Subject(s)
Autoimmune Diseases/complications , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/immunology , Killer Cells, Natural/immunology , Liver Cirrhosis, Biliary/complications , Liver Cirrhosis, Biliary/immunology , Liver/immunology , Adoptive Transfer/methods , Animals , Autoimmune Diseases/blood , B-Lymphocytes/immunology , Basic-Leucine Zipper Transcription Factors/deficiency , Basic-Leucine Zipper Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , Cytokines/blood , Disease Models, Animal , Disease Progression , Female , Hybridomas , Liver Cirrhosis, Biliary/blood , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptor, Transforming Growth Factor-beta Type II/genetics
8.
Cell Mol Immunol ; 16(3): 216-224, 2019 03.
Article in English | MEDLINE | ID: mdl-30787416

ABSTRACT

Although lymphocytes are known to circulate throughout lymphoid tissues and blood, they also establish residency in nonlymphoid organs, most prominently in barrier tissues, such as the intestines. The adaptation of T lymphocytes to intestinal environments requires constant discrimination between natural stimulation from commensal flora and food and pathogens that need to be cleared. Genetic variations that cause a defective defense or a break in tolerance along with environmental cues, such as infection or imbalances in the gut microbiota known as dysbiosis, can trigger several immune disorders via the activation of T lymphocytes in the intestines. Elucidation of the immune mechanisms that distinguish between commensal flora and pathogenic organisms may reveal therapeutic targets for the prevention or modulation of inflammatory diseases and boost the efficacy of cancer immunotherapy. In this review, we discuss the development and adaptation of T lymphocytes in the intestine, how these cells protect the host against pathogenic infections while tolerating food antigens and commensal microbiota, and the potential implications of targeting these cells for disease management and therapeutics.


Subject(s)
Dysbiosis/immunology , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Adaptive Immunity , Animals , Host-Pathogen Interactions , Humans , Immune Tolerance , Lymphocyte Activation , Microbiota/immunology
9.
J Autoimmun ; 95: 47-57, 2018 12.
Article in English | MEDLINE | ID: mdl-30340822

ABSTRACT

Gut microbiota and bacterial translocation have been implicated as significant contributors to mucosal immune responses and tolerance; alteration of microbial molecules, termed pathogen-associated molecular patterns (PAMP) and bacterial translocation are associated with immune pathology. However, the mechanisms by which dysregulated gut microbiota promotes autoimmunity is unclear. We have taken advantage of a well-characterized murine model of primary biliary cholangitis, dnTGFßRII mice, and an additional unique construct, toll-like receptor 2 (TLR2)-deficient dnTGFßRII mice coined dnTGFßRIITLR2-/- mice to investigate the influences of gut microbiota on autoimmune cholangitis. Firstly, we report that dnTGFßRII mice manifest altered composition of gut microbiota and that alteration of this gut microbiota by administration of antibiotics significantly alleviates T-cell-mediated infiltration and bile duct damage. Second, toll-like receptor 2 (TLR2)-deficient dnTGFßRII mice demonstrate significant exacerbation of autoimmune cholangitis when their epithelial barrier integrity was disrupted. Further, TLR2-deficiency mediates downregulated expression of tight junction-associated protein ZO-1 leading to increased gut permeability and bacterial translocation from gut to liver; use of antibiotics reduces microbiota translocation to liver and also decreases biliary pathology. In conclusion, our data demonstrates the important role of gut microbiota and bacterial translocation in the pathogenesis of murine autoimmune cholangitis.


Subject(s)
Autoimmune Diseases/microbiology , Bacterial Translocation/immunology , Bile Ducts/immunology , Liver Cirrhosis, Biliary/immunology , Receptor, Transforming Growth Factor-beta Type II/immunology , Toll-Like Receptor 2/immunology , Ampicillin/pharmacology , Animals , Anti-Bacterial Agents/pharmacology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/pathology , Bacterial Translocation/drug effects , Bile Ducts/drug effects , Bile Ducts/microbiology , Bile Ducts/pathology , Colon/drug effects , Colon/immunology , Colon/microbiology , Colon/pathology , Feces/microbiology , Female , Gastrointestinal Microbiome/drug effects , Gastrointestinal Microbiome/immunology , Gene Expression Regulation , Immunity, Mucosal/drug effects , Liver/drug effects , Liver/immunology , Liver/microbiology , Liver/pathology , Liver Cirrhosis, Biliary/drug therapy , Liver Cirrhosis, Biliary/microbiology , Liver Cirrhosis, Biliary/pathology , Metronidazole/pharmacology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neomycin/pharmacology , Receptor, Transforming Growth Factor-beta Type II/deficiency , Receptor, Transforming Growth Factor-beta Type II/genetics , Signal Transduction , Toll-Like Receptor 2/deficiency , Toll-Like Receptor 2/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/immunology
10.
Sci Rep ; 7(1): 15246, 2017 11 10.
Article in English | MEDLINE | ID: mdl-29127360

ABSTRACT

Primary biliary cholangitis (PBC) is an autoimmune liver disease characterized by the destruction of interlobular biliary ductules, which progressively leads to cholestasis, hepatic fibrosis, cirrhosis, and eventually liver failure. Several mouse models have been used to clarify the pathogenesis of PBC and are generally considered reflective of an autoimmune cholangitis. Most models focus on issues of molecular mimicry between the E2 subunit of the pyruvate dehydrogenase complex (PDC-E2), the major mitochondrial autoantigen of PBC and xenobiotic cross reactive chemicals. None have focused on the classic models of breaking tolerance, namely immunization with self-tissue. Here, we report a novel mouse model of autoimmune cholangitis via immunization with syngeneic bile duct protein (BDP). Our results demonstrate that syngeneic bile duct antigens efficiently break immune tolerance of recipient mice, capturing several key features of PBC, including liver-specific inflammation focused on portal tract areas, increased number and activation state of CD4 and CD8 T cells in the liver and spleen. Furthermore, the germinal center (GC) responses in the spleen were more enhanced in our mouse model. Finally, these mice were 100% positive for anti-mitochondrial antibodies (AMAs). In conclusion, we developed a novel mouse model of PBC that may help to elucidate the detailed mechanism of this complex disease.


Subject(s)
Autoantigens , Autoimmune Diseases , Bile Ducts , Cholangitis , Immunization , Animals , Autoantigens/immunology , Autoantigens/toxicity , Autoimmune Diseases/chemically induced , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , Bile Ducts/immunology , Bile Ducts/pathology , Cholangitis/chemically induced , Cholangitis/immunology , Cholangitis/pathology , Disease Models, Animal , Female , Mice
11.
Front Immunol ; 8: 1192, 2017.
Article in English | MEDLINE | ID: mdl-29018448

ABSTRACT

Autoimmune diseases often induce dysregulated hematopoiesis with altered number and function of hematopoietic stem and progenitor cells (HSPCs). However, there are limited studies on the direct regulation of HSPCs on T cells, which are often detrimental to autoimmunity. Here, we found that in a murine model of Concanavalin A-induced autoimmune hepatitis, LSK (Lineage-Sca-1+c-Kit+)-like cells accumulated in liver, spleen, and bone marrow (BM), which were myeloid progenitors (Lineage-Sca-1-c-Kit+) that upregulated Sca-1 expression upon T cell-derived IFN-γ stimulation. Strikingly, BM LSK-like cells from mice induced by Con A to develop autoimmune hepatitis or alternatively myeloid progenitors from wild-type mice possessed strong in vitro suppressive ability. Their suppressive function depended on T cell-derived IFN-γ in a paracrine fashion, which induced STAT1 phosphorylation, inducible nitric oxide synthase expression, and nitric oxide production. Blocking IFN-γ/IFN-γ receptor interaction, knockout of STAT1, or iNOS inhibition abrogated their suppressive function. In addition, the suppressive function was independent of differentiation; mitomycin C-treated myeloid progenitors maintained T cell suppressive ability in vitro. Our data demonstrate a mechanism of inflammation induced suppressive function of myeloid progenitors, which may participate directly in suppressing T cell-mediated immunopathology.

12.
Immunity ; 47(2): 284-297.e5, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28813659

ABSTRACT

Ten-Eleven-Translocation-2 (Tet2) is a DNA methylcytosine dioxygenase that functions as a tumor suppressor in hematopoietic malignancies. We examined the role of Tet2 in tumor-tissue myeloid cells and found that Tet2 sustains the immunosuppressive function of these cells. We found that Tet2 expression is increased in intratumoral myeloid cells both in mouse models of melanoma and in melanoma patients and that this increased expression is dependent on an IL-1R-MyD88 pathway. Ablation of Tet2 in myeloid cells suppressed melanoma growth in vivo and shifted the immunosuppressive gene expression program in tumor-associated macrophages to a proinflammatory one, with a concomitant reduction of the immunosuppressive function. This resulted in increased numbers of effector T cells in the tumor, and T cell depletion abolished the reduced tumor growth observed upon myeloid-specific deletion of Tet2. Our findings reveal a non-cell-intrinsic, tumor-promoting function for Tet2 and suggest that Tet2 may present a therapeutic target for the treatment of non-hematologic malignancies.


Subject(s)
Carcinogenesis , DNA-Binding Proteins/metabolism , Melanoma/immunology , Myeloid-Derived Suppressor Cells/immunology , Proto-Oncogene Proteins/metabolism , Skin Neoplasms/immunology , T-Lymphocytes/immunology , Animals , Dioxygenases , Female , Humans , Male , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Tumor Burden , Tumor Escape
13.
Cell Death Dis ; 8(5): e2827, 2017 05 25.
Article in English | MEDLINE | ID: mdl-28542148

ABSTRACT

Molecular mechanisms that modulate liver regeneration are of critical importance for a number of hepatic disorders. Kupffer cells and natural killer (NK) cells are two cell subsets indispensable for liver regeneration. We have focused on these two populations and, in particular, the interplay between them. Importantly, we demonstrate that deletion of the myeloid phosphatase and tensin homolog on chromosome 10 (PTEN) leading to an M2-like polarization of Kupffer cells, which results in decreased activation of NK cells. In addition, PTEN-deficient Kupffer cells secrete additional factors that facilitate the proliferation of hepatocytes. In conclusion, PTEN is critical for inhibiting M2-like polarization of Kupffer cells after partial hepatectomy, resulting in NK cell activation and thus the inhibition of liver regeneration. Furthermore, PTEN reduces growth factor secretion by Kupffer cells. Our results suggest that targeting PTEN on Kupffer cells may be useful in altering liver regeneration in patients undergoing liver resection.


Subject(s)
Liver Regeneration , Myeloid Cells/metabolism , PTEN Phosphohydrolase/deficiency , Animals , Cell Polarity , Hepatectomy , Hepatocytes/metabolism , Killer Cells, Natural/metabolism , Kupffer Cells , Mice, Inbred C57BL , Mice, Knockout , Mitogens/metabolism , Models, Biological , PTEN Phosphohydrolase/metabolism
14.
J Autoimmun ; 78: 19-28, 2017 03.
Article in English | MEDLINE | ID: mdl-28129932

ABSTRACT

CXC Chemokine Receptor 3 (CXCR3) is functionally pleiotropic and not only plays an important role in chemotaxis, but also participates in T cell differentiation and may play a critical role in inducing and maintaining immune tolerance. These observations are particularly critical for autoimmune cholangitis in which CXCR3 positive T cells are found around the portal areas of both humans and mouse models of primary biliary cholangitis (PBC). Herein, we investigated the role of CXCR3 in the pathogenesis of autoimmune cholangitis. We have taken advantage of a unique CXCR3 knockout dnTGFßRII mouse to focus on the role of CXCR3, both by direct observation of its influence on the natural course of disease, as well as through adoptive transfer studies into Rag-/- mice. We report herein that not only do CXCR3 deficient mice develop an exacerbation of autoimmune cholangitis associated with an expanded effector memory T cell number, but also selective adoptive transfer of CXCR3 deficient CD8+ T cells induces autoimmune cholangitis. In addition, gene microarray analysis of CXCR3 deficient CD8+ T cells reveal an intense pro-inflammatory profile. Our data suggests that the altered gene profiles induced by CXCR3 deficiency promotes autoimmune cholangitis through pathogenic CD8+ T cells. These data have significance for human PBC and other autoimmune liver diseases in which therapeutic intervention might be directed to chemokines and/or their receptors.


Subject(s)
Autoimmunity/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Receptors, CXCR3/deficiency , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Cytokines/blood , Cytokines/metabolism , Disease Models, Animal , Gene Expression Profiling , Immunologic Memory , Ligands , Liver Cirrhosis, Biliary/metabolism , Liver Cirrhosis, Biliary/pathology , Mice , Mice, Knockout , Receptors, CXCR3/metabolism
15.
Cell Death Dis ; 7(10): e2439, 2016 10 27.
Article in English | MEDLINE | ID: mdl-27787514

ABSTRACT

Understanding the mechanisms that lead to autoimmunity is critical for defining potential therapeutic pathways. In this regard there have been considerable efforts in investigating the interacting roles of TGF-ß and IL-2 on the function regulatory T cells. We have taken advantage of dnTGF-ßRII Il2ra-/- (abbreviated as Il2ra-/-Tg) mouse model, which allows a direct mechanistic approach to define the relative roles of TGF-ß and IL-2 on Treg development. Il2ra-/-Tg mice spontaneously developed multi-organ autoimmune diseases with expansion of pathogenic T cells and enhanced germinal center response at 3-4 weeks of age. Importantly, peripheral Treg cells from Il2ra-/-Tg mice demonstrated an activated Th1-like stable phenotype and normal in vitro suppressive function, while thymus Treg increased but manifested decreased suppressive function. Interestingly, neither thymus nor peripheral Treg cells of Il2ra-/-Tg mice contained Neuropilin-1+ or PD-1hi phenotype, resulting in defective follicular regulatory T (Tfr) cell development. Such defective Tfr development led to elevated follicular T helper cells, enhanced germinal center responses and increased plasma cell infiltration. These data demonstrate an important synergetic role of TGF-ß and IL-2 in the generation, activation and stability of Treg cells, as well as their subsequent development into Tfr cells.


Subject(s)
Immunophilins/metabolism , Interleukin-2/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Transforming Growth Factor beta/metabolism , Animals , Germinal Center/metabolism , Inflammation/pathology , Interleukin-2 Receptor alpha Subunit/metabolism , Lymphadenopathy/pathology , Lymphocyte Activation/immunology , Mice, Transgenic , Phenotype , T-Lymphocytes, Helper-Inducer/cytology
16.
Oncotarget ; 7(19): 26992-7006, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27105495

ABSTRACT

Primary biliary cholangitis (PBC) is a chronic autoimmune liver disease with progressive cholestasis and liver fibrosis. Similar to human patients with PBC, p40-/-IL-2Rα-/- mice spontaneously develop severe autoimmune cholangitis. Although there has been considerable work on immune regulation and autoimmunity, there is a relative paucity of work directed at the functional implications of the key peritoneal cavity (PC) B cell subset, coined B1a cells in PBC. We used flow cytometry and high-resolution microarrays to study the qualitative and quantitative characteristics of B cells, particularly B1a cells, in the PC of p40-/-IL-2Rα-/- mice compared to controls. Importantly, B1a cell proliferation was markedly lower as the expression of Ki67 decreased. Meanwhile, the apoptosis level was much higher. These lead to a reduction of B1a cells in the PC of p40-/-IL-2Rα-/- mice compared to controls. In contrast, there was a dramatic increase of CD4+ and CD8+ T cells accompanied by elevated production of IFN-γ. In addition, we found a negative correlation between the frequency of B1a cells and the presence of autoreactive CD8+ T cells in both liver and PC of p40-/-IL-2Rα-/- mice. From a functional perspective, B cells from p40-/-IL-2Rα-/- mice downregulated IL-10 production and CTLA-4 expression, leading to loss of B cell regulatory function. We suggest that the dysfunction of B1a cells in the PC in this murine model of autoimmune cholangitis results in defective regulatory function. This highlights a new potential therapeutic target in PBC.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocyte Subsets/immunology , Liver Cirrhosis, Biliary/immunology , Liver/immunology , Animals , Apoptosis/genetics , Apoptosis/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , B-Lymphocyte Subsets/metabolism , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-12 Subunit p40/genetics , Interleukin-12 Subunit p40/immunology , Interleukin-12 Subunit p40/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Liver/metabolism , Liver/pathology , Liver Cirrhosis, Biliary/genetics , Liver Cirrhosis, Biliary/metabolism , Mice, Inbred C57BL , Mice, Knockout , Peritoneal Cavity/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transcriptome/genetics , Transcriptome/immunology
17.
Clin Rev Allergy Immunol ; 51(3): 353-369, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26924010

ABSTRACT

The functions of macrophages that lead to effective host responses are critical for protection against Staphylococcus aureus. Deep tissue-invading S. aureus initially countered by macrophages trigger macrophage accumulation and induce inflammatory responses through surface receptors, especially toll-like receptor 2 (TLR2). Here, we found that macrophages formed sporadic aggregates in the liver during infection. Within those aggregates, macrophages co-localized with T cells and were indispensable for their infiltration. In addition, we have focused on the mechanisms underlying the polarization of macrophages in Forkhead box transcription factor O1 (FoxO1) conditional knockout Lys Cre/+ FoxO1 fl/fl mice following S. aureus infection and report herein that macrophage M1-M2 polarization via TLR2 is intrinsically regulated by FoxO1. Indeed, for effective FoxO1 activity, stimulation of TLR2 is essential. However, following S. aureus challenge, there was a decrease in macrophage FoxO1, with increased phosphorylation of FoxO1 because of TLR2-mediated activation of PI3K/Akt and c-Raf/MEK/ERK pathway. Following infection in Lys Cre/+ FoxO1 fl/fl mice, mice became more susceptible to S. aureus with reduced macrophage aggregation in the liver and attenuated Th1 and Th17 responses. FoxO1 abrogation reduced M1 pro-inflammatory responses triggered by S. aureus and enhanced M2 polarization in macrophages. In contrast, overexpression of FoxO1 in macrophages increased pro-inflammatory mediators and functional surface molecule expression. In conclusion, macrophage FoxO1 is critical to promote M1 polarization and maintain a competent T cell immune response against S. aureus infection in the liver. FoxO1 regulates macrophage M1-M2 polarization downstream of TLR2 dynamically through phosphorylation.


Subject(s)
Forkhead Box Protein O1/metabolism , Macrophages/immunology , Macrophages/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/immunology , Animals , Biomarkers , Disease Models, Animal , Forkhead Box Protein O1/genetics , Humans , Immunomodulation , Immunophenotyping , Liver/immunology , Liver/metabolism , Liver/pathology , Macrophage Activation/genetics , Macrophage Activation/immunology , Mice , Mice, Knockout , Signal Transduction , Staphylococcal Infections/genetics , Staphylococcal Infections/pathology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology , Toll-Like Receptor 2/metabolism
18.
Clin Rev Allergy Immunol ; 50(3): 390-403, 2016 Jun.
Article in English | MEDLINE | ID: mdl-26267705

ABSTRACT

Primary biliary cirrhosis (PBC) is a chronic inflammatory autoimmune disease that develops based upon the interaction of genetic and environmental factors. Recent genome-wide association studies (GWAS) have identified dozens of predisposing variants including HLA, IL12A, and CTLA4 but have been disappointed in identifying a "smoking gun." These discoveries highlight the importance of the genetic background involved in immunological dysregulation. Although concordance rate of PBC in monozygotic (MZ) twins is among the highest reported in autoimmune disorders, incomplete disease concordance in twins associated with differentially expressed genes has been demonstrated. However, little is understood about how environmental aspects contribute to the disease and why middle-aged women are more susceptible. As a result, epigenetic factors, which convert signals indicating environmental changes into dynamic and heritable alterations of transcriptional potential, are getting increased attention by researchers in both basic and clinical studies. Among epigenetic mechanisms, the instability and skewed gene expression in the X chromosome may account for the female preponderance in PBC. In addition, transcriptional regulation of histone modification and DNA methylation underscores potential involvement in disease pathogenesis. High-throughput techniques are being used to identify epigenetic regulators. In this review, we attempt to outline recent progress regarding epigenetics in PBC and other autoimmune diseases.


Subject(s)
Epigenesis, Genetic , Genetic Predisposition to Disease , Liver Cirrhosis, Biliary/genetics , Animals , Autoimmunity/genetics , DNA Methylation , Gene Expression Regulation , Histones/metabolism , Humans , Liver Cirrhosis, Biliary/immunology , Liver Cirrhosis, Biliary/metabolism , MicroRNAs/genetics , RNA, Long Noncoding/genetics , X Chromosome Inactivation
19.
J Autoimmun ; 66: 108-17, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26432598

ABSTRACT

There is a significant unmet need in the treatment of primary biliary cirrhosis (PBC) despite significant data on the effector pathways that lead to biliary duct damage. We focused attention on a murine model of PBC, the dominant negative transforming growth factor ß receptor II (Tg) mice. To further define the pathways that lead to biliary pathology in these mice, we developed Tg mice deleted of CD4 cells (CD4(-/-)Tg). Interestingly, these mice developed more severe cholangitis than control Tg mice. These mice, which lack CD4 cells, manifested increased levels of IFN-γ produced by effector CD8 cells. It appears that increased cholangitis is due to the absence of CD4 Treg cells. Based on these data, we parabiosed CD4(-/-)Tg mice with established disease at 8-9 weeks of age with C57BL/6 control mice. Such parabiotic "twins" had a significant reduction in autoimmune cholangitis, even though they had established pathology at the time of surgery. We prepared mixed bone marrow chimera mice constructed from CD4(-/-)Tg and CD8(-/-) mice and not only was cholangitis improved, but a decrease in terminally differentiated CD8(+) T effector cells in the presence of wild type CD4 cells was noted. In conclusion, "correcting" the CD4 T cell subset, even in the presence of pathogenic CD8 T cells, is effective in treating autoimmune cholangitis.


Subject(s)
Autoimmune Diseases/surgery , Cholangitis/surgery , Liver Cirrhosis, Biliary/surgery , Parabiosis/methods , Animals , Autoimmune Diseases/immunology , Bile Ducts/immunology , Bile Ducts/pathology , CD4 Antigens/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cholangitis/immunology , Disease Models, Animal , Female , Hematopoietic Stem Cell Transplantation/methods , Humans , Liver/immunology , Liver/pathology , Liver Cirrhosis, Biliary/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Receptors, Transforming Growth Factor beta/immunology
20.
J Autoimmun ; 61: 62-72, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26071985

ABSTRACT

Thymic CD4(+) FoxP3(+) regulatory T (Treg) cells are critical for the development of immunological tolerance and immune homeostasis and requires contributions of both thymic dendritic and epithelial cells. Although B cells have been reported to be present within the thymus, there has not hitherto been a definition of their role in immune cell development and, in particular, whether or how they contribute to the Treg cellular thymic compartment. Herein, using both phenotypic and functional approaches, we demonstrate that thymic B cells contribute to the maintenance of thymic Treg cells and, using an in vitro culture system, demonstrate that thymic B cells contribute to the size of the thymic Treg compartment via cell-cell MHC II contact and the involvement of two independent co-stimulatory pathways that include interactions between the CD40/CD80/CD86 co-stimulatory molecules. Our data also suggest that thymic B cells promote the generation of thymic Treg cell precursors (pre-Treg cells), but not the conversion of FoxP3(+) Treg cells from pre-Treg cells. In addition, thymic B cells directly promote the proliferation of thymic Treg cells that is MHC II contact dependent with a minimal if any role for co-stimulatory molecules including CD40/CD80/CD86. Both pathways are independent of TGFß. In conclusion, we rigorously define the critical role of thymic B cells in the development of thymic Treg cells from non-Treg to precursor stage and in the proliferation of mature thymic Treg cells.


Subject(s)
B-Lymphocytes/immunology , Cell Proliferation , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology , Animals , B-Lymphocytes/metabolism , B7-1 Antigen/immunology , B7-2 Antigen/immunology , CD40 Antigens/immunology , Cell Differentiation/immunology , Flow Cytometry , Histocompatibility Antigens Class II/immunology , Homeostasis/immunology , Immune Tolerance/immunology , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , T-Lymphocytes, Regulatory/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL