Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
bioRxiv ; 2024 Jan 24.
Article in English | MEDLINE | ID: mdl-38328096

ABSTRACT

Objectives: Sjögren's Disease (SjD) is an autoimmune disorder characterized by progressive dysfunction, inflammation and destruction of salivary and lacrimal glands, and by extraglandular manifestations. Its etiology and pathophysiology remain incompletely understood, though a role for autoreactive B cells has been considered key. Here, we investigated the role of effector and regulatory T cells in the pathogenesis of SjD. Methods: Histological analysis, RNA-sequencing and flow cytometry were conducted on glands, lungs, eyes and lymphoid tissues of mice with regulatory T cell-specific deletion of stromal interaction proteins (STIM) 1 and 2 ( Stim1/2 Foxp3 ), which play key roles in calcium signaling and T cell function. The pathogenicity of T cells from Stim1/2 Foxp3 mice was investigated through adoptively transfer into lymphopenic host mice. Additionally, single-cell transcriptomic analysis was performed on peripheral blood mononuclear cells (PBMCs) of patients with SjD and control subjects. Results: Stim1/2 Foxp3 mice develop a severe SjD-like disorder including salivary gland (SG) and lacrimal gland (LG) inflammation and dysfunction, autoantibodies and extraglandular symptoms. SG inflammation in Stim1/2 Foxp3 mice is characterized by T and B cell infiltration, and transcriptionally by a Th1 immune response that correlates strongly with the dysregulation observed in patients with SjD. Adoptive transfer of effector T cells from Stim1/2 Foxp3 mice demonstrates that the SjD-like disease is driven by interferon (IFN)-γ producing autoreactive CD4 + T cells independently of B cells and autoantiboodies. scRNA-seq analysis identifies increased Th1 responses and attenuated memory Treg function in PBMCs of patients with SjD. Conclusions: We report a more accurate mouse model of SjD while providing evidence for a critical role of Treg cells and IFN-γ producing Th1 cells in the pathogenesis of SjD, which may be effective targets for therapy.

2.
Nat Commun ; 14(1): 3611, 2023 06 17.
Article in English | MEDLINE | ID: mdl-37330549

ABSTRACT

Follicular helper T (Tfh) cells are essential for germinal center (GC) B cell responses. However, it is not clear which PD-1+CXCR5+Bcl6+CD4+ T cells will differentiate into PD-1hiCXCR5hiBcl6hi GC-Tfh cells and how GC-Tfh cell differentiation is regulated. Here, we report that the sustained Tigit expression in PD-1+CXCR5+CD4+ T cells marks the precursor Tfh (pre-Tfh) to GC-Tfh transition, whereas Tigit-PD-1+CXCR5+CD4+ T cells upregulate IL-7Rα to become CXCR5+CD4+ T memory cells with or without CCR7. We demonstrate that pre-Tfh cells undergo substantial further differentiation at the transcriptome and chromatin accessibility levels to become GC-Tfh cells. The transcription factor c-Maf appears critical in governing the pre-Tfh to GC-Tfh transition, and we identify Plekho1 as a stage-specific downstream factor regulating the GC-Tfh competitive fitness. In summary, our work identifies an important marker and regulatory mechanism of PD-1+CXCR5+CD4+ T cells during their developmental choice between memory T cell fate and GC-Tfh cell differentiation.


Subject(s)
T Follicular Helper Cells , T-Lymphocytes, Helper-Inducer , T-Lymphocytes, Helper-Inducer/metabolism , T Follicular Helper Cells/metabolism , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Germinal Center , Cell Differentiation , Receptors, CXCR5/genetics , Receptors, CXCR5/metabolism
3.
Elife ; 122023 02 21.
Article in English | MEDLINE | ID: mdl-36803766

ABSTRACT

The essential role of store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels in T cells is well established. In contrast, the contribution of individual Orai isoforms to SOCE and their downstream signaling functions in B cells are poorly understood. Here, we demonstrate changes in the expression of Orai isoforms in response to B cell activation. We show that both Orai3 and Orai1 mediate native CRAC channels in B cells. The combined loss of Orai1 and Orai3, but not Orai3 alone, impairs SOCE, proliferation and survival, nuclear factor of activated T cells (NFAT) activation, mitochondrial respiration, glycolysis, and the metabolic reprogramming of primary B cells in response to antigenic stimulation. Nevertheless, the combined deletion of Orai1 and Orai3 in B cells did not compromise humoral immunity to influenza A virus infection in mice, suggesting that other in vivo co-stimulatory signals can overcome the requirement of BCR-mediated CRAC channel function in B cells. Our results shed important new light on the physiological roles of Orai1 and Orai3 proteins in SOCE and the effector functions of B lymphocytes.


Subject(s)
B-Lymphocytes , Calcium Channels , ORAI1 Protein , Animals , Mice , B-Lymphocytes/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Calcium Signaling/physiology , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
4.
Sci Adv ; 8(40): eabn6552, 2022 10 07.
Article in English | MEDLINE | ID: mdl-36206339

ABSTRACT

T cell activation and function depend on Ca2+ signals mediated by store-operated Ca2+ entry (SOCE) through Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI1 proteins. We here investigated how SOCE controls T cell function in pulmonary inflammation during a T helper 1 (TH1) cell-mediated response to influenza A virus (IAV) infection and TH2 cell-mediated allergic airway inflammation. T cell-specific deletion of Orai1 did not exacerbate pulmonary inflammation and viral burdens following IAV infection but protected mice from house dust mite-induced allergic airway inflammation. ORAI1 controlled the expression of genes including p53 and E2F transcription factors that regulate the cell cycle in TH2 cells in response to allergen stimulation and the expression of transcription factors and cytokines that regulate TH2 cell function. Systemic application of a CRAC channel blocker suppressed allergic airway inflammation without compromising immunity to IAV infection, suggesting that inhibition of SOCE is a potential treatment for allergic airway disease.


Subject(s)
Calcium Channels , Influenza A virus , Allergens , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling , Cytokines/metabolism , E2F Transcription Factors , Inflammation , Mice , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53/metabolism
5.
EMBO Mol Med ; 14(9): e15687, 2022 09 07.
Article in English | MEDLINE | ID: mdl-35919953

ABSTRACT

Inflammatory bowel disease (IBD) is characterized by dysregulated intestinal immune responses. Using mass cytometry (CyTOF) to analyze the immune cell composition in the lamina propria (LP) of patients with ulcerative colitis (UC) and Crohn's disease (CD), we observed an enrichment of CD4+ effector T cells producing IL-17A and TNF, CD8+ T cells producing IFNγ, T regulatory (Treg) cells, and innate lymphoid cells (ILC). The function of these immune cells is regulated by store-operated Ca2+ entry (SOCE), which results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels formed by ORAI and STIM proteins. We observed that the pharmacologic inhibition of SOCE attenuated the production of proinflammatory cytokines including IL-2, IL-4, IL-6, IL-17A, TNF, and IFNγ by human colonic T cells and ILCs, reduced the production of IL-6 by B cells and the production of IFNγ by myeloid cells, but had no effect on the viability, differentiation, and function of intestinal epithelial cells. T cell-specific deletion of CRAC channel genes in mice showed that Orai1, Stim1, and Stim2-deficient T cells have quantitatively distinct defects in SOCE, which correlate with gradually more pronounced impairment of cytokine production by Th1 and Th17 cells and the severity of IBD. Moreover, the pharmacologic inhibition of SOCE with a selective CRAC channel inhibitor attenuated IBD severity and colitogenic T cell function in mice. Our data indicate that SOCE inhibition may be a suitable new approach for the treatment of IBD.


Subject(s)
Calcium Release Activated Calcium Channels , Inflammatory Bowel Diseases , Animals , CD8-Positive T-Lymphocytes/metabolism , Calcium/metabolism , Calcium Channels/metabolism , Humans , Immunity, Innate , Interleukin-17/metabolism , Interleukin-6/metabolism , Mice , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/genetics , Th17 Cells/metabolism
6.
J Gen Physiol ; 154(10)2022 10 03.
Article in English | MEDLINE | ID: mdl-35861698

ABSTRACT

Ca2+ signals regulate the function of many immune cells and promote immune responses to infection, cancer, and autoantigens. Ca2+ influx in immune cells is mediated by store-operated Ca2+ entry (SOCE) that results from the opening of Ca2+ release-activated Ca2+ (CRAC) channels. The CRAC channel is formed by three plasma membrane proteins, ORAI1, ORAI2, and ORAI3. Of these, ORAI1 is the best studied and plays important roles in immune function. By contrast, the physiological role of ORAI3 in immune cells remains elusive. We show here that ORAI3 is expressed in many immune cells including macrophages, B cells, and T cells. To investigate ORAI3 function in immune cells, we generated Orai3-/- mice. The development of lymphoid and myeloid cells in the thymus and bone marrow was normal in Orai3-/- mice, as was the composition of immune cells in secondary lymphoid organs. Deletion of Orai3 did not affect SOCE in B cells and T cells but moderately enhanced SOCE in macrophages. Orai3-deficient macrophages, B cells, and T cells had normal effector functions in vitro. Immune responses in vivo, including humoral immunity (T cell dependent or independent) and antitumor immunity, were normal in Orai3-/- mice. Moreover, Orai3-/- mice showed no differences in susceptibility to septic shock, experimental autoimmune encephalomyelitis, or collagen-induced arthritis. We conclude that despite its expression in myeloid and lymphoid cells, ORAI3 appears to be dispensable or redundant for physiological and pathological immune responses mediated by these cells.


Subject(s)
Calcium Channels , Calcium , Animals , Calcium/metabolism , Calcium Channels/genetics , Calcium Channels/metabolism , Calcium Signaling , Immunity , Lymphocytes/metabolism , Macrophages/metabolism , Mice , ORAI1 Protein/genetics , ORAI1 Protein/metabolism , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism
7.
Nat Immunol ; 23(2): 287-302, 2022 02.
Article in English | MEDLINE | ID: mdl-35105987

ABSTRACT

The volume-regulated anion channel (VRAC) is formed by LRRC8 proteins and is responsible for the regulatory volume decrease (RVD) after hypotonic cell swelling. Besides chloride, VRAC transports other molecules, for example, immunomodulatory cyclic dinucleotides (CDNs) including 2'3'cGAMP. Here, we identify LRRC8C as a critical component of VRAC in T cells, where its deletion abolishes VRAC currents and RVD. T cells of Lrrc8c-/- mice have increased cell cycle progression, proliferation, survival, Ca2+ influx and cytokine production-a phenotype associated with downmodulation of p53 signaling. Mechanistically, LRRC8C mediates the transport of 2'3'cGAMP in T cells, resulting in STING and p53 activation. Inhibition of STING recapitulates the phenotype of LRRC8C-deficient T cells, whereas overexpression of p53 inhibits their enhanced T cell function. Lrrc8c-/- mice have exacerbated T cell-dependent immune responses, including immunity to influenza A virus infection and experimental autoimmune encephalomyelitis. Our results identify cGAMP uptake through LRRC8C and STING-p53 signaling as a new inhibitory signaling pathway in T cells and adaptive immunity.


Subject(s)
Anions/metabolism , Dinucleoside Phosphates/metabolism , Ion Channels/metabolism , Membrane Proteins/metabolism , T-Lymphocytes/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Calcium/metabolism , Female , Mice , Mice, Inbred C57BL , Nucleotides, Cyclic/metabolism , Signal Transduction/physiology
8.
Cell Calcium ; 90: 102227, 2020 09.
Article in English | MEDLINE | ID: mdl-32563861

ABSTRACT

B lymphocytes are an important component of the adaptive and innate immune system because of their ability to secrete antibodies and to present antigens to T cells, which is critical for immune responses to many pathogens. Abnormal B cell function is the cause of diseases including autoimmune, paraneoplastic, and immunodeficiency disorders. The development, survival, and function of B cells depend on signaling through the B cell receptor (BCR) and costimulatory receptors. One of the signaling pathways induced by antigen binding to the BCR is store-operated Ca2+ entry (SOCE), which depends on the Ca2+ channel ORAI1 and its activators stromal interaction molecule (STIM) 1 and 2. A recent study by Berry et al. [1] reports that B cells lacking STIM1 and STIM2 fail to survive and proliferate because abolished SOCE results in impaired expression of two key anti-apoptotic genes and blunted activation of mTORC1 and c-Myc signaling. The associated Ca2+ regulated checkpoints of B cell survival and proliferation can be bypassed, at least partially, by costimulation through CD40 or TLR9. This study provides important new insights on how SOCE controls B cell function.


Subject(s)
B-Lymphocytes/metabolism , Calcium/metabolism , Animals , B-Lymphocytes/cytology , Calcium Signaling , Cell Proliferation , Cell Survival , Humans , Immunity, Humoral , Mice
9.
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
10.
J Immunol ; 202(10): 2991-2998, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30971440

ABSTRACT

T follicular helper (Tfh) cells are essential for germinal center B cell responses. The molecular mechanism underlying the initial Tfh cell differentiation, however, is still incompletely understood. In this study, we show that in vivo, despite enhanced non-Tfh cell effector functions, the deletion of transcription factor Bach2 results in preferential Tfh cell differentiation. Mechanistically, the deletion of Bach2 leads to the induction of CXCR5 expression even before the upregulation of Ascl2. Subsequently, we have identified a novel regulatory element in the murine CXCR5 locus that negatively regulates CXCR5 promoter activities in a Bach2-dependent manner. Bach2 deficiency eventually results in a collapsed CD4+ T cell response with severely impaired CD4+ T cell memory, including Tfh cell memory. Our results demonstrate that Bach2 critically regulates Tfh cell differentiation and CD4+ T cell memory.


Subject(s)
Basic-Leucine Zipper Transcription Factors/immunology , Cell Differentiation/immunology , Immunologic Memory , T-Lymphocytes, Helper-Inducer/immunology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/immunology , Basic-Leucine Zipper Transcription Factors/genetics , Cell Differentiation/genetics , Gene Expression Regulation/immunology , Mice , Mice, Transgenic , Receptors, CXCR5/genetics , Receptors, CXCR5/immunology , T-Lymphocytes, Helper-Inducer/cytology
11.
Nat Commun ; 10(1): 1183, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862784

ABSTRACT

T regulatory (Treg) cells maintain immunological tolerance and organ homeostasis. Activated Treg cells differentiate into effector Treg subsets that acquire tissue-specific functions. Ca2+ influx via Ca2+ release-activated Ca2+ (CRAC) channels formed by STIM and ORAI proteins is required for the thymic development of Treg cells, but its function in mature Treg cells remains unclear. Here we show that deletion of Stim1 and Stim2 genes in mature Treg cells abolishes Ca2+ signaling and prevents their differentiation into follicular Treg and tissue-resident Treg cells. Transcriptional profiling of STIM1/STIM2-deficient Treg cells reveals that Ca2+ signaling regulates transcription factors and signaling pathways that control the identity and effector differentiation of Treg cells. In the absence of STIM1/STIM2 in Treg cells, mice develop a broad spectrum of autoantibodies and fatal multiorgan inflammation. Our findings establish a critical role of CRAC channels in controlling lineage identity and effector functions of Treg cells.


Subject(s)
Autoimmune Diseases/immunology , Calcium Release Activated Calcium Channels/physiology , Calcium/metabolism , Cell Differentiation/immunology , T-Lymphocytes, Regulatory/physiology , Animals , Autoantibodies/immunology , Autoimmune Diseases/genetics , Bone Marrow Transplantation , Cations, Divalent/metabolism , Disease Models, Animal , Female , Forkhead Transcription Factors/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Stromal Interaction Molecule 1/genetics , Stromal Interaction Molecule 1/metabolism , Stromal Interaction Molecule 2/genetics , Stromal Interaction Molecule 2/metabolism , Transplantation Chimera
12.
J Immunol ; 200(2): 586-594, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29212910

ABSTRACT

T follicular helper (Tfh) cells play an essential role in the formation of germinal centers (GC) and generation of high-affinity Abs. The homing of activated CD4+ T cells into B cell follicles and the involvement of key costimulatory and coinhibitory molecules are critical in controlling both the initiation and the magnitude of GC responses. Meanwhile, studies have shown that a high number of single clone B cells leads to intraclonal competition, which inhibits the generation of high-affinity Abs. Our previous work has shown that transcription factor Foxp1 is a critical negative regulator of Tfh cell differentiation. In this study, we report that the deletion of Foxp1 leads to a high proportion of activated CD4+ T cells homing into B cell follicles with faster kinetics, resulting in earlier GC formation. In addition, we show that Foxp1-deficient Tfh cells restore the generation of high-affinity Abs when cotransferred with high numbers of single clone B cells. We find that Foxp1 regulates the expression levels of cytotoxic T lymphocyte-associated Ag-4 (CTLA-4) in activated CD4+ T cells and that Ctla4 is a direct Foxp1 target. Finally, we demonstrate that CTLA-4 expression on conventional CD4+ T cells plays a cell-intrinsic role in Tfh cell differentiation in vivo, and CTLA-4 blockade helps abolish the intraclonal competition of B cells in generating high-affinity Abs.


Subject(s)
CTLA-4 Antigen/metabolism , Cell Differentiation/immunology , Cell Movement/immunology , Forkhead Transcription Factors/metabolism , Germinal Center/immunology , Repressor Proteins/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CTLA-4 Antigen/genetics , Immunomodulation , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Mice, Transgenic , T-Lymphocytes, Helper-Inducer/cytology
13.
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
14.
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
15.
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
16.
J Immunol ; 196(9): 3537-41, 2016 05 01.
Article in English | MEDLINE | ID: mdl-27001958

ABSTRACT

Previously we have shown that transcription factor Foxp1 plays an essential role in maintaining naive T cell quiescence; in the absence of Foxp1, mature naive CD8(+) T cells proliferate in direct response to homeostatic cytokine IL-7. In this study, we report that the deletion of Foxp1 in naive CD8(+) T cells leads to enhanced activation of the PI3K/Akt/mammalian target of rapamycin signaling pathway and its downstream cell growth and metabolism targets in response to IL-7. We found that Foxp1 directly regulates PI3K interacting protein 1, a negative regulator of PI3K. Additionally, we found that deletion of Foxp1 in naive CD8(+) T cells results in increased expression levels of E2fs, the critical components for cell cycle progression and proliferation, in a manner that is not associated with increased phosphorylation of retinoblastoma protein. Taken together, our studies suggest that Foxp1 enforces naive CD8(+) T cell quiescence by simultaneously repressing key pathways in both cellular metabolism and cell cycle progression.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Cycle , Forkhead Transcription Factors/metabolism , Gene Expression Regulation , Interleukin-7/metabolism , Repressor Proteins/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Cycle/physiology , Cell Proliferation , Forkhead Transcription Factors/deficiency , Forkhead Transcription Factors/genetics , Homeostasis , Interleukin-7/immunology , Interleukin-7/pharmacology , Intracellular Signaling Peptides and Proteins , Membrane Proteins , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Repressor Proteins/deficiency , Repressor Proteins/genetics , Retinoblastoma Protein/immunology , Retinoblastoma Protein/metabolism , Signal Transduction , TOR Serine-Threonine Kinases/metabolism
17.
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
18.
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
19.
J Autoimmun ; 59: 26-37, 2015 May.
Article in English | MEDLINE | ID: mdl-25701076

ABSTRACT

CD4(+)Foxp3(+) regulatory T cells (Tregs) play a non-redundant role in control of excessive immune responses, and defects in Tregs have been shown both in patients and murine models of primary biliary cirrhosis (PBC), a progressive autoimmune biliary disease. Herein, we took advantage of a murine model of PBC, the dominant negative transforming growth factor ß receptor II (dnTGFßRII) mice, to assess Treg genetic defects and their functional effects in PBC. By using high-resolution microarrays with verification by PCR and protein expression, we found profound and wide-ranging differences between dnTGFßRII and normal, wild type Tregs. Critical transcription factors were down-regulated including Eos, Ahr, Klf2, Foxp1 in dnTGFßRII Tregs. Functionally, dnTGFßRII Tregs expressed an activated, pro-inflammatory phenotype with upregulation of Ccl5, Granzyme B and IFN-γ. Genetic pathway analysis suggested that the primary effect of loss of TGFß pathway signaling was to down regulate immune regulatory processes, with a secondary upregulation of inflammatory processes. These findings provide new insights into T regulatory genetic defects; aberrations of the identified genes or genetic pathways should be investigated in human PBC Tregs. This approach which takes advantage of biologic pathway analysis illustrates the ability to identify genes/pathways that are affected both independently and dependent on abnormalities in TGFß signaling. Such approaches will become increasingly useful in human autoimmunity.


Subject(s)
Liver Cirrhosis, Biliary/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Disease Models, Animal , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation/genetics , Granzymes/metabolism , Humans , Inflammation Mediators/metabolism , Interferon-gamma/metabolism , Liver Cirrhosis, Biliary/genetics , Mice , Mice, Mutant Strains , Microarray Analysis , Mutation/genetics , Protein Serine-Threonine Kinases/genetics , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/genetics , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism
20.
Autoimmun Rev ; 14(3): 183-91, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25315744

ABSTRACT

The intestinal microbiome plays a significant role in the development of autoimmune diseases, in particular, inflammatory bowel diseases. But the interplay between the intestinal tract and the liver may explain the increased association with autoimmune liver diseases and inflammatory bowel diseases. The gut-liver axis involves multiple inflammatory cell types and cytokines, chemokines and other molecules which lead to the destruction of normal liver architecture. Triggers for the initiation of these events are unclear, but appear to include multiple environmental factors, including pathogenic or even commensal microbial agents. The variation in the gut microbiome has been cited as a major factor in the pathogenesis of autoimmune liver disease and even other autoimmune diseases. The unique positioning of the liver at the juncture of the peripheral circulation and the portal circulation augments the interaction between naïve T cells and other hepatic cells and leads to the disruption in the development of tolerance to commensal bacteria and other environmental agents. Finally, the innate immune system and in particular toll-like receptors play a significant role in the pathogenesis of autoimmune liver disease.


Subject(s)
Intestines/microbiology , Microbiota , Animals , B-Lymphocytes/immunology , Humans , Immunity, Innate , Intestines/immunology , Liver/immunology , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...