Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Clin Transl Med ; 13(11): e1471, 2023 11.
Article in English | MEDLINE | ID: mdl-37962000

ABSTRACT

BACKGROUND: The NLRP3 inflammasome drives release of pro-inflammatory cytokines including interleukin (IL)-1ß and IL-18 and is a potential target for ulcerative colitis (UC). Selnoflast (RO7486967) is an orally active, potent, selective and reversible small molecule NLRP3 inhibitor. We conducted a randomized, placebo-controlled Phase 1b study to assess the safety, tolerability, pharmacokinetics (PK) and pharmacodynamics (PD) of selnoflast. METHODS: Nineteen adults with previous diagnosis of UC and current active moderate to severe disease were randomized 2:1 to selnoflast or placebo for 7 days. A dose of 450 mg QD (once daily) was selected to achieve 90% IL-1ß inhibition in plasma and colon tissue. Consecutive blood, sigmoid colon biopsies and stool samples were analyzed for a variety of PD markers. Safety and PK were also evaluated. RESULTS: Selnoflast was well-tolerated. Plasma concentrations increased rapidly after oral administration, reaching Tmax 1 h post-dose. Mean plasma concentrations stayed above the IL-1ß IC90 level throughout the dosing interval (mean Ctrough on Day 1 and Day 5: 2.55 µg/mL and 2.66 µg/mL, respectively). At steady state, post-dose selnoflast concentrations in sigmoid colon (5-20 µg/g) were above the IC90 . Production of IL-1ß was reduced in whole blood following ex vivo stimulation with lipopolysaccharide (LPS) (in the selnoflast arm). No changes were observed in plasma IL-18 levels. There were no meaningful differences in the expression of an IL-1-related gene signature in sigmoid colon tissue, and no differences in the expression of stool biomarkers. CONCLUSIONS: Selnoflast was safe and well-tolerated. Selnoflast 450 mg QD achieved plasma and tissue exposure predicted to maintain IL-1ß IC90 over the dosing interval. However, PD biomarker results showed no robust differences between treatment arms, suggesting no major therapeutic effects are to be expected in UC. The limitations of this study are its small sample size and indirect assessment of the effect on IL-1ß in tissue. TRIAL REGISTRATION: ISRCTN16847938.


Subject(s)
Colitis, Ulcerative , Adult , Humans , Colitis, Ulcerative/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-18/therapeutic use , Inflammasomes/metabolism , Cytokines/metabolism , Biomarkers
2.
bioRxiv ; 2023 Jul 01.
Article in English | MEDLINE | ID: mdl-37425769

ABSTRACT

Extraintestinal autoimmune diseases are multifactorial with translocating gut pathobionts implicated as instigators and perpetuators in mice. However, the microbial contributions to autoimmunity in humans remain largely unclear, including whether specific pathological human adaptive immune responses are triggered by such pathobionts. We show here that the translocating pathobiont Enterococcus gallinarum induces human IFNγ + Th17 differentiation and IgG3 subclass switch of anti- E. gallinarum RNA and correlating anti-human RNA autoantibody responses in patients with systemic lupus erythematosus and autoimmune hepatitis. Human Th17 induction by E. gallinarum is cell-contact dependent and involves TLR8-mediated human monocyte activation. In murine gnotobiotic lupus models, E. gallinarum translocation triggers IgG3 anti-RNA autoantibody titers that correlate with renal autoimmune pathophysiology and with disease activity in patients. Overall, we define cellular mechanisms of how a translocating pathobiont induces human T- and B-cell-dependent autoimmune responses, providing a framework for developing host- and microbiota-derived biomarkers and targeted therapies in extraintestinal autoimmune diseases. One Sentence Summary: Translocating pathobiont Enterococcus gallinarum promotes human Th17 and IgG3 autoantibody responses linked to disease activity in autoimmune patients.

3.
J Med Chem ; 65(17): 11533-11549, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35969159

ABSTRACT

The chemokine receptor CXCR3 allows the selective recruitment of innate and adaptive inflammatory immune cells into inflamed tissue. CXCR3 ligands are secreted after exposure to pro-inflammatory cytokines. Upon binding to CXCR3 ligands, CXCR3 expressing T-lymphocytes migrate toward sites of inflammation and can promote tissue damage. Therefore, antagonizing this receptor may provide clinical benefits for patients suffering from autoimmune diseases characterized by high concentrations of CXCR3 ligands. Herein, we report the second part of our CXCR3 discovery program where we explored the benzimidazolo-thiazole core scaffold. The optimization of potency and the mitigation of an hERG liability are described. Further pharmacokinetic considerations led to the identification of the potent CXCR3 antagonist ACT-672125 (29). The compound showed good physicochemical properties and safety profile. In a proof-of-mechanism model of lung inflammation, ACT-672125 inhibited the recruitment of CXCR3 expressing T cells into the inflamed lung in a dose-dependent manner.


Subject(s)
Autoimmune Diseases , Thiazoles , Autoimmune Diseases/drug therapy , Cytokines , Humans , Ligands , Receptors, CXCR3/metabolism , T-Lymphocytes/metabolism , Thiazoles/pharmacology , Thiazoles/therapeutic use
4.
J Med Chem ; 65(17): 11513-11532, 2022 09 08.
Article in English | MEDLINE | ID: mdl-35947786

ABSTRACT

The chemokine receptor CXCR3 is a seven-transmembrane G-protein-coupled receptor (GPCR) involved in various pathologies, in particular autoimmune diseases. It is activated by the three chemokine ligands CXCL9, CXCL10, and CXCL11 and enables the recruitment of immune cell subsets leading to damage of inflamed tissues. Starting from a high-throughput screening hit, we describe the iterative optimization of a chemical series culminating in the discovery of the selective CXCR3 antagonist ACT-660602 (9j). The careful structural modifications during the lead optimization phase led to a compound with high biological potency in inhibiting cell migration together with improvements of the metabolic stability and hERG issue. In a LPS-induced lung inflammation model in mice, ACT-660602 led to significantly reduced recruitment of the CXCR3+ CD8+ T cell in the bronchoalveolar lavage compartment when administered orally at a dose of 30 mg/kg.


Subject(s)
Autoimmune Diseases , Chemokine CXCL10 , Animals , Autoimmune Diseases/drug therapy , CD8-Positive T-Lymphocytes/metabolism , Chemokine CXCL9/metabolism , Ligands , Mice , Receptors, CXCR3/metabolism
5.
J Transl Med ; 19(1): 245, 2021 06 05.
Article in English | MEDLINE | ID: mdl-34090480

ABSTRACT

In the era of precision medicine, digital technologies and artificial intelligence, drug discovery and development face unprecedented opportunities for product and business model innovation, fundamentally changing the traditional approach of how drugs are discovered, developed and marketed. Critical to this transformation is the adoption of new technologies in the drug development process, catalyzing the transition from serendipity-driven to data-driven medicine. This paradigm shift comes with a need for both translation and precision, leading to a modern Translational Precision Medicine approach to drug discovery and development. Key components of Translational Precision Medicine are multi-omics profiling, digital biomarkers, model-based data integration, artificial intelligence, biomarker-guided trial designs and patient-centric companion diagnostics. In this review, we summarize and critically discuss the potential and challenges of Translational Precision Medicine from a cross-industry perspective.


Subject(s)
Artificial Intelligence , Precision Medicine , Biomarkers , Drug Discovery , Humans , Translational Research, Biomedical
6.
Innov Clin Neurosci ; 16(3-4): 22-30, 2019 Mar 01.
Article in English | MEDLINE | ID: mdl-31214480

ABSTRACT

Background: Despite the recent approval of new oral therapies for the treatment of multiple sclerosis (MS), a significant percentage of patients are still not free from disease activity. In view of the complex pathogenesis and the relapsing and progressive nature of MS, combination therapy, a classical approach to treat many chronic diseases, could improve disease control over monotherapy. Ponesimod, a selective and rapidly reversible sphingosine-1-phosphate receptor Type 1 (S1P1) modulator, currently in Phase III clinical trial stage in relapsing MS (RMS), and dimethyl fumarate (DMF) would potentially be an ideal combination due to their differing mechanisms of action and oral administration. Objective: The goal of the study was to evaluate the therapeutic effect of ponesimod monotherapy and investigate the potential additive, or synergistic, activity of ponesimod-DMF combination therapy in experimental autoimmune encephalomyelitis (EAE) animal models of MS. Methods: Efficacy was evaluated in the myelin oligodendrocyte glycoprotein (MOG)-induced EAE model in C57BL/6 mice (ponesimod monotherapy) and in the myelin basic protein (MBP)-induced EAE model in Lewis rats (monotherapies and combination therapy). The principal readout was the clinical score assessing paralysis. Additional readouts, such as histopathology, survival, and disease prevalence, were generated in parallel when applicable. Results: Ponesimod monotherapy in the mouse EAE model showed significant efficacy in both preventative and therapeutic settings. In the rat EAE model, ponesimod demonstrated significant dose-dependent efficacy on clinical scores, while DMF showed only modest activity. Combination therapy synergistically reduced the severity and prevalence of disease. Only the combination treatment of ponesimod and DMF fully suppressed clinical disease activity by the end of the study. Conclusion: The results support the potential therapeutic benefits of combining ponesimod with DMF to improve disease activity control in patients with MS. Additionally, the results suggest that combining ponesimod with other oral agents that have different mechanisms of action might also be therapeutically beneficial to patients with MS.

7.
Eur J Immunol ; 49(3): 462-475, 2019 03.
Article in English | MEDLINE | ID: mdl-30578679

ABSTRACT

Immune checkpoints are regulators of immune cells and play key roles in the modulation of immune responses. The role of checkpoints in autoimmune disease is poorly understood but likely to be central since checkpoint inhibition during cancer treatment can cause autoimmunity. We generated a high-dimensional single-cell proteomics data set from PBMCs of healthy individuals and patients with ulcerative colitis (UC) by mass cytometry, enabling systems-wide analyses of immune cell frequencies and cell type-specific expression patterns of 12 immune checkpoints. Subtle but significant changes in immune cell frequencies and checkpoint expression were observed between UC patients on different treatment regimens and between patients and healthy controls. Most strikingly, UC patients showed a reduced number of peripheral NK-cells and those cells showed an altered phenotype including increased TIGIT expression. Based on these results, we modulated NK-cell function ex vivo through targeting of TIGIT pathway members. In summary, we describe a pattern of changes in immune cell abundance and checkpoint expression as a basis for UC patient stratification and we show modulation of a corresponding immune cell subset through checkpoint targeting. Our approach can be used for the identification of pathogenic immune cell subsets and guide target selection in autoimmunity and chronic inflammation.


Subject(s)
Colitis, Ulcerative/metabolism , Killer Cells, Natural/metabolism , Proteomics/methods , Single-Cell Analysis/methods , Adult , Aged , Aged, 80 and over , Aminosalicylic Acid/therapeutic use , Antibodies, Monoclonal, Humanized/therapeutic use , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/genetics , Female , Gastrointestinal Agents/therapeutic use , Humans , Immunosuppressive Agents/therapeutic use , Infliximab/therapeutic use , Killer Cells, Natural/drug effects , Male , Middle Aged , Receptors, Immunologic/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/metabolism
8.
Article in English | MEDLINE | ID: mdl-29226621

ABSTRACT

Sphingosine-1-phosphate receptor 1 (S1P1 ) modulators sequester circulating lymphocytes within lymph nodes, thereby preventing potentially pathogenic autoimmune cells from exiting into the blood stream and reaching inflamed tissues. S1P1 receptor modulation may thus offer potential to treat various autoimmune diseases. The first nonselective S1P1-5 receptor modulator FTY720/fingolimod/Gilenya® has successfully demonstrated clinical efficacy in relapsing forms of multiple sclerosis. However, cardiovascular, hepatic, and respiratory side-effects were reported and there is a need for novel S1P1 receptor modulators with better safety profiles. Here, we describe the discovery of cenerimod, a novel, potent and selective S1P1 receptor modulator with unique S1P1 receptor signaling properties and absence of broncho- and vasoconstrictor effects ex vivo and in vivo. Cenerimod dose-dependently lowered circulating lymphocyte counts in rats and mice after oral administration and effectively attenuated disease parameters in a mouse experimental autoimmune encephalitis (EAE) model. Cenerimod has potential as novel therapy with improved safety profile for autoimmune diseases with high unmet medical need.


Subject(s)
Encephalomyelitis, Autoimmune, Experimental/drug therapy , Immunosuppressive Agents/administration & dosage , Lymphocytes/drug effects , Oxadiazoles/administration & dosage , Pyridines/administration & dosage , Receptors, Lysosphingolipid/agonists , Administration, Oral , Animals , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , Encephalomyelitis, Autoimmune, Experimental/immunology , Encephalomyelitis, Autoimmune, Experimental/metabolism , Human Umbilical Vein Endothelial Cells , Humans , Immunosuppressive Agents/chemistry , Immunosuppressive Agents/pharmacology , Lymphocyte Count , Mice , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Pyridines/chemistry , Pyridines/pharmacology , Rats , Receptors, Lysosphingolipid/metabolism , Signal Transduction/drug effects
9.
Immunol Lett ; 192: 1-6, 2017 12.
Article in English | MEDLINE | ID: mdl-28987474

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) are a heterogeneous cell population expanded in cancer, infection and autoimmunity capable of suppressing T-cell functions. Checkpoint inhibitors have emerged as a key therapeutic strategy in immune-oncology. While checkpoint molecules were initially associated with T cell functions, recent evidence suggests a broader expression and function in innate myeloid cells. Previous studies provided first evidence for a potential role for checkpoints on MDSCs, yet the human relevance remained poorly understood. Therefore, we investigated the expression and functional relevance of checkpoint molecules in human MDSC-T-cell interactions. Our studies demonstrate that programmed death-ligand 1 (PD-L1) is expressed on granulocytic MDSCs upon co-culture with T cells. Transwell experiments showed that cell-to-cell contact was required for MDSC-T-cell interactions and antibody blocking studies showed that targeting PD-L1 partially impaired MDSC-mediated T-cell suppression. Collectively, these studies suggest a role for PD-L1 in human MDSC function and thereby expand the functionality of this checkpoint beyond T cells, which could pave the way for further understanding and therapeutic targeting of PD-1/PD-L1 in innate immune-mediated diseases.


Subject(s)
B7-H1 Antigen/metabolism , Immunotherapy/methods , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes/immunology , Antibodies, Blocking/pharmacology , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Cell Communication , Cells, Cultured , Coculture Techniques , Humans , Immune Tolerance , Immunity, Innate , Molecular Targeted Therapy
10.
PLoS One ; 8(10): e77296, 2013.
Article in English | MEDLINE | ID: mdl-24204793

ABSTRACT

In the present study, we investigated the therapeutic potential of a selective S1P1 receptor modulator, ponesimod, to protect and reverse autoimmune diabetes in non-obese diabetic (NOD) mice. Ponesimod was administered orally to NOD mice starting at 6, 10, 13 and 16 weeks of age up to 35 weeks of age or to NOD mice showing recent onset diabetes. Peripheral blood and spleen B and T cell counts were significantly reduced after ponesimod administration. In pancreatic lymph nodes, B lymphocytes were increased and expressed a transitional 1-like phenotype. Chronic oral ponesimod treatment efficiently prevented autoimmune diabetes in 6, 10 and 16 week-old pre-diabetic NOD mice. Treatment withdrawal led to synchronized disease relapse. Ponesimod did not inhibit the differentiation of autoreactive T cells as assessed by adoptive transfer of lymphocytes from treated disease-free NOD mice. In addition, it did not affect the migration, proliferation and activation of transgenic BDC2.5 cells into the target tissue. However, ponesimod inhibited spreading of the T cell responses to islet-specific glucose-6-phosphatase catalytic subunit-related protein (IGRP). Treatment of diabetic NOD mice with ponesimod induced disease remission. However, here again, upon treatment cessation, the disease rapidly recurred. This recurrence was effectively prevented by combination treatment with a CD3 antibody leading to the restoration of self-tolerance. In conclusion, treatment with a selective S1P1 modulator in combination with CD3 antibody represents a promising therapeutic approach for the treatment of autoimmune diabetes.


Subject(s)
Antibodies/pharmacology , Diabetes Mellitus, Type 1/drug therapy , Hypoglycemic Agents/pharmacology , Receptors, Lysosphingolipid/immunology , Thiazoles/pharmacology , Administration, Oral , Adoptive Transfer , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , CD3 Complex/genetics , CD3 Complex/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/pathology , Drug Administration Schedule , Gene Expression , Glucose-6-Phosphatase/antagonists & inhibitors , Glucose-6-Phosphatase/genetics , Glucose-6-Phosphatase/immunology , Lymph Nodes/drug effects , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Count , Mice , Mice, Inbred NOD , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/genetics , Recurrence , Self Tolerance , Spleen/drug effects , Spleen/immunology , Spleen/pathology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/pathology
12.
J Pharmacol Exp Ther ; 337(2): 547-56, 2011 May.
Article in English | MEDLINE | ID: mdl-21345969

ABSTRACT

Lymphocyte exit from lymph nodes and their recirculation into blood is controlled by the sphingolipid sphingosine 1-phosphate (S1P). The cellular receptor mediating lymphocyte exit is S1P(1), one of five S1P receptors. Nonselective agonists for S1P receptors lead to blood lymphocyte count reduction. The effects of selective S1P(1) agonists on blood lymphocyte count and their impact in models of lymphocyte-mediated tissue inflammation have been less investigated. We describe here the general pharmacology of ponesimod, (Z,Z)-5-[3-chloro-4-((2R)-2,3-dihydroxy-propoxy)-benzylidene]-2-propylimino-3-o-tolyl-thiazolidin-4-one, a new, potent, and orally active selective S1P(1) agonist. Ponesimod activated S1P(1)-mediated signal transduction with high potency (EC(50) of 5.7 nM) and selectivity. Oral administration of ponesimod to rats led to a dose-dependent decrease of blood lymphocyte count. After discontinuation of dosing, blood lymphocyte count returned to baseline within 48 h. Ponesimod prevented edema formation, inflammatory cell accumulation, and cytokine release in the skin of mice with delayed-type hypersensitivity. Ponesimod also prevented the increase in paw volume and joint inflammation in rats with adjuvant-induced arthritis. These data show that selective activation of S1P(1) using ponesimod leads to blood lymphocyte count reduction and efficacy in models of lymphocyte-mediated tissue inflammation. Immunomodulation with a rapidly reversible S1P(1)-selective agonist may represent a new therapeutic approach in lymphocyte-mediated autoimmune diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Inflammation/pathology , Inflammation/prevention & control , Lymphocytes/drug effects , Lymphocytes/physiology , Receptors, Lysosphingolipid/agonists , Thiazoles/pharmacology , Animals , Arthritis, Experimental/drug therapy , CHO Cells , Cricetinae , Cricetulus , Dose-Response Relationship, Drug , Guanosine 5'-O-(3-Thiotriphosphate)/metabolism , Hypersensitivity, Delayed/immunology , Lymphocyte Count , Lymphocyte Subsets/drug effects , Male , Mice , Mice, Inbred BALB C , Monocytes/drug effects , Rats , Rats, Inbred Lew , Rats, Wistar , Thiazoles/blood
13.
Blood ; 105(1): 31-9, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15358618

ABSTRACT

During embryonic development, T-lymphoid precursor cells colonize the thymus. Chemoattraction by the fetal thymus is thought to mediate T-precursor cell colonization. However, the molecules that attract T-precursor cells to the thymus remain unclear. By devising time-lapse visualization in culture, the present results show that alymphoid fetal thymus lobes attract T-precursor cells from fetal liver or fetal blood. CD4(-)CD8(-)CD25(-)CD44+ fetal thymocytes retained the activity to specifically re-enter the thymus. The attraction was predominantly due to I-A-expressing thymic epithelial cells and was mediated by pertussis toxin-sensitive G-protein signals. Among the chemokines produced by the fetal thymus, CCL21, CCL25, and CXCL12 could attract CD4(-)CD8(-)CD25(-)CD44+ fetal thymocytes. However, fetal thymus colonization was markedly diminished by neutralizing antibodies specific for CCL21 and CCL25, but not affected by anti-CXCL12 antibody. Fetal thymus colonization was partially defective in CCL21-deficient plt/plt mice and was further diminished by anti-CCL25 antibody. These results indicate that CCL21 is involved in the recruitment of T-cell precursors to the fetal thymus and suggest that the combination of CCL21 and CCL25 plays a major role in fetal thymus colonization.


Subject(s)
Chemokines, CC/metabolism , Stem Cells/cytology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymus Gland/embryology , Thymus Gland/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Cells, Cultured , Chemokine CCL21 , Chemokine CXCL12 , Chemokines, CC/antagonists & inhibitors , Chemokines, CC/immunology , Chemokines, CXC/immunology , Chemokines, CXC/metabolism , Chemotaxis, Leukocyte , Fetal Blood/cytology , Fetus/embryology , Fetus/immunology , Fetus/metabolism , Histocompatibility Antigens Class II/immunology , Liver/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/immunology , T-Lymphocytes/metabolism , Thymus Gland/cytology , Thymus Gland/metabolism , Time Factors
14.
Immunology ; 108(3): 365-74, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12603603

ABSTRACT

Granuloma is a typical feature of tuberculosis. We evaluated the chemotaxis of selected human leucocyte subsets induced by macrophages incubated with Mycobacterium tuberculosis (MT)-derived products in vitro. The release of monocyte chemotactic protein 1 (MCP-1) and interleukin-8 (IL-8) correlated with the specific induction of strong chemotaxis towards monocytes and polymorphonuclear leucocytes (PMNs). gammadelta and T helper type 1 (Th1) alphabeta lymphocytes were chemoattracted, while T-resting, IL-2-activated and Th2 lymphocytes were unaffected. Activation with mycobacterium-derived, phosphate-containing components, modulated the chemokine receptor profile of gammadelta T lymphocytes as well as their pattern of cyto-chemokine production, disclosing a potential for their active participation in granuloma formation. In particular, CXCR3 and IP-10, which we found to be released by MT-pulsed alveolar macrophages, seem to represent the receptor-counter-receptor pair implicated in the chemotaxis of gammadelta lymphocytes. Immunohistochemical analysis and in situ hybridization revealed the in vivo presence of IL-8, MCP-1 and IL-10 in lymph node and lung tuberculous granulomas. Our results underscore the role of MT extracts in the induction of macrophage-derived chemokines responsible for the orchestrated recruitment of PMNs, monocytes, and Th1 and gammadelta T cells, as well as in the regulation of gammadelta function.


Subject(s)
Chemokines/metabolism , Macrophages, Alveolar/immunology , Mycobacterium tuberculosis/immunology , Receptors, Antigen, T-Cell, gamma-delta/analysis , T-Lymphocyte Subsets/immunology , Antigens, Bacterial/immunology , Cells, Cultured , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Chemotaxis, Leukocyte/immunology , Gene Expression , Granuloma/immunology , Humans , Interleukin-8/genetics , Interleukin-8/metabolism , Lung/immunology , Lymph Nodes/immunology , Monocytes/immunology , Neutrophils/immunology , RNA, Messenger/genetics
15.
Nat Med ; 8(12): 1414-20, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12415259

ABSTRACT

T cell-mediated loss of insulin-secreting beta cells in the islets of Langerhans is the hallmark of type 1 diabetes. The molecular basis for the directed migration of autoreactive T cells leading to insulitis is presently unknown. Here we demonstrate that in response to inflammation, beta cells secrete the chemokines CXC ligand 10 and CXC ligand 9, which specifically attract T-effector cells via the CXC chemokine receptor 3. In mice deficient for this receptor, the onset of type 1 diabetes is substantially delayed. Thus, in the absence of known etiological agents, CXC receptor 3 represents a novel target for therapeutic interference early in type 1 diabetes.


Subject(s)
Islets of Langerhans/immunology , Islets of Langerhans/pathology , Receptors, Chemokine/physiology , T-Lymphocytes/physiology , Animals , Cells, Cultured , Chemokine CXCL10 , Chemokines, CXC/physiology , Diabetes Mellitus, Type 1/etiology , Inflammation/etiology , Inflammation/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Receptors, CXCR3
16.
Nat Immunol ; 3(11): 1102-8, 2002 Nov.
Article in English | MEDLINE | ID: mdl-12379851

ABSTRACT

T cell development and selection require the fully mature and diverse epithelial microenvironment of the thymus. Acquisition of these characteristics is dependent on expression of the forkhead (also known as winged-helix) transcription factor FoxN1, as a lack of functional FoxN1 results in aberrant epithelial morphogenesis and an inability to attract lymphoid precursors to the thymus primordium. However, the transcriptional control of Foxn1 expression has not been elucidated. Here we report that secreted Wnt glycoproteins, expressed by thymic epithelial cells and thymocytes, regulate epithelial Foxn1 expression in both autocrine and paracrine fashions. Wnt molecules therefore provide regulatory signals critical for thymic function.


Subject(s)
DNA-Binding Proteins/biosynthesis , Gene Expression Regulation , Glycoproteins/physiology , Lymphopoiesis/genetics , Protein Serine-Threonine Kinases , Proteins/physiology , Proto-Oncogene Proteins/physiology , Receptors, G-Protein-Coupled , T-Lymphocytes/cytology , Transcription Factors/biosynthesis , Transcription Factors/physiology , Xenopus Proteins , Zebrafish Proteins , 3T3 Cells , Adaptor Proteins, Signal Transducing , Animals , Autocrine Communication , Cell Differentiation , Chromones/pharmacology , Cytoskeletal Proteins/biosynthesis , Cytoskeletal Proteins/genetics , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dishevelled Proteins , Enzyme Inhibitors/pharmacology , Epithelial Cells/metabolism , Epithelial Cells/ultrastructure , Forkhead Transcription Factors , Frizzled Receptors , Genes, Reporter , Glycoproteins/biosynthesis , Glycoproteins/genetics , Lymphoid Enhancer-Binding Factor 1 , Mice , Mice, Nude , Morphogenesis , Morpholines/pharmacology , Paracrine Communication , Phosphatidylinositol 3-Kinases/physiology , Phosphoinositide-3 Kinase Inhibitors , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Phosphorylation , Protein Biosynthesis , Protein Processing, Post-Translational , Proteins/genetics , Proto-Oncogene Proteins/biosynthesis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt , Receptors, Cell Surface/biosynthesis , Receptors, Cell Surface/genetics , Recombinant Fusion Proteins/physiology , TCF Transcription Factors , Thymus Gland/cytology , Thymus Gland/metabolism , Trans-Activators/biosynthesis , Trans-Activators/genetics , Transcription Factor 7-Like 2 Protein , Transcription Factors/deficiency , Transcription Factors/genetics , Transcription, Genetic , Wnt Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...