Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters

Country/Region as subject
Publication year range
1.
EMBO Rep ; 21(6): e48927, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32363653

ABSTRACT

CD1d-restricted invariant natural killer T (iNKT) cells constitute a common glycolipid-reactive innate-like T-cell subset with a broad impact on innate and adaptive immunity. While several microbial glycolipids are known to activate iNKT cells, the cellular mechanisms leading to endogenous CD1d-dependent glycolipid responses remain largely unclear. Here, we show that endoplasmic reticulum (ER) stress in APCs is a potent inducer of CD1d-dependent iNKT cell autoreactivity. This pathway relies on the presence of two transducers of the unfolded protein response: inositol-requiring enzyme-1a (IRE1α) and protein kinase R-like ER kinase (PERK). Surprisingly, the neutral but not the polar lipids generated within APCs undergoing ER stress are capable of activating iNKT cells. These data reveal that ER stress is an important mechanism to elicit endogenous CD1d-restricted iNKT cell responses through induction of distinct classes of neutral lipids.


Subject(s)
Natural Killer T-Cells , Antigen-Presenting Cells , Antigens, CD1d/genetics , Endoribonucleases , Lipids , Lymphocyte Activation , Protein Serine-Threonine Kinases
2.
EMBO J ; 34(4): 466-74, 2015 Feb 12.
Article in English | MEDLINE | ID: mdl-25599993

ABSTRACT

Antinuclear antibodies are a hallmark feature of generalized autoimmune diseases, including systemic lupus erythematosus and systemic sclerosis. However, the processes underlying the loss of tolerance against nuclear self-constituents remain largely unresolved. Using mice deficient in lymphotoxin and Hox11, we report that approximately 25% of mice lacking secondary lymphoid organs spontaneously develop specific antinuclear antibodies. Interestingly, we find this phenotype is not caused by a defect in central tolerance. Rather, cell-specific deletion and in vivo lymphotoxin blockade link these systemic autoimmune responses to the formation of gut-associated lymphoid tissue in the neonatal period of life. We further demonstrate antinuclear antibody production is influenced by the presence of commensal gut flora, in particular increased colonization with segmented filamentous bacteria, and IL-17 receptor signaling. Together, these data indicate that neonatal colonization of gut microbiota influences generalized autoimmunity in adult life.


Subject(s)
Autoimmunity/immunology , Microbiota/immunology , Animals , Antibodies, Antinuclear/genetics , Antibodies, Antinuclear/immunology , Autoimmunity/genetics , Female , Flow Cytometry , Lymphotoxin-alpha/genetics , Lymphotoxin-alpha/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
3.
Ann Rheum Dis ; 76(3): 585-592, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27551052

ABSTRACT

OBJECTIVES: A20 is an important endogenous regulator of inflammation. Single nucleotide polymorphisms in A20 have been associated with various immune-mediated inflammatory diseases, and cell-specific deletion of A20 results in diverse inflammatory phenotypes. Our goal was to delineate the underlying mechanisms of joint inflammation in myeloid-specific A20-deficient mice (A20myelKO mice). METHODS: Inflammation in A20myelKO mice was assessed in a time-dependent manner. Western blot analysis and quantitative PCR analysis were performed on bone marrow-derived macrophages from A20myelKO and littermate control mice to study the effect of A20 on STAT1/STAT3 expression and STAT1/STAT3-dependent gene transcription in myeloid cells. The in vivo role of Janus kinase-Signal Transducer and Activator of Transcription (JAK-STAT) signalling in the development of enthesitis in A20myelKO mice was assessed following administration of a JAK inhibitor versus placebo control. RESULTS: Enthesitis was found to be an early inflammatory lesion in A20myelKO mice. A20 negatively modulated STAT1-dependent, but generally not STAT3-dependent gene transcription in myeloid cells by suppressing STAT1 but not STAT3 expression, both in unstimulated conditions and after interferon-γ or interleukin-6 stimulation. The increase in STAT1 gene transcription in the absence of A20 was shown to be JAK-STAT-dependent. Moreover, JAK inhibition in vivo resulted in significant reduction of enthesitis, both clinically and histopathologically. CONCLUSIONS: Our data reveal an important and novel interplay between myeloid cells and tissue resident cells at entheseal sites that is regulated by A20. In the absence of A20, STAT1 but not STAT3 expression is enhanced leading to STAT1-dependent inflammation. Therefore, A20 acts as a novel endogenous regulator of STAT1 that prevents onset of enthesitis.


Subject(s)
Enthesopathy/genetics , Enthesopathy/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Animals , Cells, Cultured , Enthesopathy/etiology , Enthesopathy/pathology , Inflammation/complications , Inflammation/genetics , Inflammation/metabolism , Interferon-gamma/pharmacology , Interleukin-6/pharmacology , Janus Kinases/metabolism , Macrophages , Mice , Mice, Knockout , Piperidines/pharmacology , Pyrimidines/pharmacology , Pyrroles/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/genetics
4.
J Immunol ; 193(12): 5960-72, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25381434

ABSTRACT

The regulatory role of the thymic microenvironment during trafficking and differentiation of the invariant NKT (iNKT) cell lineage remains poorly understood. In this study, we show that fractalkine receptor expression marks emigrating subpopulations of the NKT1, NKT2, and NKT17 sublineages in the thymus and peripheral organs of naive mice. Moreover, NKT1 sublineage cells can be subdivided into two subsets, namely NKT1(a) and NKT1(b), which exhibit distinct developmental and tissue-specific distribution profiles. More specifically, development and trafficking of the NKT1(a) subset are selectively dependent upon lymphotoxin (LT)α1ß2-LTß receptor-dependent differentiation of thymic stroma, whereas the NKT1(b), NKT2, and NKT17 sublineages are not. Furthermore, we identify a potential cellular source for LTα1ß2 during thymic organogenesis, marked by expression of IL-7Rα, which promotes differentiation of the NKT1(a) subset in a noncell-autonomous manner. Collectively, we propose a mechanism by which thymic differentiation and retention of the NKT1 sublineage are developmentally coupled to LTα1ß2-LTß receptor-dependent thymic organogenesis.


Subject(s)
Cell Movement , Cellular Microenvironment , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/metabolism , Thymus Gland/immunology , Thymus Gland/metabolism , Animals , CX3C Chemokine Receptor 1 , Cell Differentiation/genetics , Cell Differentiation/immunology , Cluster Analysis , Female , Gene Expression , Gene Expression Profiling , Immunohistochemistry , Immunophenotyping , Interleukin-7 Receptor alpha Subunit/genetics , Interleukin-7 Receptor alpha Subunit/metabolism , Lymph Nodes/immunology , Lymph Nodes/metabolism , Lymph Nodes/pathology , Lymphotoxin alpha1, beta2 Heterotrimer/metabolism , Lymphotoxin beta Receptor/metabolism , Lymphotoxin-beta/deficiency , Male , Mice , Mice, Transgenic , Phenotype , Pregnancy , Receptors, Cytokine/genetics , Receptors, Cytokine/metabolism , Receptors, HIV/genetics , Receptors, HIV/metabolism , Signal Transduction , Thymocytes/immunology , Thymocytes/metabolism
5.
EMBO J ; 30(11): 2294-305, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21552205

ABSTRACT

Invariant natural killer T (iNKT) cells are known to have marked immunomodulatory capacity due to their ability to produce copious amounts of effector cytokines. Here, we report the structure and function of a novel class of aromatic α-galactosylceramide structurally related glycolipids with marked Th1 bias in both mice and men, leading to superior tumour protection in vivo. The strength of the Th1 response correlates well with enhanced lipid binding to CD1d as a result of an induced fit mechanism that binds the aromatic substitution as a third anchor, in addition to the two lipid chains. This induced fit is in contrast to another Th1-biasing glycolipid, α-C-GalCer, whose CD1d binding follows a conventional key-lock principle. These findings highlight the previously unexploited flexibility of CD1d in accommodating galactose-modified glycolipids and broaden the range of glycolipids that can stimulate iNKT cells. We speculate that glycolipids can be designed that induce a similar fit, thereby leading to superior and more sustained iNKT cell responses in vivo.


Subject(s)
Antigens, CD1d/metabolism , Galactosylceramides/metabolism , Natural Killer T-Cells/immunology , Neoplasm Metastasis/prevention & control , Neoplasms/prevention & control , Animals , Mice , Neoplasm Metastasis/immunology , Neoplasms/immunology , Protein Binding
6.
J Hepatol ; 60(1): 175-82, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23973929

ABSTRACT

BACKGROUND & AIMS: Immunometabolism is an emerging field of clinical investigation due to the obesity epidemic worldwide. A reciprocal involvement of immune mediators in the body energy metabolism has been recognized for years, but is only partially understood. We hypothesized that the adipokine leptin could provide an important modulator of iNKT cells. METHODS: The expression of leptin receptor (LR) on resting and activated iNKT cells was measured by flow cytometry. FACS-sorted hepatic iNKT cells were stimulated with anti-CD3/CD28Ab coated beads in the absence or presence of a neutralizing anti-leptin Ab. Furthermore, we evaluated the outcome of LR blocking nanobody treatment in ConA induced hepatitis and towards metabolic parameters in WT and iNKT cell deficient mice. RESULTS: The LR is expressed on iNKT cells and leptin suppresses iNKT cell proliferation and cytokine production in vitro. LR deficient iNKT cells are hyper-responsive further enforcing the role of leptin as an important inhibitor of iNKT cell function. Consistently, in vivo blockade of LR signaling exacerbated ConA hepatitis in wild-type but not in iNKT cell deficient mice, through both Janus kinase (JAK)2 and mitogen-activated protein kinase (MAPK) dependent mechanisms. Moreover, LR inhibition altered fat pad features and was accompanied by insulin resistance, only in wild-type mice. Curiously, this interaction was strictly dependent on MAPK mediated LR signaling in iNKT cells and uncoupled from the more central effects of leptin. CONCLUSIONS: Our data support a new concept of immune regulation by which leptin protects towards T cell mediated hepatitis via modulation of iNKT cells.


Subject(s)
Adipocytes/physiology , Cell Communication , Hepatitis/etiology , Leptin/physiology , Natural Killer T-Cells/physiology , T-Lymphocytes/immunology , Adipose Tissue/metabolism , Animals , Disease Susceptibility , Lymphocyte Activation , MAP Kinase Signaling System , Mice , Receptors, Antigen, T-Cell/physiology , Receptors, Leptin/physiology , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/physiology
7.
J Med Chem ; 67(7): 5233-5258, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38552030

ABSTRACT

The salt-inducible kinases (SIKs) SIK1, SIK2, and SIK3 belong to the adenosine monophosphate-activated protein kinase (AMPK) family of serine/threonine kinases. SIK inhibition represents a new therapeutic approach modulating pro-inflammatory and immunoregulatory pathways that holds potential for the treatment of inflammatory diseases. Here, we describe the identification of GLPG3970 (32), a first-in-class dual SIK2/SIK3 inhibitor with selectivity against SIK1 (IC50 of 282.8 nM on SIK1, 7.8 nM on SIK2 and 3.8 nM on SIK3). We outline efforts made to increase selectivity against SIK1 and improve CYP time-dependent inhibition properties through the structure-activity relationship. The dual activity of 32 in modulating the pro-inflammatory cytokine TNFα and the immunoregulatory cytokine IL-10 is demonstrated in vitro in human primary myeloid cells and human whole blood, and in vivo in mice stimulated with lipopolysaccharide. Compound 32 shows dose-dependent activity in disease-relevant mouse pharmacological models.


Subject(s)
Protein Kinases , Protein Serine-Threonine Kinases , Mice , Humans , Animals , Protein Kinases/metabolism , Cytokines , Tumor Necrosis Factor-alpha
8.
Arthritis Rheum ; 62(4): 988-99, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20131252

ABSTRACT

OBJECTIVE: To investigate the role of invariant natural killer T (iNKT) cells in TNF(DeltaARE/+) mice, an animal model of spondylarthritis (SpA) with both gut and joint inflammation. METHODS: The frequency and activation of iNKT cells were analyzed on mononuclear cells from the lymph nodes and livers of mice, using flow cytometry with alpha-galactosylceramide/CD1d tetramers and quantitative polymerase chain reaction for the invariant V(alpha)14-J(alpha)18 rearrangement. Bone marrow-derived dendritic cells (DCs) were obtained by expansion of primary cells with granulocyte-macrophage colony-stimulating factor followed by coculture with iNKT cell hybridomas, and interleukin-2 release into the cocultures was then measured by enzyme-linked immunosorbent assay (ELISA). Cytokine levels were determined by ELISA or cytometric bead array analyses of freshly isolated DCs and iNKT cells in mixed cocultures. TNF(DeltaARE/+) mice were backcrossed onto J(alpha)18(-/-) and CD1d(-/-) mice, and disease onset was evaluated by clinical scoring, positron emission tomography, and histology. CD1d levels were analyzed on mononuclear cells in paired blood and synovial fluid samples from patients with SpA compared with healthy control subjects. RESULTS: In the absence of iNKT cells, symptoms of gut and joint inflammation in TNF(DeltaARE/+)mice were aggravated. Invariant NKT cells were activated during the course of the disease. This was linked to an enrichment of inflammatory DCs, characterized by high levels of CD1d, particularly at draining sites of inflammation. A similar increase in CD1d levels was observed on DCs from patients with SpA. Inflammatory DCs from TNF(DeltaARE/+) mice stimulated iNKT cells to produce immunomodulatory cytokines, in the absence of exogenous stimulation. Prolonged, continuous exposure, but not short-term exposure, to tumor necrosis factor (TNF) was found to be responsible for the enhanced DC-NKT cell crosstalk. CONCLUSION: This mode of iNKT cell activation represents a natural counterregulatory mechanism for the dampening of TNF-driven inflammation.


Subject(s)
Killer Cells, Natural/immunology , Spondylarthritis/immunology , Animals , Crosses, Genetic , Disease Models, Animal , Flow Cytometry , Gene Rearrangement , Homeostasis/immunology , Inflammation/genetics , Inflammation/immunology , Joint Diseases/genetics , Joint Diseases/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Polymerase Chain Reaction , Spondylarthritis/genetics , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
9.
J Immunol ; 183(4): 2213-6, 2009 Aug 15.
Article in English | MEDLINE | ID: mdl-19620294

ABSTRACT

The current model used to define T cell export from the thymus suggests that emigrating lymphocytes seed the peripheral organs as functionally mature cells. This model holds true for the majority of T cells exported from the thymus with the exception of invariant NK T (iNKT) cells. iNKT cells undergo lineage expansion after positive selection and acquire NK receptor expression once fully mature; yet, the majority of mature iNKT cells are retained in the thymus by an as of yet unidentified mechanism. In this study we demonstrate that mature iNKT cells are retained in the thymus by the chemokine receptor CXCR3. We propose that the expression of CXCR3 ligands in the thymic medullary epithelium promotes the chemotactic retention of mature iNKT thymocytes and prevents leakage of iNKT cells into the peripheral circulation.


Subject(s)
Natural Killer T-Cells/cytology , Natural Killer T-Cells/immunology , Receptors, CXCR3/physiology , Thymus Gland/cytology , Thymus Gland/immunology , Animals , Antigens, Ly/biosynthesis , Cell Differentiation/immunology , Chemotaxis, Leukocyte/immunology , Inflammation Mediators/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , NK Cell Lectin-Like Receptor Subfamily B/biosynthesis , Natural Killer T-Cells/metabolism , Receptors, CXCR3/biosynthesis , Receptors, CXCR3/deficiency , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Thymus Gland/metabolism , Up-Regulation/immunology
10.
J Public Health Manag Pract ; 17(6): 506-12, 2011.
Article in English | MEDLINE | ID: mdl-21964361

ABSTRACT

CONTEXT: Panel management is a central component of the primary care medical home, but faces numerous challenges in the safety net setting. In the San Francisco Department of Public Health, many of our community-based primary care clinics have difficulty accommodating all patients seeking care. OBJECTIVE: We evaluated patient panel size in our 7 clinics providing cradle-to-grave primary care services to more than 25,000 active patients. DESIGN: We adjusted panel size for age, gender, diagnoses, homelessness, and substance abuse; set related policies; and assessed the effects on our clinics. On the basis of our previous data and targets set by other safety net providers, we established a minimum of 1125 patients per full-time paid primary care provider (ie, full-time equivalent [FTE]) in April 2009. We calculated the target panel size each clinic would have if all their providers reached the minimum panel size goal and compared it with the panel size attained by the clinic. RESULTS: Nine months after establishing panel size policy, providers reached 82% of the aggregate target panel size. Five of the 7 clinics increased their adjusted panel size per FTE in the range of 2% to 23%. Two data-oriented and innovative clinics with some of the highest adjusted panel sizes per FTE largely maintained their panel size. Two clinics that had the lowest adjusted panel size per FTE realized a 23% and 8% respective gain; both clinics reduced barriers to new patient appointments. Two clinics acquired new providers and experienced a concomitant drop in panel size per FTE while the new clinicians expanded their panels. One of these clinics had difficulty managing high no-show rates and creating effective appointment templates. CONCLUSIONS: Routine data generation, review of data with administrators and providers, data-driven policies and panel size standards, and interventions to bolster team-based care are important tools for increasing capacity at our primary care clinics.


Subject(s)
Health Services Accessibility , Patient-Centered Care/organization & administration , Public Health Practice , Ambulatory Care Facilities/organization & administration , Capacity Building , Efficiency, Organizational , Humans , Public Policy , San Francisco
11.
Eur J Immunol ; 39(4): 925-30, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19224640

ABSTRACT

The thymus is a primary lymphoid organ supporting the development of self-tolerant T cells. Key events in T-cell development in the thymus include lineage commitment, selection events, and thymic emigration. This review discusses the proposed role of sphingosine-1-phosphate and its receptors in the emigration of both conventional and unconventional T-cell subsets from the thymus, and the molecular machinery currently understood to regulate this process. Furthermore, we highlight a role for chemokines and actin-associated proteins in T-cell motility as recent data suggest that T-cell emigration is regulated by more than just a sphingosine-1-phosphate receptor-1-dependent chemotactic axis.


Subject(s)
Cell Movement/physiology , Kruppel-Like Transcription Factors/metabolism , Receptors, Lysosphingolipid/metabolism , T-Lymphocyte Subsets/immunology , Thymus Gland/immunology , Animals , Cell Movement/immunology , Fingolimod Hydrochloride , Humans , Kruppel-Like Transcription Factors/immunology , Propylene Glycols/pharmacology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/immunology , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , T-Lymphocyte Subsets/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism
12.
PLoS Pathog ; 4(8): e1000122, 2008 Aug 08.
Article in English | MEDLINE | ID: mdl-18688274

ABSTRACT

African trypanosomes are extracellular parasitic protozoa, predominantly transmitted by the bite of the haematophagic tsetse fly. The main mechanism considered to mediate parasitemia control in a mammalian host is the continuous interaction between antibodies and the parasite surface, covered by variant-specific surface glycoproteins. Early experimental studies have shown that B-cell responses can be strongly protective but are limited by their VSG-specificity. We have used B-cell (microMT) and IgM-deficient (IgM(-/-)) mice to investigate the role of B-cells and IgM antibodies in parasitemia control and the in vivo induction of trypanosomiasis-associated anemia. These infection studies revealed that that the initial setting of peak levels of parasitemia in Trypanosoma brucei-infected microMT and IgM(-/-) mice occurred independent of the presence of B-cells. However, B-cells helped to periodically reduce circulating parasites levels and were required for long term survival, while IgM antibodies played only a limited role in this process. Infection-associated anemia, hypothesized to be mediated by B-cell responses, was induced during infection in microMT mice as well as in IgM(-/-) mice, and as such occurred independently from the infection-induced host antibody response. Antigenic variation, the main immune evasion mechanism of African trypanosomes, occurred independently from host antibody responses against the parasite's ever-changing antigenic glycoprotein coat. Collectively, these results demonstrated that in murine experimental T. brucei trypanosomiasis, B-cells were crucial for periodic peak parasitemia clearance, whereas parasite-induced IgM antibodies played only a limited role in the outcome of the infection.


Subject(s)
Anemia/immunology , Antibodies, Protozoan/immunology , B-Lymphocytes/immunology , Immunoglobulin M/immunology , Parasitemia/immunology , Trypanosomiasis, African/immunology , Anemia/genetics , Anemia/pathology , Anemia/physiopathology , Animals , Antibodies, Protozoan/genetics , B-Lymphocytes/parasitology , Female , Immunoglobulin M/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Parasitemia/genetics , Parasitemia/pathology , Trypanosoma brucei brucei , Trypanosomiasis, African/genetics , Trypanosomiasis, African/pathology
13.
J Public Health Manag Pract ; 15(4): 337-44, 2009.
Article in English | MEDLINE | ID: mdl-19525778

ABSTRACT

Patients with a medical home tend to fare better. One of the first steps toward establishing a medical home is to create panels by designating a clinic responsible for each patient. In 2006, we defined active clinic panels (all patients assigned to a clinic and seen there for one or more outpatient medical visits during the past 2 years) for the San Francisco Department of Public Health's 13 community- and four public hospital-based primary care clinics and began automatically assigning previously unassigned patients to clinics based on utilization. In 2007, we created a Web-based user interface for managing panels from within the electronic medical record. Providers and medical directors can now view and verify their panels and link to patient demographic and utilization data. Through April 2008, 14 508 patients have been auto-assigned to a clinic; on average 320 patients were assigned monthly. A total of 82,637 primary care patients were on a clinic panel, and 73.6 percent of them were active. Patient demographics, panel size, and productivity vary considerably by clinic. By establishing active panels and providing Web-based access to panel data, we can systematically assign patients a clinical home; enable providers to manage their panels; accurately measure utilization, capacity, and productivity; assess patient characteristics; and generate clinical quality indicators based on an accurate denominator. These management tools will allow us to set policies and work toward our goal of establishing a medical home for San Franciscans who rely on publicly funded care.


Subject(s)
Patient-Centered Care , Public Health Practice , Demography , Efficiency, Organizational , Health Services/statistics & numerical data , Humans , Program Development , San Francisco
14.
Nat Commun ; 9(1): 5340, 2018 12 17.
Article in English | MEDLINE | ID: mdl-30559399

ABSTRACT

Activated invariant natural killer T (iNKT) cells rapidly produce large amounts of cytokines, but how cytokine mRNAs are induced, stabilized and mobilized following iNKT activation is still unclear. Here we show that an endoplasmic reticulum stress sensor, inositol-requiring enzyme 1α (IRE1α), links key cellular processes required for iNKT cell effector functions in specific iNKT subsets, in which TCR-dependent activation of IRE1α is associated with downstream activation of p38 MAPK and the stabilization of preformed cytokine mRNAs. Importantly, genetic deletion of IRE1α in iNKT cells reduces cytokine production and protects mice from oxazolone colitis. We therefore propose that an IRE1α-dependent signaling cascade couples constitutive cytokine mRNA expression to the rapid induction of cytokine secretion and effector functions in activated iNKT cells.


Subject(s)
Cytokines/genetics , Endoplasmic Reticulum Stress/physiology , Endoribonucleases/genetics , Lymphocyte Activation/immunology , Natural Killer T-Cells/immunology , Protein Serine-Threonine Kinases/genetics , Animals , Cells, Cultured , Colitis/genetics , Gene Deletion , Mice , Mice, Knockout , Oxazolone/toxicity , RNA, Messenger/genetics , Signal Transduction , Unfolded Protein Response/genetics , Unfolded Protein Response/physiology , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Nat Rev Rheumatol ; 12(7): 398-411, 2016 07.
Article in English | MEDLINE | ID: mdl-27305853

ABSTRACT

The human gut harbours a tremendously diverse and abundant microbial community that correlates with, and even modulates, many health-related processes. The mucosal interfaces are particularly active sites of microorganism-host interplay. Growing insight into the characteristic composition and functionality of the mucosal microbiota has revealed that the microbiota is involved in mucosal barrier integrity and immune function. This involvement affects proinflammatory and anti-inflammatory processes not only at the epithelial level, but also at remote sites such as the joints. Here, we review the role of the gut microbiota in shaping local and systemic immune responses and how disturbances in the host-microorganism interplay can potentially affect the development and progression of rheumatic diseases. Increasing our understanding of how to promote host-microorganism homeostasis could therefore reveal novel strategies for the prevention or alleviation of rheumatic disease.


Subject(s)
Gastrointestinal Microbiome/immunology , Homeostasis/immunology , Probiotics/therapeutic use , Rheumatic Diseases/drug therapy , Rheumatic Diseases/immunology , Animals , Arthritis, Rheumatoid/immunology , Dysbiosis/immunology , Evidence-Based Medicine , Humans , Lupus Erythematosus, Systemic/immunology , Microbiota/immunology , Rheumatic Diseases/microbiology , Rheumatic Diseases/pathology , Spondylarthritis/immunology , Treatment Outcome
17.
J Ambul Care Manage ; 39(4): 333-42, 2016.
Article in English | MEDLINE | ID: mdl-27576054

ABSTRACT

We compared prospective risk adjustment models for adjusting patient panels at the San Francisco Department of Public Health. We used 4 statistical models (linear regression, two-part model, zero-inflated Poisson, and zero-inflated negative binomial) and 4 subsets of predictor variables (age/gender categories, chronic diagnoses, homelessness, and a loss to follow-up indicator) to predict primary care visit frequency. Predicted visit frequency was then used to calculate patient weights and adjusted panel sizes. The two-part model using all predictor variables performed best (R = 0.20). This model, designed specifically for safety net patients, may prove useful for panel adjustment in other public health settings.


Subject(s)
Models, Statistical , Patient Satisfaction , Primary Health Care/standards , Public Health , Risk Adjustment , Adult , Female , Humans , Male , Prospective Studies , Regression Analysis , Safety-net Providers , San Francisco
18.
J Exp Med ; 213(10): 1973-81, 2016 09 19.
Article in English | MEDLINE | ID: mdl-27551157

ABSTRACT

Natural killer T (NKT) cells are innate lymphocytes that differentiate into NKT1, NKT2, and NKT17 sublineages during development. However, the signaling events that control NKT sublineage specification and differentiation remain poorly understood. Here, we demonstrate that the ubiquitin-modifying enzyme TNFAIP3/A20, an upstream regulator of T cell receptor (TCR) signaling in T cells, is an essential cell-intrinsic regulator of NKT differentiation. A20 is differentially expressed during NKT cell development, regulates NKT cell maturation, and specifically controls the differentiation and survival of NKT1 and NKT2, but not NKT17, sublineages. Remaining A20-deficient NKT1 and NKT2 thymocytes are hyperactivated in vivo and secrete elevated levels of Th1 and Th2 cytokines after TCR ligation in vitro. Defective NKT development was restored by compound deficiency of MALT1, a key downstream component of TCR signaling in T cells. These findings therefore show that negative regulation of TCR signaling during NKT development controls the differentiation and survival of NKT1 and NKT2 cells.


Subject(s)
Cell Lineage , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/metabolism , Ubiquitin/metabolism , Animals , CD4 Antigens/metabolism , Caspases/deficiency , Caspases/metabolism , Cell Survival , Integrases/metabolism , Lymphocyte Activation/immunology , Mice, Inbred C57BL , Mice, Transgenic , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tumor Necrosis Factor alpha-Induced Protein 3/genetics , Up-Regulation/genetics
19.
J Vis Exp ; (105): e53256, 2015 Oct 29.
Article in English | MEDLINE | ID: mdl-26555769

ABSTRACT

The ability to rapidly secrete cytokines upon stimulation is a functional characteristic of the invariant natural killer T (iNKT) cell lineage. iNKT cells are therefore characterized as an innate T cell population capable of activating and steering adaptive immune responses. The development of improved techniques for the culture and expansion of murine iNKT cells facilitates the study of iNKT cell biology in in vitro and in vivo model systems. Here we describe an optimized procedure for the isolation and expansion of murine splenic iNKT cells. Spleens from C57Bl/6 mice are removed, dissected and strained and the resulting cellular suspension is layered over density gradient media. Following centrifugation, splenic mononuclear cells (MNCs) are collected and CD5-positive (CD5(+)) lymphocytes are enriched for using magnetic beads. iNKT cells within the CD5(+) fraction are subsequently stained with αGalCer-loaded CD1d tetramer and purified by fluorescence activated cell sorting (FACS). FACS sorted iNKT cells are then initially cultured in vitro using a combination of recombinant murine cytokines and plate-bound T cell receptor (TCR) stimuli before being expanded in the presence of murine recombinant IL-7. Using this technique, approximately 10(8) iNKT cells can be generated within 18-20 days of culture, after which they can be used for functional assays in vitro, or for in vivo transfer experiments in mice.


Subject(s)
Flow Cytometry/methods , Natural Killer T-Cells/cytology , Spleen/cytology , Animals , Cytokines/immunology , Female , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Natural Killer T-Cells/immunology , Receptors, Antigen, T-Cell/immunology , Spleen/immunology
20.
Microbes Infect ; 6(10): 946-59, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15310472

ABSTRACT

The control of Mycobacterium tuberculosis infection depends on recognition of the pathogen and the activation of both the innate and adaptive immune responses. Toll-like receptors (TLR) were shown to play a critical role in the recognition of several pathogens. Mycobacterial antigens recognise distinct TLR resulting in rapid activation of cells of the innate immune system. Recent evidence from in vitro and in vivo investigations, summarised in this review demonstrates TLR-dependent activation of innate immune response, while the induction of adaptive immunity to mycobacteria may be TLR independent.


Subject(s)
Lymphocyte Activation/immunology , Membrane Glycoproteins/immunology , Mycobacterium tuberculosis/immunology , Receptors, Cell Surface/immunology , Tuberculosis/immunology , Animals , Apoptosis/immunology , Humans , Lipopolysaccharides/immunology , Membrane Glycoproteins/chemistry , Receptors, Cell Surface/chemistry , Signal Transduction/immunology , Toll-Like Receptors , Tuberculosis/microbiology
SELECTION OF CITATIONS
SEARCH DETAIL