Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Biotechnol ; 41(9): 1229-1238, 2023 09.
Article in English | MEDLINE | ID: mdl-36658341

ABSTRACT

Muscle-specific tyrosine kinase myasthenia gravis (MuSK MG) is an autoimmune disease that causes life-threatening muscle weakness due to anti-MuSK autoantibodies that disrupt neuromuscular junction signaling. To avoid chronic immunosuppression from current therapies, we engineered T cells to express a MuSK chimeric autoantibody receptor with CD137-CD3ζ signaling domains (MuSK-CAART) for precision targeting of B cells expressing anti-MuSK autoantibodies. MuSK-CAART demonstrated similar efficacy as anti-CD19 chimeric antigen receptor T cells for depletion of anti-MuSK B cells and retained cytolytic activity in the presence of soluble anti-MuSK antibodies. In an experimental autoimmune MG mouse model, MuSK-CAART reduced anti-MuSK IgG without decreasing B cells or total IgG levels, reflecting MuSK-specific B cell depletion. Specific off-target interactions of MuSK-CAART were not identified in vivo, in primary human cell screens or by high-throughput human membrane proteome array. These data contributed to an investigational new drug application and phase 1 clinical study design for MuSK-CAART for the treatment of MuSK autoantibody-positive MG.


Subject(s)
Myasthenia Gravis, Autoimmune, Experimental , Receptors, Cholinergic , Humans , Mice , Animals , Receptors, Cholinergic/therapeutic use , Autoantigens/therapeutic use , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , T-Lymphocytes , Autoantibodies/therapeutic use , Immunoglobulin G , Protein-Tyrosine Kinases/therapeutic use , Muscles
2.
3.
Cell Mol Gastroenterol Hepatol ; 14(4): 731-750, 2022.
Article in English | MEDLINE | ID: mdl-35835390

ABSTRACT

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) is characterized by severe gastrointestinal inflammation, but many patients experience extra-intestinal disease. Bone loss is one common extra-intestinal manifestation of IBD that occurs through dysregulated interactions between osteoclasts and osteoblasts. Systemic inflammation has been postulated to contribute to bone loss, but the specific pathologic mechanisms have not yet been fully elucidated. We hypothesized that intestinal inflammation leads to bone loss through increased abundance and altered function of osteoclast progenitors. METHODS: We used chemical, T cell driven, and infectious models of intestinal inflammation to determine the impact of intestinal inflammation on bone volume, the skeletal cytokine environment, and the cellular changes to pre-osteoclast populations within bone marrow. Additionally, we evaluated the potential for monoclonal antibody treatment against an inflammation-induced osteoclast co-receptor, myeloid DNAX activation protein 12-associating lectin-1 (MDL-1) to reduce bone loss during colitis. RESULTS: We observed significant bone loss across all models of intestinal inflammation. Bone loss was associated with an increase in pro-osteoclastogenic cytokines within the bone and an expansion of a specific Cd11b-/loLy6Chi osteoclast precursor (OCP) population. Intestinal inflammation led to altered OCP expression of surface receptors involved in osteoclast differentiation and function, including the pro-osteoclastogenic co-receptor MDL-1. OCPs isolated from mice with intestinal inflammation demonstrated enhanced osteoclast differentiation ex vivo compared to controls, which was abrogated by anti-MDL-1 antibody treatment. Importantly, in vivo anti-MDL-1 antibody treatment ameliorated bone loss during intestinal inflammation. CONCLUSIONS: Collectively, these data implicate the pathologic expansion and altered function of OCPs expressing MDL-1 in bone loss during IBD.


Subject(s)
Bone Resorption , Inflammatory Bowel Diseases , Lectins, C-Type , Osteoclasts , Osteogenesis , Receptors, Cell Surface , Animals , Antibodies, Monoclonal/metabolism , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation/physiology , Cytokines/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Inflammatory Bowel Diseases/genetics , Inflammatory Bowel Diseases/metabolism , Inflammatory Bowel Diseases/pathology , Intestines/metabolism , Lectins/metabolism , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Mice , Osteoclasts/metabolism , Osteoclasts/pathology , Osteogenesis/genetics , Osteogenesis/physiology , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism
4.
J Invest Dermatol ; 142(3 Pt B): 907-914, 2022 03.
Article in English | MEDLINE | ID: mdl-34756581

ABSTRACT

Pemphigus and pemphigoid are paradigms for understanding the mechanisms of antibody-mediated autoimmune disease in humans. In pemphigus, IgG4-predominant autoantibodies cause intraepidermal blistering by direct interference with desmoglein interactions and subsequent disruption of desmosomes and signaling pathways. In pemphigoid, IgG1, IgG4, and IgE autoantibodies against basement membrane zone antigens directly interfere with hemidesmosomal adhesion, activating complement and Fc receptor‒mediated effector pathways. Unraveling disease mechanisms in pemphigus and pemphigoid has identified numerous opportunities for clinical trials, which hold promise to identify safer and more effective therapies for these potentially life-threatening diseases.


Subject(s)
Autoimmune Diseases , Pemphigoid, Bullous , Pemphigus , Autoantibodies , Humans , Immunoglobulin G , Pemphigoid, Bullous/drug therapy , Pemphigus/drug therapy
5.
Cancer Res ; 81(12): 3241-3254, 2021 06 15.
Article in English | MEDLINE | ID: mdl-33619116

ABSTRACT

Fusion genes including NPM-ALK can promote T-cell transformation, but the signals required to drive a healthy T cell to become malignant remain undefined. In this study, we introduce NPM-ALK into primary human T cells and demonstrate induction of the epithelial-to-mesenchymal transition (EMT) program, attenuation of most T-cell effector programs, reemergence of an immature epigenomic profile, and dynamic regulation of c-Myc, E2F, and PI3K/mTOR signaling pathways early during transformation. A mutant of NPM-ALK failed to bind several signaling complexes including GRB2/SOS, SHC1, SHC4, and UBASH3B and was unable to transform T cells. Finally, T-cell receptor (TCR)-generated signals were required to achieve T-cell transformation, explaining how healthy individuals can harbor T cells with NPM-ALK translocations. These findings describe the fundamental mechanisms of NPM-ALK-mediated oncogenesis and may serve as a model to better understand factors that regulate tumor formation. SIGNIFICANCE: This investigation into malignant transformation of T cells uncovers a requirement for TCR triggering, elucidates integral signaling complexes nucleated by NPM-ALK, and delineates dynamic transcriptional changes as a T cell transforms.See related commentary by Spasevska and Myklebust, p. 3160.


Subject(s)
Cell Dedifferentiation , Cell Transformation, Neoplastic/pathology , Cellular Reprogramming , Lymphoma, Large-Cell, Anaplastic/pathology , Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Apoptosis , Cell Proliferation , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/metabolism , Humans , Lymphoma, Large-Cell, Anaplastic/genetics , Lymphoma, Large-Cell, Anaplastic/immunology , Lymphoma, Large-Cell, Anaplastic/metabolism , Phosphorylation , Protein-Tyrosine Kinases/genetics , Receptors, Antigen, T-Cell/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Tumor Cells, Cultured
6.
Front Pharmacol ; 11: 1153, 2020.
Article in English | MEDLINE | ID: mdl-32848762

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAID)s relieve pain, inflammation, and fever by inhibiting the activity of cyclooxygenase isozymes (COX-1 and COX-2). Despite their clinical efficacy, NSAIDs can cause gastrointestinal (GI) and cardiovascular (CV) complications. Moreover, NSAID use is characterized by a remarkable individual variability in the extent of COX isozyme inhibition, therapeutic efficacy, and incidence of adverse effects. The interaction between the gut microbiota and host has emerged as a key player in modulating host physiology, gut microbiota-related disorders, and metabolism of xenobiotics. Indeed, host-gut microbiota dynamic interactions influence NSAID disposition, therapeutic efficacy, and toxicity. The gut microbiota can directly cause chemical modifications of the NSAID or can indirectly influence its absorption or metabolism by regulating host metabolic enzymes or processes, which may have consequences for drug pharmacokinetic and pharmacodynamic properties. NSAID itself can directly impact the composition and function of the gut microbiota or indirectly alter the physiological properties or functions of the host which may, in turn, precipitate in dysbiosis. Thus, the complex interconnectedness between host-gut microbiota and drug may contribute to the variability in NSAID response and ultimately influence the outcome of NSAID therapy. Herein, we review the interplay between host-gut microbiota and NSAID and its consequences for both drug efficacy and toxicity, mainly in the GI tract. In addition, we highlight progress towards microbiota-based intervention to reduce NSAID-induced enteropathy.

8.
Pharmacol Res ; 149: 104456, 2019 11.
Article in English | MEDLINE | ID: mdl-31553935

ABSTRACT

Prostaglandins (PG) are pleiotropic bioactive lipids involved in the control of many physiological processes, including key roles in regulating inflammation. This links PG to the modulation of the quality and magnitude of immune responses. T cells, as a core part of the immune system, respond readily to inflammatory cues from their environment, and express a diverse array of PG receptors that contribute to their function and phenotype. Here we put in context our knowledge about how PG affect T cell biology, and review advances that bring light into how specific T cell functions that have been newly discovered are modulated through PG. We will also comment on drugs that target PG metabolism and sensing, their effect on T cell function during disease, and we will finally discuss how we can design new approaches that modulate PG in order to maximize desired therapeutic T cell effects.


Subject(s)
Prostaglandins/immunology , T-Lymphocytes/immunology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Cell Differentiation/drug effects , Drug Discovery , Humans , Immunity/drug effects , Inflammation/drug therapy , Inflammation/immunology , Inflammation/metabolism , Prostaglandins/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism
9.
mBio ; 10(1)2019 01 08.
Article in English | MEDLINE | ID: mdl-30622186

ABSTRACT

Clostridium difficile infection (CDI) is a major public health threat worldwide. The use of nonsteroidal anti-inflammatory drugs (NSAIDs) is associated with enhanced susceptibility to and severity of CDI; however, the mechanisms driving this phenomenon have not been elucidated. NSAIDs alter prostaglandin (PG) metabolism by inhibiting cyclooxygenase (COX) enzymes. Here, we found that treatment with the NSAID indomethacin prior to infection altered the microbiota and dramatically increased mortality and the intestinal pathology associated with CDI in mice. We demonstrated that in C. difficile-infected animals, indomethacin treatment led to PG deregulation, an altered proinflammatory transcriptional and protein profile, and perturbed epithelial cell junctions. These effects were paralleled by increased recruitment of intestinal neutrophils and CD4+ cells and also by a perturbation of the gut microbiota. Together, these data implicate NSAIDs in the disruption of protective COX-mediated PG production during CDI, resulting in altered epithelial integrity and associated immune responses.IMPORTANCEClostridium difficile infection (CDI) is a spore-forming anaerobic bacterium and leading cause of antibiotic-associated colitis. Epidemiological data suggest that use of nonsteroidal anti-inflammatory drugs (NSAIDs) increases the risk for CDI in humans, a potentially important observation given the widespread use of NSAIDs. Prior studies in rodent models of CDI found that NSAID exposure following infection increases the severity of CDI, but mechanisms to explain this are lacking. Here we present new data from a mouse model of antibiotic-associated CDI suggesting that brief NSAID exposure prior to CDI increases the severity of the infectious colitis. These data shed new light on potential mechanisms linking NSAID use to worsened CDI, including drug-induced disturbances to the gut microbiome and colonic epithelial integrity. Studies were limited to a single NSAID (indomethacin), so future studies are needed to assess the generalizability of our findings and to establish a direct link to the human condition.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Clostridium Infections/mortality , Clostridium Infections/pathology , Gastrointestinal Microbiome/drug effects , Indomethacin/adverse effects , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , CD4-Positive T-Lymphocytes/immunology , Indomethacin/administration & dosage , Mice , Neutrophils/immunology , Prostaglandins/analysis , Survival Analysis
10.
Cell ; 175(7): 1780-1795.e19, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30392958

ABSTRACT

Activated T cells differentiate into functional subsets with distinct metabolic programs. Glutaminase (GLS) converts glutamine to glutamate to support the tricarboxylic acid cycle and redox and epigenetic reactions. Here, we identify a key role for GLS in T cell activation and specification. Though GLS deficiency diminished initial T cell activation and proliferation and impaired differentiation of Th17 cells, loss of GLS also increased Tbet to promote differentiation and effector function of CD4 Th1 and CD8 CTL cells. This was associated with altered chromatin accessibility and gene expression, including decreased PIK3IP1 in Th1 cells that sensitized to IL-2-mediated mTORC1 signaling. In vivo, GLS null T cells failed to drive Th17-inflammatory diseases, and Th1 cells had initially elevated function but exhausted over time. Transient GLS inhibition, however, led to increased Th1 and CTL T cell numbers. Glutamine metabolism thus has distinct roles to promote Th17 but constrain Th1 and CTL effector cell differentiation.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Glutaminase/immunology , Lymphocyte Activation , Th1 Cells/immunology , Th17 Cells/immunology , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/genetics , Glutaminase/genetics , Male , Mice , Mice, Transgenic , Th1 Cells/cytology , Th17 Cells/cytology
11.
J Immunol ; 201(3): 861-873, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29950508

ABSTRACT

Early breaches in B cell tolerance are central to type 1 diabetes progression in mouse and man. Conventional BCR transgenic mouse models (VH125.Tg NOD) reveal the power of B cell specificity to drive disease as APCs. However, in conventional fixed IgM models, comprehensive assessment of B cell development is limited. To provide more accurate insight into the developmental and functional fates of anti-insulin B cells, we generated a new NOD model (VH125SDNOD) in which anti-insulin VDJH125 is targeted to the IgH chain locus to generate a small (1-2%) population of class switch-competent insulin-binding B cells. Tracking of this rare population in a polyclonal repertoire reveals that anti-insulin B cells are preferentially skewed into marginal zone and late transitional subsets known to have increased sensitivity to proinflammatory signals. Additionally, IL-10 production, characteristic of regulatory B cell subsets, is increased. In contrast to conventional models, class switch-competent anti-insulin B cells proliferate normally in response to mitogenic stimuli but remain functionally silent for insulin autoantibody production. Diabetes development is accelerated, which demonstrates the power of anti-insulin B cells to exacerbate disease without differentiation into Ab-forming or plasma cells. Autoreactive T cell responses in VH125SDNOD mice are not restricted to insulin autoantigens, as evidenced by increased IFN-γ production to a broad array of diabetes-associated epitopes. Together, these results independently validate the pathogenic role of anti-insulin B cells in type 1 diabetes, underscore their diverse developmental fates, and demonstrate the pathologic potential of coupling a critical ß cell specificity to predominantly proinflammatory Ag-presenting B cell subsets.


Subject(s)
Antigen Presentation/immunology , B-Lymphocyte Subsets/immunology , Diabetes Mellitus, Type 1/immunology , Insulin Antibodies/immunology , Insulin/immunology , Animals , Autoantibodies/immunology , Autoantigens/immunology , Female , Immune Tolerance/immunology , Inflammation/immunology , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred NOD , Mice, Transgenic , Receptors, Antigen, B-Cell/immunology
12.
Front Immunol ; 9: 2954, 2018.
Article in English | MEDLINE | ID: mdl-30619314

ABSTRACT

PGE2 is a lipid mediator of the initiation and resolution phases of inflammation, as well as a regulator of immune system responses to inflammatory events. PGE2 is produced and sensed by T cells, and autocrine or paracrine PGE2 can affect T cell phenotype and function. In this study, we use a T cell-dependent model of colitis to evaluate the role of PGE2 on pathological outcome and T-cell phenotypes. CD4+ T effector cells either deficient in mPGES-1 or the PGE2 receptor EP4 are less colitogenic. Absence of T cell autocrine mPGES1-dependent PGE2 reduces colitogenicity in association with an increase in CD4+RORγt+ cells in the lamina propria. In contrast, recipient mice deficient in mPGES-1 exhibit more severe colitis that corresponds with a reduced capacity to generate FoxP3+ T cells, especially in mesenteric lymph nodes. Thus, our research defines how mPGES-1-driven production of PGE2 by different cell types in distinct intestinal locations impacts T cell function during colitis. We conclude that PGE2 has profound effects on T cell phenotype that are dependent on the microenvironment.


Subject(s)
Colitis/immunology , Dinoprostone/immunology , Prostaglandin-E Synthases/immunology , Receptors, Prostaglandin E, EP4 Subtype/immunology , T-Lymphocytes/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Colitis/genetics , Colitis/metabolism , Dinoprostone/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP2 Subtype/immunology , Receptors, Prostaglandin E, EP2 Subtype/metabolism , Receptors, Prostaglandin E, EP4 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/metabolism , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism
13.
J Immunol ; 200(2): 725-736, 2018 01 15.
Article in English | MEDLINE | ID: mdl-29237778

ABSTRACT

The integration of inflammatory signals is paramount in controlling the intensity and duration of immune responses. Eicosanoids, particularly PGE2, are critical molecules in the initiation and resolution of inflammation and in the transition from innate to acquired immune responses. Microsomal PGE synthase 1 (mPGES1) is an integral membrane enzyme whose regulated expression controls PGE2 levels and is highly expressed at sites of inflammation. PGE2 is also associated with modulation of autoimmunity through altering the IL-23/IL-17 axis and regulatory T cell (Treg) development. During a type II collagen-CFA immunization response, lack of mPGES1 impaired the numbers of CD4+ regulatory (Treg) and Th17 cells in the draining lymph nodes. Ag-experienced mPGES1-/- CD4+ cells showed impaired IL-17A, IFN-γ, and IL-6 production when rechallenged ex vivo with their cognate Ag compared with their wild-type counterparts. Additionally, production of PGE2 by cocultured APCs synergized with that of Ag-experienced CD4+ T cells, with mPGES1 competence in the APC compartment enhancing CD4+ IL-17A and IFN-γ responses. However, in contrast with CD4+ cells that were Ag primed in vivo, exogenous PGE2 inhibited proliferation and skewed IL-17A to IFN-γ production under Th17 polarization of naive T cells in vitro. We conclude that mPGES1 is necessary in vivo to mount optimal Treg and Th17 responses during an Ag-driven primary immune response. Furthermore, we uncover a coordination of autocrine and paracrine mPGES1-driven PGE2 production that impacts effector T cell IL-17A and IFN-γ responses.


Subject(s)
Autocrine Communication , Dinoprostone/metabolism , Paracrine Communication , Prostaglandin-E Synthases/genetics , Th1 Cells/immunology , Th1 Cells/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Animals , Epitopes, T-Lymphocyte/immunology , Gene Expression Regulation , Immunization , Immunomodulation , Lymphocyte Activation/immunology , Mice , Phenotype , Prostaglandin-E Synthases/metabolism , Receptors, Prostaglandin E, EP2 Subtype/genetics , Receptors, Prostaglandin E, EP4 Subtype/genetics
14.
Proc Natl Acad Sci U S A ; 113(19): 5370-5, 2016 May 10.
Article in English | MEDLINE | ID: mdl-27114551

ABSTRACT

Natural killer (NK) cells are innate lymphocytes that recognize and lyse virally infected or transformed cells. This latter property is being pursued in clinics to treat leukemia with the hope that further breakthroughs in NK cell biology can extend treatments to other cancers. At issue is the ability to expand transferred NK cells and prolong their functionality within the context of a tumor. In terms of NK cell expansion and survival, we now report that Kruppel-like factor 2 (KLF2) is a key transcription factor that underpins both of these events. Excision of Klf2 using gene-targeted mouse models promotes spontaneous proliferation of immature NK cells in peripheral tissues, a phenotype that is replicated under ex vivo conditions. Moreover, KLF2 imprints a homeostatic migration pattern on mature NK cells that allows these cells to access IL-15-rich microenvironments. KLF2 accomplishes this feat within the mature NK cell lineage via regulation of a subset of homing receptors that respond to homeostatic ligands while leaving constitutively expressed receptors that recognize inflammatory cytokines unperturbed. Under steady-state conditions, KLF2-deficient NK cells alter their expression of homeostatic homing receptors and subsequently undergo apoptosis due to IL-15 starvation. This novel mechanism has implications regarding NK cell contraction following the termination of immune responses including the possibility that retention of an IL-15 transpresenting support system is key to extending NK cell activity in a tumor environment.


Subject(s)
Cell Proliferation/physiology , Cell Survival/physiology , Interleukin-15/metabolism , Killer Cells, Natural/cytology , Killer Cells, Natural/physiology , Kruppel-Like Transcription Factors/metabolism , Animals , Cells, Cultured , Gene Expression Regulation/physiology , Homeostasis/physiology , Mice , Mice, Inbred C57BL
15.
Arthritis Rheumatol ; 68(8): 1856-68, 2016 08.
Article in English | MEDLINE | ID: mdl-26945549

ABSTRACT

OBJECTIVE: Bruton's tyrosine kinase (BTK) is a B cell signaling protein that also contributes to innate immunity. BTK inhibitors prevent autoimmune arthritis but have off-target effects, and the mechanisms of protection remain unknown. We undertook these studies using genetic deletion to investigate the role of BTK in adaptive and innate immune responses that drive inflammatory arthritis. METHODS: BTK-deficient K/BxN mice were generated to study the role of BTK in a spontaneous model that requires both adaptive and innate immunity. The K/BxN serum-transfer model was used to bypass the adaptive system and elucidate the role of BTK in innate immune contributions to arthritis. RESULTS: BTK deficiency conferred disease protection to K/BxN mice, confirming outcomes of BTK inhibitors. B lymphocytes were profoundly reduced, more than in other models of BTK deficiency. Subset analysis revealed loss of B cells at all developmental stages. Germinal center B cells were also decreased, with downstream effects on numbers of follicular helper T cells and greatly reduced autoantibodies. In contrast, total IgG was only mildly decreased. Strikingly, and in contrast to small molecule inhibitors, BTK deficiency had no effect in the serum-transfer model of arthritis. CONCLUSION: BTK contributes to autoimmune arthritis primarily through its role in B cell signaling and not through innate immune components.


Subject(s)
Antigen-Antibody Complex/immunology , Arthritis/enzymology , Arthritis/immunology , Autoimmune Diseases/enzymology , Autoimmune Diseases/etiology , Protein-Tyrosine Kinases/deficiency , Agammaglobulinaemia Tyrosine Kinase , Animals , Male , Mice
16.
Proc Natl Acad Sci U S A ; 111(26): 9579-84, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24979767

ABSTRACT

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


Subject(s)
Autoimmunity/immunology , Cell Differentiation/immunology , Kruppel-Like Transcription Factors/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmunity/genetics , Chromatin Immunoprecipitation , DNA Primers/genetics , Flow Cytometry , Kruppel-Like Transcription Factors/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic
17.
Expert Rev Clin Immunol ; 10(6): 747-62, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24734886

ABSTRACT

B cells mediate multiple functions that influence immune and inflammatory responses in rheumatoid arthritis. Production of a diverse array of autoantibodies can happen at different stages of the disease, and are important markers of disease outcome. In turn, the magnitude and quality of acquired humoral immune responses is strongly dependent on signals delivered by innate immune cells. Additionally, the milieu of cells and chemokines that constitute a niche for plasma cells rely strongly on signals provided by stromal cells at different anatomical locations and times. The chronic inflammatory state therefore importantly impacts the developing humoral immune response and its intensity and specificity. We focus this review on B cell biology and the role of the innate immune system in the development of autoimmunity in patients with rheumatoid arthritis.


Subject(s)
Arthritis, Rheumatoid/immunology , B-Lymphocyte Subsets/immunology , Immunity, Innate , Plasma Cells/immunology , Signal Transduction/immunology , Animals , Arthritis, Rheumatoid/pathology , Autoantibodies/immunology , B-Lymphocyte Subsets/pathology , Chemokines/immunology , Chronic Disease , Humans , Plasma Cells/pathology , Stromal Cells/immunology , Stromal Cells/pathology
18.
J Immunol ; 191(5): 2780-2795, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-23918988

ABSTRACT

The spleen regulatory B cell subset with the functional capacity to express IL-10 (B10 cells) modulates both immune responses and autoimmune disease severity. However, the peritoneal cavity also contains relatively high frequencies of functionally defined IL-10-competent B10 cells. In this study, peritoneal cavity B10 cells shared similar cell surface phenotypes with their spleen counterparts. However, peritoneal cavity B10 cells were 10-fold more frequent among B cells than occurred within the spleen, intestinal tract, or mesenteric lymph nodes and were present at higher proportions among the phenotypically defined peritoneal B1a > B1b > B2 cell subpopulations. The development or localization of B10 cells within the peritoneal cavity was not dependent on the presence of commensal microbiota, T cells, IL-10 or B10 cell IL-10 production, or differences between their fetal liver or adult bone marrow progenitor cell origins. The BCR repertoire of peritoneal cavity B10 cells was diverse, as occurs in the spleen, and predominantly included germline-encoded VH and VL regions commonly found in either the conventional or B1 B cell compartments. Thereby, the capacity to produce IL-10 appears to be an intrinsic functional property acquired by clonally diverse B cells. Importantly, IL-10 production by peritoneal cavity B cells significantly reduced disease severity in spontaneous and induced models of colitis by regulating neutrophil infiltration, colitogenic CD4(+) T cell activation, and proinflammatory cytokine production during colitis onset. Thus, the numerically small B10 cell subset within the peritoneal cavity has regulatory function and is important for maintaining homeostasis within gastrointestinal tissues and the immune system.


Subject(s)
B-Lymphocytes, Regulatory/immunology , CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Peritoneal Cavity/cytology , Adoptive Transfer , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Fluorescent Antibody Technique , Interferon-gamma/immunology , Interleukin-10/biosynthesis , Interleukin-10/immunology , Lymphocyte Subsets/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
19.
J Immunol ; 188(3): 1036-48, 2012 Feb 01.
Article in English | MEDLINE | ID: mdl-22198952

ABSTRACT

Regulatory B cells that are functionally defined by their capacity to express IL-10 (B10 cells) downregulate inflammation and autoimmunity. In studies using well-defined IL-10 reporter mice, this rare B10 cell subset was also found to maintain a capacity for plasma cell differentiation. During a transient period of il10 transcription, the blimp1 and irf4 transcription factors were induced in B10 cells, whereas pax5 and bcl6 were downregulated as a significant fraction of B10 cells completed the genetic and phenotypic program leading to Ab-secreting cell differentiation in vitro and in vivo. B10 cell-derived IgM reacted with both self- and foreign Ags, whereas B10 cells generated Ag-specific IgG in response to immunizations. Moreover, B10 cells represented a significant source of serum IgM and IgG during adoptive-transfer experiments and produced Ag-specific, polyreactive and autoreactive Ab specificities that were consistent with their expression of a diverse AgR repertoire. Thereby, B10 cells limit inflammation and immune responses by the transient production of IL-10, and may facilitate clearance of their eliciting Ags through an inherent capacity to quickly generate polyreactive and/or Ag-specific Abs during humoral immune responses.


Subject(s)
Antibody-Producing Cells/cytology , B-Lymphocyte Subsets/cytology , B-Lymphocytes, Regulatory/immunology , Cell Differentiation/immunology , Interleukin-10/biosynthesis , Animals , Antigens , Autoimmunity , B-Lymphocyte Subsets/immunology , B-Lymphocytes, Regulatory/cytology , Gene Expression Regulation , Immunity, Humoral , Immunoglobulin G , Immunoglobulin M , Inflammation , Mice
20.
Nat Med ; 14(7): 748-55, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18542049

ABSTRACT

Autoantibody-mediated diseases like myasthenia gravis, autoimmune hemolytic anemia and systemic lupus erythematosus represent a therapeutic challenge. In particular, long-lived plasma cells producing autoantibodies resist current therapeutic and experimental approaches. Recently, we showed that the sensitivity of myeloma cells toward proteasome inhibitors directly correlates with their immunoglobulin synthesis rates. Therefore, we hypothesized that normal plasma cells are also hypersensitive to proteasome inhibition owing to their extremely high amount of protein biosynthesis. Here we show that the proteasome inhibitor bortezomib, which is approved for the treatment of multiple myeloma, eliminates both short- and long-lived plasma cells by activation of the terminal unfolded protein response. Treatment with bortezomib depleted plasma cells producing antibodies to double-stranded DNA, eliminated autoantibody production, ameliorated glomerulonephritis and prolonged survival of two mouse strains with lupus-like disease, NZB/W F1 and MRL/lpr mice. Hence, the elimination of autoreactive plasma cells by proteasome inhibitors might represent a new treatment strategy for antibody-mediated diseases.


Subject(s)
Boronic Acids/pharmacology , Lupus Erythematosus, Systemic/drug therapy , Nephritis/prevention & control , Plasma Cells/drug effects , Protease Inhibitors/pharmacology , Proteasome Inhibitors , Pyrazines/pharmacology , Animals , Bortezomib , Lupus Erythematosus, Systemic/enzymology , Lupus Erythematosus, Systemic/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred MRL lpr , Mice, Inbred NZB , Models, Immunological , Nephritis/immunology , Ovalbumin , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL