Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 40
Filter
1.
Clin Immunol ; 262: 110168, 2024 May.
Article in English | MEDLINE | ID: mdl-38458301

ABSTRACT

Systemic lupus erythematosus (SLE) is a complex autoimmune disorder impacting various organs, notably prevalent in women of reproductive age. This review explores the involvement of a disintegrin and metalloproteinases (ADAMs) in SLE pathogenesis. Despite advancements in understanding SLE through genome and transcriptome studies, the role of ADAMs in post-translational regulations remains insufficiently explored. ADAMs, transmembrane proteins with diverse functions, impact cell adhesion, migration, and inflammation by shedding cell surface proteins, growth factors, and receptors. Notably, ADAM9 is implicated in Th17 cell differentiation, which is crucial in SLE pathology. ADAM10 and ADAM17 play pivotal roles in T-cell biology, influencing immune cell development and differentiation. Elevated soluble ADAM substrates in SLE patients serve as potential biomarkers correlating with disease activity. Targeting ADAMs or their substrates offers promising therapeutic avenues for SLE management and treatment enhancement.


Subject(s)
Disintegrins , Lupus Erythematosus, Systemic , Humans , Female , Disintegrins/metabolism , ADAM10 Protein/metabolism , Inflammation , Cell Differentiation , Membrane Proteins , ADAM Proteins
2.
J Immunol ; 209(3): 621-628, 2022 08 01.
Article in English | MEDLINE | ID: mdl-35831019

ABSTRACT

We had shown previously that the protein phosphatase 2A regulatory subunit PPP2R2D suppresses IL-2 production, and PPP2R2D deficiency in T cells potentiates the suppressive function of regulatory T (Treg) cells and alleviates imiquimod-induced lupus-like pathology. In this study, in a melanoma xenograft model, we noted that the tumor grew in larger sizes in mice lacking PPP2R2D in T cells (LckCreR2Dfl/fl) compared with wild type (R2Dfl/fl) mice. The numbers of intratumoral T cells in LckCreR2Dfl/fl mice were reduced compared with R2Dfl/fl mice, and they expressed a PD-1+CD3+CD44+ exhaustion phenotype. In vitro experiments confirmed that the chromatin of exhaustion markers PD-1, LAG3, TIM3, and CTLA4 remained open in LckCreR2Dfl/fl CD4 T conventional compared with R2Dfl/fl T conventional cells. Moreover, the percentage of Treg cells (CD3+CD4+Foxp3+CD25hi) was significantly increased in the xenografted tumor of LckCreR2Dfl/fl mice compared with R2Dfl/fl mice probably because of the increase in the percentage of IL-2-producing LckCreR2Dfl/fl T cells. Moreover, using adoptive T cell transfer in mice xenografted with melanoma, we demonstrated that PPP2R2D deficiency in T cells enhanced the inhibitory effect of Treg cells in antitumor immunity. At the translational level, analysis of publicly available data from 418 patients with melanoma revealed that PPP2R2D expression levels correlated positively with tumor-infiltration level of CD4 and CD8 T cells. The data demonstrate that PPP2R2D is a negative regulator of immune checkpoint receptors, and its absence exacerbates effector T cell exhaustion and promotes Treg cell expansion. We conclude that PPP2R2D protects against melanoma growth, and PPP2R2D-promoting regimens can have therapeutic value in patients with melanoma.


Subject(s)
Melanoma , T-Lymphocytes, Regulatory , Animals , Cell Proliferation , Humans , Interleukin-2/metabolism , Melanoma/metabolism , Mice , Programmed Cell Death 1 Receptor/metabolism , Protein Phosphatase 2/metabolism
3.
Proc Natl Acad Sci U S A ; 118(18)2021 05 04.
Article in English | MEDLINE | ID: mdl-33911034

ABSTRACT

The a disintegrin and metalloproteinase (ADAM) family of proteinases alter the extracellular environment and are involved in the development of T cells and autoimmunity. The role of ADAM family members in Th17 cell differentiation is unknown. We identified ADAM9 to be specifically expressed and to promote Th17 differentiation. Mechanistically, we found that ADAM9 cleaved the latency-associated peptide to produce bioactive transforming growth factor ß1, which promoted SMAD2/3 phosphorylation and activation. A transcription factor inducible cAMP early repressor was found to bind directly to the ADAM9 promoter and to promote its transcription. Adam9-deficient mice displayed mitigated experimental autoimmune encephalomyelitis, and transfer of Adam9-deficient myelin oligodendrocyte globulin-specific T cells into Rag1-/- mice failed to induce disease. At the translational level, an increased abundance of ADAM9 levels was observed in CD4+ T cells from patients with systemic lupus erythematosus, and ADAM9 gene deletion in lupus primary CD4+ T cells clearly attenuated their ability to differentiate into Th17 cells. These findings revealed that ADAM9 as a proteinase provides Th17 cells with an ability to activate transforming growth factor ß1 and accelerates its differentiation, resulting in aberrant autoimmunity.


Subject(s)
ADAM Proteins/genetics , Autoimmunity/genetics , Homeodomain Proteins/genetics , Membrane Proteins/genetics , T-Lymphocytes/immunology , Transforming Growth Factor beta1/genetics , Adult , Animals , Autoimmunity/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation/genetics , Cyclic AMP/genetics , Female , Humans , Lupus Erythematosus, Systemic , Male , Mice , Middle Aged , Myelin Sheath/genetics , Oligodendroglia/metabolism , Phosphorylation/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , T-Lymphocytes/pathology , Th17 Cells/immunology , Young Adult
4.
Rheumatology (Oxford) ; 62(2): 861-871, 2023 02 01.
Article in English | MEDLINE | ID: mdl-35781320

ABSTRACT

OBJECTIVE: To investigate the role of calcium/calmodulin-dependent protein kinase IV (CaMK4) in the development of joint injury in a mouse model of arthritis and patients with RA. METHODS: Camk4-deficient, Camk4flox/floxLck-Cre, and mice treated with CaMK4 inhibitor KN-93 or KN-93 encapsulated in nanoparticles tagged with CD4 or CD8 antibodies were subjected to collagen-induced arthritis (CIA). Inflammatory cytokine levels, humoral immune response, synovitis, and T-cell activation were recorded. CAMK4 gene expression was measured in CD4+ T cells from healthy participants and patients with active RA. Micro-CT and histology were used to assess joint pathology. CD4+ and CD14+ cells in patients with RA were subjected to Th17 or osteoclast differentiation, respectively. RESULTS: CaMK4-deficient mice subjected to CIA displayed improved clinical scores and decreased numbers of Th17 cells. KN-93 treatment significantly reduced joint destruction by decreasing the production of inflammatory cytokines. Furthermore, Camk4flox/floxLck-Cre mice and mice treated with KN93-loaded CD4 antibody-tagged nanoparticles developed fewer Th17 cells and less severe arthritis. CaMK4 inhibition mitigated IL-17 production by CD4+ cells in patients with RA. The number of in vitro differentiated osteoclasts from CD14+ cells in patients with RA was significantly decreased with CaMK4 inhibitors. CONCLUSION: Using global and CD4-cell-targeted pharmacologic approaches and conditionally deficient mice, we demonstrate that CaMK4 is important in the development of arthritis. Using ex vivo cell cultures from patients with RA, CaMK4 is important for both Th17 generation and osteoclastogenesis. We propose that CaMK4 inhibition represents a new approach to control the development of arthritis.


Subject(s)
Arthritis, Experimental , Osteogenesis , Animals , Mice , Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Calcium/therapeutic use , Th17 Cells , Cytokines/metabolism , Arthritis, Experimental/metabolism , Cell Differentiation
5.
J Immunol ; 206(8): 1719-1728, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33762326

ABSTRACT

Protein phosphatase 2A (PP2A) composed of a scaffold subunit, a catalytic subunit, and multiple regulatory subunits is a ubiquitously expressed serine/threonine phosphatase. We have previously shown that the PP2A catalytic subunit is increased in T cells from patients with systemic lupus erythematosus and promotes IL-17 production by enhancing the activity of Rho-associated kinase (ROCK) in T cells. However, the molecular mechanism whereby PP2A regulates ROCK activity is unknown. In this study, we show that the PP2A regulatory subunit PPP2R2A is increased in T cells from people with systemic lupus erythematosus and binds to, dephosphorylates, and activates the guanine nucleotide exchange factor GEF-H1 at Ser885, which in turn increases the levels of RhoA-GTP and the activity of ROCK in T cells. Genetic PPP2R2A deficiency in murine T cells reduced Th1 and Th17, but not regulatory T cell differentiation and mice with T cell-specific PPP2R2A deficiency displayed less autoimmunity when immunized with myelin oligodendrocyte glycoprotein peptide. Our studies indicate that PPP2R2A is the regulatory subunit that dictates the PP2A-directed enhanced Th1 and Th17 differentiation, and therefore, it represents a therapeutic target for pathologies linked to Th1 and Th17 cell expansion.


Subject(s)
Carboxylic Ester Hydrolases/metabolism , Lupus Erythematosus, Systemic/metabolism , Protein Phosphatase 2/metabolism , Th1 Cells/immunology , Th17 Cells/immunology , Animals , Carboxylic Ester Hydrolases/genetics , Cell Differentiation , Cells, Cultured , Gene Expression Regulation , Humans , Lupus Erythematosus, Systemic/genetics , Lymphocyte Activation , Mice , Mice, Knockout , Protein Phosphatase 2/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , rho-Associated Kinases/metabolism , rhoA GTP-Binding Protein/metabolism
6.
J Immunol ; 207(1): 55-64, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34135066

ABSTRACT

Effector CD4+ T lymphocytes contribute to inflammation and tissue damage in psoriasis, but the underlying molecular mechanisms remain poorly understood. The transcription factor CREMα controls effector T cell function in people with systemic autoimmune diseases. The inhibitory surface coreceptor PD-1 plays a key role in the control of effector T cell function and its therapeutic inhibition in patients with cancer can cause psoriasis. In this study, we show that CD4+ T cells from patients with psoriasis and psoriatic arthritis exhibit increased production of IL-17 but decreased expression of IL-2 and PD-1. In genetically modified mice and Jurkat T cells CREMα expression was linked to low PD-1 levels. We demonstrate that CREMα is recruited to the proximal promoter of PDCD1 in which it trans-represses gene expression and corecruits DNMT3a-mediating DNA methylation. As keratinocytes limit inflammation by PD-1 ligand expression and, in this study, reported reduced expression of PD-1 on CD4+ T cells is linked to low IL-2 and high IL-17A production, our studies reveal a molecular pathway in T cells from people with psoriasis that can deserve clinical exploitation.


Subject(s)
Arthritis, Psoriatic/immunology , CD4-Positive T-Lymphocytes/immunology , Cyclic AMP Response Element Modulator/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Humans , Mice , Mice, Inbred C57BL
7.
Curr Opin Rheumatol ; 33(2): 181-189, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33332887

ABSTRACT

PURPOSE OF REVIEW: This review gives an overview of the recently published clinical trials in systemic lupus erythematosus (SLE). RECENT FINDINGS: Our continuously improving understanding of the cellular and molecular mechanisms, which are involved in the pathogenesis of SLE, has inspired the performance of multiple clinical trials in an attempt to modify recognized targets. Here, we summarize results obtained from recent trials, which used monoclonal antibodies blocking cytokines, blockers of costimulatory molecules or deleting immune cells, small drug inhibitors of kinases and replenishment of cytokines. SUMMARY: The therapeutic options for patients with SLE grow continuously and in parallel it raises the need for pathogenetic mechanism-based precision medicine so that we may select the right treatment for the right patient.


Subject(s)
Lupus Erythematosus, Systemic , Antibodies, Monoclonal/therapeutic use , Cytokines , Humans , Lupus Erythematosus, Systemic/drug therapy
8.
Proc Natl Acad Sci U S A ; 115(10): 2478-2483, 2018 03 06.
Article in English | MEDLINE | ID: mdl-29463741

ABSTRACT

Glutaminolysis is a well-known source of energy for effector T cells but its contribution to each T cell subset and the mechanisms which are responsible for the control of involved metabolic enzymes are not fully understood. We report that Th17 but not Th1, Th2, or Treg cell induction in vitro depends on glutaminolysis and the up-regulation of glutaminase 1 (Gls1), the first enzyme in the glutaminolysis pathway. Both pharmacological and siRNA-based selective inhibition of Gls1 reduced in vitro Th17 differentiation and reduced the CD3/TCR-mediated increase of the mammalian target of rapamycin complex 1 activity. Treatment of mice with a Gls1 inhibitor ameliorated experimental autoimmune encephalomyelitis. Furthermore, RAG1-deficient mice that received Gls1-shRNA-transfected 2D2 T cells had reduced experimental autoimmune encephalomyelitis scores compared with those that received control-shRNA-treated cells. Next we found that T cells deficient in inducible cAMP early repressor (ICER), a transcriptional factor known to promote Th17 differentiation, display reduced activity of oxidative phosphorylation rates in the presence of glutamine and reduced Gls1 expression, both of which could be restored by ICER overexpression. Finally, we demonstrate that ICER binds to the gls1 promoter directly and increases its activity. These findings demonstrate the importance of glutaminolysis in the generation of Th17 and the direct control of Gls1 activity by the IL-17-promoting transcription factor ICER. Pharmaceutical modulation of the glutaminolysis pathway should be considered to control Th17-mediated pathology.


Subject(s)
Cyclic AMP Response Element Modulator , Glutaminase , Th17 Cells , Animals , Autoimmunity , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/metabolism , Encephalomyelitis, Autoimmune, Experimental/immunology , Glutaminase/antagonists & inhibitors , Glutaminase/metabolism , Glutamine/metabolism , Mice , Mice, Transgenic , Th17 Cells/cytology , Th17 Cells/immunology , Th17 Cells/metabolism
9.
Proc Natl Acad Sci U S A ; 115(37): 9288-9293, 2018 09 11.
Article in English | MEDLINE | ID: mdl-30150402

ABSTRACT

Th17 cells favor glycolytic metabolism, and pyruvate dehydrogenase (PDH) is the key bifurcation enzyme, which in its active dephosphorylated form advances the oxidative phosphorylation from glycolytic pathway. The transcriptional factor, inducible cAMP early repressor/cAMP response element modulator (ICER/CREM), has been shown to be induced in Th17 cells and to be overexpressed in CD4+ T cells from the patients with systemic lupus erythematosus (SLE). We found that glycolysis and lactate production in in vitro Th17-polarized T cells was reduced and that the expression of pyruvate dehydrogenase phosphatase catalytic subunit 2 (PDP2), an enzyme that converts the inactive PDH to its active form, and PDH enzyme activity were increased in Th17 cells from ICER/CREM-deficient animals. ICER was found to bind to the Pdp2 promoter and suppress its expression. Furthermore, forced expression of PDP2 in CD4+ cells reduced the in vitro Th17 differentiation, whereas shRNA-based suppression of PDP2 expression increased in vitro Th17 differentiation and augmented experimental autoimmune encephalomyelitis. At the translational level, PDP2 expression was decreased in memory Th17 cells from patients with SLE and forced expression of PDP2 in CD4+ T cells from lupus-prone MRL/lpr mice and patients with SLE suppressed Th17 differentiation. These data demonstrate the direct control of energy production during Th17 differentiation in health and disease by the transcription factor ICER/CREM at the PDH metabolism bifurcation level.


Subject(s)
Cell Differentiation , Gene Expression Regulation, Enzymologic , Phosphoprotein Phosphatases/biosynthesis , Response Elements , Th17 Cells/enzymology , Animals , Catalytic Domain , Cyclic AMP Response Element Modulator/genetics , Cyclic AMP Response Element Modulator/immunology , Cyclic AMP Response Element Modulator/metabolism , Encephalomyelitis, Autoimmune, Experimental/enzymology , Encephalomyelitis, Autoimmune, Experimental/genetics , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/pathology , Female , Humans , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/genetics , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Male , Mice , Mice, Knockout , Phosphoprotein Phosphatases/genetics , Phosphoprotein Phosphatases/immunology , Th17 Cells/immunology , Th17 Cells/pathology
10.
Curr Opin Rheumatol ; 32(2): 192-199, 2020 03.
Article in English | MEDLINE | ID: mdl-31842032

ABSTRACT

PURPOSE OF REVIEW: Th1, Th17, and Treg cells play distinct roles in autoimmune diseases, including systemic lupus erythematosus, multiple sclerosis, and rheumatoid arthritis. During the last 5 years we have learned that T-cell metabolism affects cell survival, differentiation and fate of T cells. RECENT FINDINGS: We highlight recent studies which have reported on T-cell metabolism in autoimmune diseases, differences in cellular metabolisms in T-cell subsets among various diseases and transcription factors which control the expression and function of central metabolic enzymes. SUMMARY: Distinct metabolic processes control the function of T-cell subsets in autoimmune disease and known transcription factors control the activity of metabolic enzymes. The revealed insights into the metabolic events of immune cells offer opportunities for new therapeutic approaches.


Subject(s)
Autoimmune Diseases/metabolism , Autoimmunity/physiology , T-Lymphocytes/metabolism , Animals , Autoimmune Diseases/immunology , Cell Differentiation/immunology , Humans , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes/immunology
11.
J Immunol ; 198(11): 4268-4276, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28438897

ABSTRACT

Systemic lupus erythematosus (SLE) damages multiple organs by producing various autoantibodies. In this study, we report that decreased microRNA (miR)-200a-3p causes IL-2 hypoproduction through zinc finger E-box binding homeobox (ZEB)1 and C-terminal binding protein 2 (CtBP2) in a lupus-prone mouse. First, we performed RNA sequencing to identify candidate microRNAs and mRNAs involved in the pathogenesis of SLE. We found that miR-200a-3p was significantly downregulated, whereas its putative targets, ZEB2 and CtBP2, were upregulated in CD4+ T cells from MRL/lpr-Tnfrsf6lpr mice compared with C57BL/6J mice. ZEB1 and ZEB2 comprise the ZEB family and suppress various genes, including IL-2 by recruiting CtBP2. IL-2 plays a critical role in immune tolerance, and insufficient IL-2 production upon stimulation has been recognized in SLE pathogenesis. Therefore, we hypothesized that decreased miR-200a-3p causes IL-2 deficit through the ZEB1-CtBP2 and/or ZEB2-CtBP2 complex in SLE CD4+ T cells. Overexpression of miR-200a-3p induced IL-2 production by downregulating ZEB1, ZEB2, and CtBP2 in EL4 cell lines. We further revealed that miR-200a-3p promotes IL-2 expression by reducing the binding of suppressive ZEB1-CtBP2 and ZEB2-CtBP2 complexes on negative regulatory element A in the IL-2 promoter in EL4 cells. Interestingly, the ZEB1-CtBP2 complex on negative regulatory element A was significantly upregulated after PMA/ionomycin stimulation in lupus CD4+ T cells. Our studies have revealed a new epigenetic pathway in the control of IL-2 production in SLE whereby low levels of miR-200a-3p accumulate the binding of the ZEB1-CtBP2 complex to the IL-2 promoter and suppress IL-2 production.


Subject(s)
DNA-Binding Proteins/genetics , Down-Regulation , Interleukin-2/biosynthesis , Interleukin-2/genetics , Lupus Erythematosus, Systemic/immunology , MicroRNAs/genetics , Phosphoproteins/genetics , T-Lymphocytes/immunology , Alcohol Oxidoreductases , Animals , Cell Line , Co-Repressor Proteins , DNA-Binding Proteins/metabolism , Interleukin-2/immunology , Lupus Erythematosus, Systemic/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Phosphoproteins/metabolism , T-Lymphocytes/pathology , Transcriptional Activation , Zinc Finger E-box-Binding Homeobox 1/metabolism
12.
Proc Natl Acad Sci U S A ; 113(33): 9321-6, 2016 08 16.
Article in English | MEDLINE | ID: mdl-27482100

ABSTRACT

Signaling lymphocytic activation molecule family 3 (SLAMF3/Ly9) is a coregulatory molecule implicated in T-cell activation and differentiation. Systemic lupus erythematosus (SLE) is characterized by aberrant T-cell activation and compromised IL-2 production, leading to abnormal regulatory T-cell (Treg) development/function. Here we show that SLAMF3 functions as a costimulator on CD4(+) T cells and influences IL-2 response and T helper cell differentiation. SLAMF3 ligation promotes T-cell responses to IL-2 via up-regulation of CD25 in a small mothers against decapentaplegic homolog 3 (Smad3)-dependent mechanism. This augments the activation of the IL-2/IL-2R/STAT5 pathway and enhances cell proliferation in response to exogenous IL-2. SLAMF3 costimulation promotes Treg differentiation from naïve CD4(+) T cells. Ligation of SLAMF3 receptors on SLE CD4(+) T cells restores IL-2 responses to levels comparable to those seen in healthy controls and promotes functional Treg generation. Taken together, our results suggest that SLAMF3 acts as potential therapeutic target in SLE patients by augmenting sensitivity to IL-2.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Interleukin-2/pharmacology , Lupus Erythematosus, Systemic/immunology , Signaling Lymphocytic Activation Molecule Family/physiology , T-Lymphocytes, Regulatory/physiology , Adult , Aged , CD4-Positive T-Lymphocytes/immunology , Cell Differentiation , Cell Polarity , Female , Humans , Interleukin-2/biosynthesis , Interleukin-2 Receptor alpha Subunit/analysis , Interleukin-2 Receptor alpha Subunit/genetics , Male , Middle Aged
13.
J Immunol ; 195(12): 5533-7, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26561550

ABSTRACT

Treatment of autoimmune diseases is still largely based on the use of systemically acting immunosuppressive drugs, which invariably cause severe side effects. Calcium/calmodulin-dependent protein kinase IV is involved in the suppression of IL-2 and the production of IL-17. Its pharmacologic or genetic inhibition limits autoimmune disease in mice. In this study, we demonstrate that KN93, a small-molecule inhibitor of calcium/calmodulin-dependent protein kinase IV, targeted to CD4(+) T cells via a nanolipogel delivery system, markedly reduced experimental autoimmune encephalomyelitis and was 10-fold more potent than the free systemically delivered drug in the lupus mouse models. The targeted delivery of KN93 did not deplete T cells but effectively blocked Th17 cell differentiation and expansion as measured in the spinal cords and kidneys of mice developing experimental autoimmune encephalomyelitis or lupus, respectively. These results highlight the promise of cell-targeted inhibition of molecules involved in the pathogenesis of autoimmunity as a means of advancing the treatment of autoimmune diseases.


Subject(s)
Benzylamines/administration & dosage , CD4-Positive T-Lymphocytes/drug effects , Encephalomyelitis, Autoimmune, Experimental/drug therapy , Lupus Erythematosus, Systemic/drug therapy , Polyethylene Glycols/administration & dosage , Polyethyleneimine/administration & dosage , Sulfonamides/administration & dosage , Th17 Cells/drug effects , Animals , Benzylamines/pharmacology , CD4-Positive T-Lymphocytes/immunology , Calcium-Calmodulin-Dependent Protein Kinase Type 4/antagonists & inhibitors , Cell Differentiation/drug effects , Cells, Cultured , Encephalomyelitis, Autoimmune, Experimental/immunology , Humans , Immunosuppression Therapy , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred C57BL , Mice, Inbred MRL lpr , Nanogels , Sulfonamides/pharmacology , Th17 Cells/immunology
14.
J Immunol ; 193(5): 2168-77, 2014 Sep 01.
Article in English | MEDLINE | ID: mdl-25063876

ABSTRACT

IL-2, a cytokine with pleiotropic effects, is critical for immune cell activation and peripheral tolerance. Although the therapeutic potential of IL-2 has been previously suggested in autoimmune diseases, the mechanisms whereby IL-2 mitigates autoimmunity and prevents organ damage remain unclear. Using an inducible recombinant adeno-associated virus vector, we investigated the effect of low systemic levels of IL-2 in lupus-prone MRL/Fas(lpr/lpr) (MRL/lpr) mice. Treatment of mice after the onset of disease with IL-2-recombinant adeno-associated virus resulted in reduced mononuclear cell infiltration and pathology of various tissues, including skin, lungs, and kidneys. In parallel, we noted a significant decrease of IL-17-producing CD3(+)CD4(-)CD8(-) double-negative T cells and an increase in CD4(+)CD25(+)Foxp3(+) immunoregulatory T cells (Treg) in the periphery. We also show that IL-2 can drive double-negative (DN) T cell death through an indirect mechanism. Notably, targeted delivery of IL-2 to CD122(+) cytotoxic lymphocytes effectively reduced the number of DN T cells and lymphadenopathy, whereas selective expansion of Treg by IL-2 had no effect on DN T cells. Collectively, our data suggest that administration of IL-2 to lupus-prone mice protects against end-organ damage and suppresses inflammation by dually limiting IL-17-producing DN T cells and expanding Treg.


Subject(s)
Antineoplastic Agents/pharmacology , CD4 Antigens , CD8 Antigens , Interleukin-2/pharmacology , Lupus Erythematosus, Systemic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Female , Interleukin-17/immunology , Interleukin-2 Receptor beta Subunit/immunology , Lupus Erythematosus, Systemic/pathology , Mice , Mice, Inbred MRL lpr , T-Lymphocytes, Regulatory/pathology
16.
J Biol Chem ; 288(2): 826-36, 2013 Jan 11.
Article in English | MEDLINE | ID: mdl-23209284

ABSTRACT

Somatic mutations accumulate in senescent cells. Bcl6, which functions as a transcriptional repressor, has been identified as a potent inhibitor of cell senescence, but a role of Bcl6 in the accumulation of somatic mutations has remained unclear. Ig class-switch recombination simultaneously induces somatic mutations in an IgM class-switch (Ig-Sµ) region of IgG B cells. Surprisingly, mutations were detected in the Ig-Sµ region of Bcl6-deficient IgM B cells without class-switch recombination, and these mutations were mainly generated by conversion of adenosine to guanosine, suggesting a novel DNA mutator in the B cells. The ADAR1 (adenosine deaminase acting on RNA1) gene was overexpressed in Bcl6-deficient cells, and its promoter analysis revealed that ADAR1 is a molecular target of Bcl6. Exogenous ADAR1 induced adenosine-targeted DNA mutations in IgM B cells from ADAR1-transgenic mice and in wild-type mouse embryonic fibroblasts (MEFs). These mutations accumulated in senescent MEFs accompanied with endogenous ADAR1 expression, and the frequency in senescent Bcl6-deficient MEFs was higher than senescent wild-type MEFs. Thus, Bcl6 protects senescent cells from accumulation of adenosine-targeted DNA mutations induced by ADAR1.


Subject(s)
Adenosine Deaminase/physiology , Adenosine/metabolism , DNA-Binding Proteins/physiology , DNA/genetics , Mutation , Animals , Cells, Cultured , DNA-Binding Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Proto-Oncogene Proteins c-bcl-6 , RNA-Binding Proteins
17.
Nat Commun ; 15(1): 840, 2024 Jan 29.
Article in English | MEDLINE | ID: mdl-38287012

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by dysregulated B cell compartment responsible for the production of autoantibodies. Here, we show that T cell-specific expression of calcium/calmodulin-dependent protein kinase IV (CaMK4) leads to T follicular helper (Tfh) cells expansion in models of T-dependent immunization and autoimmunity. Mechanistically, CaMK4 controls the Tfh-specific transcription factor B cell lymphoma 6 (Bcl6) at the transcriptional level through the cAMP responsive element modulator α (CREMα). In the absence of CaMK4 in T cells, germinal center formation and humoral immunity is impaired in immunized mice, resulting in reduced anti-dsDNA titres, as well as IgG and complement kidney deposition in the lupus-prone B6.lpr mouse. In human Tfh cells, CaMK4 inhibition reduced BCL6 expression and IL-21 secretion ex vivo, resulting in impaired plasmablast formation and IgG production. In patients with SLE, CAMK4 mRNA levels in Tfh cells correlated with those of BCL6. In conclusion, we identify CaMK4/CREMα as a driver of T cell-dependent B cell dysregulation in autoimmunity.


Subject(s)
Lupus Erythematosus, Systemic , T Follicular Helper Cells , Animals , Humans , Mice , Autoimmunity , Cell Differentiation/genetics , Immunoglobulin G/metabolism , T Follicular Helper Cells/metabolism , T-Lymphocytes, Helper-Inducer
18.
Nat Commun ; 15(1): 8304, 2024 Sep 27.
Article in English | MEDLINE | ID: mdl-39333474

ABSTRACT

CD38 has emerged as a potential therapeutic target for patients with systemic lupus erythematosus (SLE) but it is not known whether CD38 alters CD4+ T cell function. Using primary human T cells and CD38-sufficient and CD38-deficient Jurkat T cells, we demonstrate that CD38 shifts the T cell lipid profile of gangliosides from GM3 to GM2 by upregulating B4GALNT1 in a Sirtuin 1-dependent manner. Enhanced expression of GM2 causes ER stress by enhancing Ca2+ flux through the PLCγ1-IP3 pathway. Interestingly, correction of the calcium overload by an IP3 receptor inhibitor, but not by a store-operated calcium entry (SOCE) inhibitor, improves IL-2 production by CD4+ T cells in SLE. This study demonstrates that CD38 affects calcium homeostasis in CD4+ T cells by controlling cell membrane lipid composition that results in suppressed IL-2 production. CD38 inhibition with biologics or small drugs should be expected to benefit patients with SLE.


Subject(s)
ADP-ribosyl Cyclase 1 , CD4-Positive T-Lymphocytes , Calcium , Cell Membrane , Interleukin-2 , Lupus Erythematosus, Systemic , Female , Humans , ADP-ribosyl Cyclase 1/metabolism , ADP-ribosyl Cyclase 1/genetics , Calcium/metabolism , CD4-Positive T-Lymphocytes/metabolism , CD4-Positive T-Lymphocytes/immunology , Cell Membrane/metabolism , Interleukin-2/metabolism , Jurkat Cells , Lupus Erythematosus, Systemic/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/genetics , Membrane Glycoproteins/metabolism , Membrane Glycoproteins/genetics , Phospholipase C gamma/metabolism , Phospholipase C gamma/genetics
19.
Arthritis Rheumatol ; 2024 Sep 15.
Article in English | MEDLINE | ID: mdl-39279154

ABSTRACT

OBJECTIVE: Enhanced expression of transforming growth factor (TGF) ß in the kidneys of patients with lupus nephritis (LN) can lead to progressive fibrosis, resulting in end-organ damage. ADAM9 activates TGFß1 by cleaving the latency-associated peptide (LAP). We hypothesized that ADAM9 in the kidney may accelerate fibrogenesis by activating TGFß1. METHODS: We assessed the expression of ADAM9 in the kidneys of mice and humans who were lupus prone. In vitro experiments were conducted using tubular epithelial cells (TECs) isolated from mice and explored the mechanisms responsible for the up-regulation of ADAM9 and the subsequent activation of TGFß1. To assess the role of ADAM9 in the development of tubular-intestinal fibrosis in individuals with LN, we generated MRL/lpr mice who were Adam9 deficient. RESULTS: ADAM9 was highly expressed in tubules from MRL/lpr mice. The transcription factor hypoxia-inducible factor-1α was found to promote the transcription of ADAM9 in TECs. TECs from mice who were Adam9 deficient and exposed to the hypoxia mimetic agent dimethyloxalylglycine failed to cleave the LAP to produce bioactive TGFß1 from latent TGFß1. Coculture of TECs from mice who were Adam9 deficient with fibroblasts in the presence of dimethyloxalylglycine and latent TGFß1 produced decreased amounts of type I collagen and α-smooth muscle actin (SMA) by fibroblasts. MRL/lpr mice who were Adam9 deficient showed reduced interstitial fibrosis. At the translational level, ADAM9 expression in tissues and urine of patients with LN was found to increase. CONCLUSION: Hypoxia promotes the expression of ADAM9 by TECs, which is responsible for the development of interstitial fibrosis in patients with LN by enhancing the TGFß1 activation, which promotes fibroblasts to produce collagen and α-SMA.

20.
J Immunol ; 186(5): 2800-8, 2011 Mar 01.
Article in English | MEDLINE | ID: mdl-21270405

ABSTRACT

CXCR4 expression is critical for localization of centroblasts in the dark zone of germinal centers (GCs), and centrocytes downregulate CXCR4 and thus leave the dark zone to reside in the light zone. However, mechanisms governing CXCR4 downregulation on centrocytes are not known. In this study, we show that the amount of intracellular CXCR4 in centroblasts was similar to that in centrocytes, suggesting differential control of CXCR4 protein expression in these GC B cells. Restimulation of activated B cells with IL-21, which is a major cytokine produced by T follicular helper cells, accelerated CXCR4 internalization by inducing endocytosis-related GRK6 expression. Although CXCR4 expression was downregulated on GC B cells by IL-21 stimulation, CXCR4(low) centrocytes developed in the spleens of IL-21R-deficient mice, suggesting other mechanisms for downregulation. The level of CD63 (which recruits CXCR4 to late endosome in CD4 T cells) in centrocytes was more than that in centroblasts and was strikingly elevated in activated Bcl6-deficient B cells. Bcl6, a transcriptional repressor, was detected on the chromatin of the CD63 gene in resting B cells, therefore CD63 is a molecular target of Bcl6. Downregulation of CD63 mRNA in activated Bcl6-deficient B cells by small interfering RNA upregulated CXCR4 expression on the B cells. Furthermore, addition of Bcl6 inhibitor to activated B cell cultures increased CD63 mRNA expression in (and downregulated CXCR4 expression on) those activated B cells. Thus, CXCR4 can be downregulated on activated B cells by IL-21-induced endocytosis and CD63-mediated endosomal recruitment, and these mechanisms may contribute to downregulation of CXCR4 on centrocytes.


Subject(s)
Antigens, CD/physiology , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Down-Regulation/immunology , Interleukins/physiology , Lymphocyte Activation/immunology , Platelet Membrane Glycoproteins/physiology , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/biosynthesis , Animals , Antigens, CD/biosynthesis , Antigens, CD/genetics , B-Lymphocyte Subsets/cytology , Cells, Cultured , Endocytosis/immunology , Endosomes/immunology , Endosomes/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Platelet Membrane Glycoproteins/biosynthesis , Platelet Membrane Glycoproteins/genetics , Receptors, CXCR4/metabolism , Tetraspanin 30
SELECTION OF CITATIONS
SEARCH DETAIL