Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 103
Filter
1.
PLoS One ; 14(3): e0213203, 2019.
Article in English | MEDLINE | ID: mdl-30845158

ABSTRACT

Although sphingosine-1-phosphate receptor 1 (S1P1) has been shown to trigger several S1P targeted functions such as immune cell trafficking, cell proliferation, migration, or angiogenesis, tools that allow the accurate detection of endogenous S1P1 localization and trafficking remain to be obtained and validated. In this study, we developed and characterized a novel monoclonal S1P1 antibody. Mice were immunized with S1P1 produced in the yeast Pichia pastoris and nine hybridoma clones producing monoclonal antibodies were created. Using different technical approaches including Western blot, immunoprecipitation and immunocytochemistry, we show that a selected clone, hereinafter referred to as 2B9, recognizes human and mouse S1P1 in various cell lineages. The interaction between 2B9 and S1P1 is specific over receptor subtypes, as the antibody does not binds to S1P2 or S1P5 receptors. Using cell-imaging methods, we demonstrate that 2B9 binds to an epitope located at the intracellular domain of S1P1; reveals cytosolic and membrane localization of the endogenous S1P1; and receptor internalization upon S1P or FTY720-P stimulation. Finally, loss of 2B9 signal upon knockdown of endogenous S1P1 by specific small interference RNAs further confirms its specificity. 2B9 was also able to detect S1P1 in human kidney and spinal cord tissue by immunohistochemistry. Altogether, our results suggest that 2B9 could be a useful tool to detect, quantify or localize low amounts of endogenous S1P1 in various physiological and pathological processes.


Subject(s)
Antibodies, Monoclonal/immunology , Breast Neoplasms/metabolism , Fibroblasts/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Image Processing, Computer-Assisted/methods , Receptors, Lysosphingolipid/immunology , Receptors, Lysosphingolipid/metabolism , Animals , Breast Neoplasms/pathology , Cells, Cultured , Female , Fibroblasts/cytology , Humans , Immunization , Kidney/metabolism , Mice , Microscopy, Fluorescence , Sphingosine-1-Phosphate Receptors , Spinal Cord/metabolism
2.
Front Immunol ; 9: 2189, 2018.
Article in English | MEDLINE | ID: mdl-30319641

ABSTRACT

About 40% of patients with systemic lupus erythematosus experience diffuse neuropsychiatric manifestations, including impaired cognition and depression. Although the pathogenesis of diffuse neuropsychiatric SLE (NPSLE) is not fully understood, loss of brain barrier integrity, autoreactive antibodies, and pro-inflammatory cytokines are major contributors to disease development. Fingolimod, a sphingosine-1-phosphate (S1P) receptor modulator, prevents lymphocyte egress from lymphoid organs through functional antagonism of S1P receptors. In addition to reducing the circulation of autoreactive lymphocytes, fingolimod has direct neuroprotective effects such as preserving brain barrier integrity and decreasing pro-inflammatory cytokine secretion by astrocytes and microglia. Given these effects, we hypothesized that fingolimod would attenuate neurobehavioral deficits in MRL-lpr/lpr (MRL/lpr) mice, a validated neuropsychiatric lupus model. Fingolimod treatment was initiated after the onset of disease, and mice were assessed for alterations in cognitive function and emotionality. We found that fingolimod significantly attenuated spatial memory deficits and depression-like behavior in MRL/lpr mice. Immunofluorescent staining demonstrated a dramatic lessening of brain T cell and macrophage infiltration, and a significant reduction in cortical leakage of serum albumin, in fingolimod treated mice. Astrocytes and endothelial cells from treated mice exhibited reduced expression of inflammatory genes, while microglia showed differential regulation of key immune pathways. Notably, cytokine levels within the cortex and hippocampus were not appreciably decreased with fingolimod despite the improved neurobehavioral profile. Furthermore, despite a reduction in splenomegaly, lymphadenopathy, and circulating autoantibody titers, IgG deposition within the brain was unaffected by treatment. These findings suggest that fingolimod mediates attenuation of NPSLE through a mechanism that is not dependent on reduction of autoantibodies or cytokines, and highlight modulation of the S1P signaling pathway as a novel therapeutic target in lupus involving the central nervous system.


Subject(s)
Depression/immunology , Fingolimod Hydrochloride/pharmacology , Lupus Vasculitis, Central Nervous System/psychology , Lysophospholipids/metabolism , Signal Transduction/drug effects , Sphingosine/analogs & derivatives , Animals , Astrocytes/drug effects , Astrocytes/immunology , Autoantibodies/immunology , Behavior Observation Techniques , Behavior, Animal/drug effects , Brain/cytology , Brain/immunology , Brain/physiology , Cognition/drug effects , Cognition/physiology , Cytokines/immunology , Depression/drug therapy , Depression/psychology , Disease Models, Animal , Endothelial Cells/drug effects , Endothelial Cells/immunology , Female , Fingolimod Hydrochloride/therapeutic use , Humans , Lupus Vasculitis, Central Nervous System/drug therapy , Lupus Vasculitis, Central Nervous System/genetics , Lupus Vasculitis, Central Nervous System/immunology , Lysophospholipids/immunology , Mice , Mice, Inbred MRL lpr , Microglia/drug effects , Microglia/immunology , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/immunology , Receptors, Lysosphingolipid/metabolism , Signal Transduction/immunology , Sphingosine/immunology , Sphingosine/metabolism , Treatment Outcome
4.
Cell Physiol Biochem ; 48(3): 1188-1200, 2018.
Article in English | MEDLINE | ID: mdl-30045015

ABSTRACT

BACKGROUND/AIMS: Epithelial cells line the intestinal mucosa and form an important barrier for maintaining host health. This study aimed to explore the mechanism of the Sphingosine-1-phosphate (S1P)/Sphingosine-1-phosphate receptor 2 (S1PR2) pathway in intestinal epithelial cells (IECs) that participate in the intestinal barrier function. METHODS: In this study, we constructed a knockout of the S1PR2 gene in mice, and Dextra sulfate sodium (DSS) was used to induce colitis. We isolated IECs from wild type (WT) and S1PR2-/- mice, and the endogenous expression of S1PR2 and Zonula occludens 1 (ZO-1) in IEC were detected by Western blot. Next, the major histocompatibility complex II (MHC-II) expression was analyzed by reverse transcription quantitative real-time (RT-qPCR) and flow cytometry. The in vivo and in vitro intestinal permeability were evaluated by serum fluorescein isothiocyanate (FITC) concentration. The tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and interferon-γ (IFN-γ) levels in cell suspension were analyzed by enzyme-linked immuno sorbent assay (ELISA). A carboxyfluorescein diacetate succinimidyl ester (CFSE) assay was used to detect the T-cell proliferation in a co-culture system. RESULTS: The intestinal mucosal barrier damage in S1PR2-/- mice was more severe than in the WT mice, and there were more CD4+T-cells in the colon tissue of DSS-treated S1PR2-/- mice. Either the mouse colon carcinoma cell line (CT26. WT) or the IECs upregulated MHC-II expression, which then promoted CD4+T-cell proliferation. The S1P/S1PR2 pathway controlled MHC-II expression to regulate CD4+T-cell proliferation via the extracellular signal-regulated kinase (ERK) pathway. In addition, the IFN-γ that was secreted by CD4+T-cells increased DSS-induced damage of intestinal epithelial cell barrier function. ZO-1 expression was increased by S1P in CT26.WT cells, while S1PR2 antagonist JTE-013 expression was downregulated. However, in CT26.WTsi-S1PR2 cells, S1P had no effect on ZO-1 expression. CONCLUSIONS: The S1P/S1PR2 axis in IECs mediated CD4+T-cell activation via the ERK pathway and MHC-II expression to regulate intestinal barrier function.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Intestinal Mucosa/immunology , Lysophospholipids/immunology , Receptors, Lysosphingolipid/immunology , Signal Transduction , Sphingosine/analogs & derivatives , Animals , CD4-Positive T-Lymphocytes/pathology , Cell Communication , Cell Line, Tumor , Cell Membrane Permeability , Cell Proliferation , Cells, Cultured , Colitis/genetics , Colitis/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Female , Intestinal Absorption , Intestinal Mucosa/pathology , MAP Kinase Signaling System , Male , Mice, Inbred C57BL , Mice, Knockout , Receptors, Lysosphingolipid/genetics , Sphingosine/immunology , Sphingosine-1-Phosphate Receptors
5.
Int J Mol Med ; 42(2): 1116-1124, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29749428

ABSTRACT

In patients with chronic rhinosinusitis with nasal polyps (CRSwNP), a relative accumulation of cluster of differentiation (CD)8+ T cells over CD4+ T cells occurs in nasal polyps compared with the peripheral blood. Nasal CD8+ T cells and CD4+ T cells predominantly present an effector memory phenotype. Immunological studies have reported that memory T cells recirculate from the tissues to the peripheral blood and a high percentage of these T cells persist within the tissue. The aim of the present study was to characterize CD69+ sphingosine­1­phosphate receptor 1 (S1PR1)­ tissue resident memory T cells (Trm) in the polyps of patients with CRSwNP. Tissue and blood samples were collected from 10 patients undergoing nasal sinus surgery. Expression of specific extra­ and intracellular molecules were analyzed using multicolor flow cytometry. A significantly higher level of CD8+ T cells than CD4+ T cells was present in nasal polyps, while significantly more CD4+ T cells than CD8+ T cells were detected in the peripheral blood of patients with CRSwNP. The frequency of CD69+ T cells was significantly higher in CD8+ and CD4+ T cells in nasal polyps compared with the peripheral blood. The frequency of CD69+ S1PR1­ Trm was also significantly higher in CD4+ and CD8+ T cells from nasal polyps compared with the peripheral blood. Within polyps, the frequency of CD69+ S1PR1­ Trm was again significantly higher in CD8+ compared with CD4+ T cells. In summary, a significantly higher frequency of CD69+ S1PR1­ T cells was observed in the nasal polyps compared with the peripheral blood in patients with CRSwNP. The results of the present study suggest that local regulation of the immune response occurs within nasal polyps. As such, Trm should be considered a potential stimulus in the pathogenesis of nasal polyps. However, the role of Trm in nasal polyps as a pathogenic trigger of the local inflammatory reaction requires further investigation.


Subject(s)
Antigens, CD/immunology , Antigens, Differentiation, T-Lymphocyte/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lectins, C-Type/immunology , Nasal Polyps/immunology , Sinusitis/immunology , Adult , Antigens, CD/analysis , Antigens, Differentiation, T-Lymphocyte/analysis , CD4-Positive T-Lymphocytes/pathology , CD8-Positive T-Lymphocytes/pathology , Chronic Disease , Female , Humans , Lectins, C-Type/analysis , Lymphocyte Activation , Male , Middle Aged , Nasal Polyps/pathology , Receptors, Lysosphingolipid/analysis , Receptors, Lysosphingolipid/immunology , Sinusitis/pathology , Sphingosine-1-Phosphate Receptors
6.
Nat Commun ; 9(1): 1970, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29773802

ABSTRACT

T cell-mediated responses have been implicated in the development of fibrosis, impaired lymphangiogenesis, and lymphatic dysfunction in secondary lymphedema. Here we show that CD4+ T cells are necessary for lymphedema pathogenesis by utilizing adoptive transfer techniques in CD4 knockout mice that have undergone tail skin and lymphatic excision or popliteal lymph node dissection. We also demonstrate that T cell activation following lymphatic injury occurs in regional skin-draining lymph nodes after interaction with antigen-presenting cells such as dendritic cells. CD4+ T cell activation is associated with differentiation into a mixed T helper type 1 and 2 phenotype, as well as upregulation of adhesion molecules and chemokines that promote migration to the skin. Most importantly, we find that blocking T cell release from lymph nodes using a sphingosine-1-phosphate receptor modulator prevents lymphedema, suggesting that this approach may have clinical utility.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Immunosuppressive Agents/therapeutic use , Lymphedema/immunology , Lymphocyte Activation/immunology , Adoptive Transfer , Animals , CD4 Antigens/genetics , CD4 Antigens/metabolism , CD4-Positive T-Lymphocytes/drug effects , Cell Differentiation/immunology , Dendritic Cells/immunology , Disease Models, Animal , Female , Fingolimod Hydrochloride/pharmacology , Fingolimod Hydrochloride/therapeutic use , Humans , Immunosuppressive Agents/pharmacology , Lymph Nodes/cytology , Lymph Nodes/pathology , Lymphangiogenesis/immunology , Lymphatic Vessels/cytology , Lymphatic Vessels/immunology , Lymphatic Vessels/pathology , Lymphedema/drug therapy , Lymphedema/pathology , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Lysosphingolipid/immunology , Receptors, Lysosphingolipid/metabolism , Skin/cytology , Skin/immunology
7.
Cell Microbiol ; 20(5): e12836, 2018 05.
Article in English | MEDLINE | ID: mdl-29498184

ABSTRACT

Sphingosine-1-phosphate (S1P) is a signalling lipid that regulates many cellular processes in mammals. One well-studied role of S1P signalling is to modulate T-cell trafficking, which has a major impact on adaptive immunity. Compounds that target S1P signalling pathways are of interest for immune system modulation. Recent studies suggest that S1P signalling regulates many more cell types and processes than previously appreciated. This review will summarise current understanding of S1P signalling, focusing on recent novel findings in the roles of S1P receptors in innate immunity.


Subject(s)
Immunity, Innate/genetics , Inflammation/immunology , Receptors, Lysosphingolipid/genetics , T-Lymphocytes/immunology , Animals , Cell Movement , Humans , Lysophospholipids/genetics , Lysophospholipids/metabolism , Receptors, Lysosphingolipid/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Sphingosine/analogs & derivatives , Sphingosine/genetics , Sphingosine/metabolism
8.
J Immunol ; 200(5): 1580-1592, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29367208

ABSTRACT

Chronic inflammatory demyelinating polyneuropathy (CIDP) is a debilitating condition caused by autoimmune demyelination of peripheral nerves. CIDP is associated with increased IL-10, a cytokine with well-described anti-inflammatory effects. However, the role of IL-10 in CIDP is unclear. In this study, we demonstrate that IL-10 paradoxically exacerbates autoimmunity against peripheral nerves. In IL-10-deficient mice, protection from neuropathy was associated with an accrual of highly activated CD4+ T cells in draining lymph nodes and absence of infiltrating immune cells in peripheral nerves. Accumulated CD4+ T cells in draining lymph nodes of IL-10-deficient mice expressed lower sphingosine-1-phosphate receptor 1 (S1pr1), a protein important in lymphocyte egress. Additionally, IL-10 stimulation in vitro induced S1pr1 expression in lymph node cells in a STAT3-dependent manner. Together, these results delineate a novel mechanism in which IL-10-induced STAT3 increases S1pr1 expression and CD4+ T cell migration to accelerate T cell-mediated destruction of peripheral nerves.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cell Movement/immunology , Interleukin-10/immunology , Neuritis, Autoimmune, Experimental/immunology , Receptors, Lysosphingolipid/immunology , Animals , Demyelinating Diseases/immunology , Female , Lymph Nodes/immunology , Lymphocyte Activation/immunology , Mice , STAT3 Transcription Factor/immunology , Sphingosine-1-Phosphate Receptors
9.
World J Gastroenterol ; 24(47): 5322-5330, 2018 Dec 21.
Article in English | MEDLINE | ID: mdl-30598577

ABSTRACT

Ulcerative colitis (UC), a chronic, relapsing, remitting disease of the colon and rectum, is characterized by inflammatory ulceration of the mucosa. Current UC therapy relies on controlling acute episodes and preventing relapse. To predict modifications in the natural course of UC, mucosal healing (MH) has emerged as a major treatment goal. Endoscopic evaluation is considered the gold standard for assessing MH, which can be achieved by conventional drugs and biologics in many, but not all, patients. Consequently, interest is focusing on the development of new substances for UC therapy, and new oral agents are in the pipeline. This review will focus on the ability of newly developed oral drugs to induce and maintain MH in UC patients.


Subject(s)
Colitis, Ulcerative/drug therapy , Colon/drug effects , Gastrointestinal Agents/therapeutic use , Intestinal Mucosa/drug effects , Administration, Oral , Colitis, Ulcerative/diagnostic imaging , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Colon/diagnostic imaging , Colon/immunology , Colon/pathology , Colonoscopy , Gastrointestinal Agents/pharmacology , Humans , Integrin alpha4/antagonists & inhibitors , Integrin alpha4/immunology , Intestinal Mucosa/diagnostic imaging , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Janus Kinase Inhibitors/pharmacology , Janus Kinase Inhibitors/therapeutic use , Phosphatidylcholines/pharmacology , Phosphatidylcholines/therapeutic use , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/immunology , Severity of Illness Index , Treatment Outcome , Wound Healing/drug effects
10.
Front Immunol ; 9: 2980, 2018.
Article in English | MEDLINE | ID: mdl-30619336

ABSTRACT

Background: The pathophysiology of non-alcoholic steatohepatitis involves hepatocyte lipotoxicity due to excess saturated free fatty acids and concomitant proinflammatory macrophage effector responses. These include the infiltration of macrophages into hepatic cords in response to incompletely understood stimuli. Stressed hepatocytes release an increased number of extracellular vesicles (EVs), which are known to participate in intercellular signaling and coordination of the behavior of immune cell populations via their cargo. We hypothesized that hepatocyte-derived lipotoxic EVs that are enriched in sphingosine 1-phosphate (S1P) are effectors of macrophage infiltration in the hepatic microenvironment. Methods: Lipotoxic EVs were isolated from palmitate treated immortalized mouse hepatocytes and characterized by nanoparticle tracking analysis. Lipotoxic EV sphingolipids were quantified using tandem mass spectrometry. Wildtype and S1P1 receptor knockout bone marrow-derived macrophages were exposed to lipotoxic EV gradients in a microfluidic gradient generator. Macrophage migration toward EV gradients was captured by time-lapse microscopy and analyzed to determine directional migration. Fluorescence-activated cell sorting along with quantitative PCR and immunohistochemistry were utilized to characterize the cell surface expression of S1P1 receptor on intrahepatic leukocytes and hepatic expression of S1P1 receptor, respectively. Results: Palmitate treatment induced the release of EVs. These EVs were enriched in S1P. Palmitate-induced S1P enriched EVs were chemoattractive to macrophages. EV S1P enrichment depended on the activity of sphingosine kinases 1 and 2, such that, pharmacological inhibition of sphingosine kinases 1 and 2 resulted in a significant reduction in EV S1P cargo without affecting the number of EVs released. When exposed to EVs derived from cells treated with palmitate in the presence of a pharmacologic inhibitor of sphingosine kinases 1 and 2, macrophages displayed diminished chemotactic behavior. To determine receptor-ligand specificity, we tested the migration responses of macrophages genetically deleted in the S1P1 receptor toward lipotoxic EVs. S1P1 receptor knockout macrophages displayed a marked reduction in their chemotactic responses toward lipotoxic palmitate-induced EVs. Conclusions:Palmitate-induced lipotoxic EVs are enriched in S1P through sphingosine kinases 1 and 2. S1P-enriched EVs activate persistent and directional macrophage chemotaxis mediated by the S1P1 receptor, a potential signaling axis for macrophage infiltration during hepatic lipotoxicity, and a potential therapeutic target for non-alcoholic steatohepatitis.


Subject(s)
Extracellular Vesicles/immunology , Hepatocytes/immunology , Lysophospholipids/immunology , Macrophages/immunology , Non-alcoholic Fatty Liver Disease/immunology , Sphingosine/analogs & derivatives , Animals , Cell Line , Chemotaxis/immunology , Diet, Atherogenic/adverse effects , Diet, Carbohydrate Loading/adverse effects , Diet, High-Fat/adverse effects , Disease Models, Animal , Extracellular Vesicles/metabolism , Gene Knockout Techniques , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/cytology , Liver/immunology , Liver/pathology , Lysophospholipids/metabolism , Male , Mice , Mice, Inbred C57BL , Non-alcoholic Fatty Liver Disease/etiology , Non-alcoholic Fatty Liver Disease/pathology , Palmitic Acid/pharmacology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/immunology , Receptors, Lysosphingolipid/metabolism , Sphingosine/immunology , Sphingosine/metabolism , Sphingosine-1-Phosphate Receptors
12.
Eur J Immunol ; 48(3): 498-508, 2018 03.
Article in English | MEDLINE | ID: mdl-29205338

ABSTRACT

Myasthenia gravis (MG) is an autoimmune disease characterized by muscle weakness and fatigue in the presence of circulating antibodies against components of the neuromuscular junction. Most patients have a good prognosis, but some are refractory to standard-of-care immunosuppressive treatment and suffer from recurrent myasthenic crises. Functional sphingosine-1-phosphate (S1P) antagonists like fingolimod and siponimod (BAF312) are successfully used for the treatment of multiple sclerosis, and fingolimod was shown to prevent the development of myasthenic symptoms in experimental autoimmune myasthenia gravis (EAMG), the standard model of MG. Here, we investigated whether fingolimod or siponimod improves outcome in EAMG mice when administered after disease onset, modeling the clinical setting in human MG. Both S1P antagonists inhibited lymphocyte egress, resulting in peripheral lymphopenia. After stimulation, there were differences in T-cell responses, but no change in either antibody titers or total or antigen-specific plasma cell populations after treatment. Most importantly, disease incidence and severity were not influenced by fingolimod or siponimod therapy. Although fingolimod and siponimod did lead to subtle changes in T-cell responses, they had no significant effect on antibody titers and disease severity. In conclusion, our data show no evidence of a therapeutic potential for S1P receptor antagonists in MG treatment.


Subject(s)
Azetidines/pharmacology , Benzyl Compounds/pharmacology , Fingolimod Hydrochloride/pharmacology , Myasthenia Gravis, Autoimmune, Experimental/drug therapy , Receptors, Lysosphingolipid/antagonists & inhibitors , Animals , Antibody Formation/drug effects , Cytokines/biosynthesis , Female , Humans , Immunosuppressive Agents/pharmacology , Lymphopenia/chemically induced , Male , Mice , Mice, Inbred C57BL , Myasthenia Gravis, Autoimmune, Experimental/immunology , Myasthenia Gravis, Autoimmune, Experimental/metabolism , Plasma Cells/drug effects , Receptors, Lysosphingolipid/immunology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology
13.
Am J Respir Crit Care Med ; 196(12): 1559-1570, 2017 12 15.
Article in English | MEDLINE | ID: mdl-28850247

ABSTRACT

RATIONALE: Efficient elimination of pathogenic bacteria is a critical determinant in the outcome of sepsis. Sphingosine-1-phosphate receptor 3 (S1PR3) mediates multiple aspects of the inflammatory response during sepsis, but whether S1PR3 signaling is necessary for eliminating the invading pathogens remains unknown. OBJECTIVES: To investigate the role of S1PR3 in antibacterial immunity during sepsis. METHODS: Loss- and gain-of-function experiments were performed using cell and murine models. S1PR3 levels were determined in patients with sepsis and healthy volunteers. MEASUREMENTS AND MAIN RESULTS: S1PR3 protein levels were up-regulated in macrophages upon bacterial stimulation. S1pr3-/- mice showed increased mortality and increased bacterial burden in multiple models of sepsis. The transfer of wild-type bone marrow-derived macrophages rescued S1pr3-/- mice from lethal sepsis. S1PR3-overexpressing macrophages further ameliorated the mortality rate of sepsis. Loss of S1PR3 led to markedly decreased bacterial killing in macrophages. Enhancing endogenous S1PR3 activity using a peptide agonist potentiated the macrophage bactericidal function and improved survival rates in multiple models of sepsis. Mechanically, the reactive oxygen species levels were decreased and phagosome maturation was delayed in S1pr3-/- macrophages due to impaired recruitment of vacuolar protein-sorting 34 to the phagosomes. In addition, S1RP3 expression levels were elevated in monocytes from patients with sepsis. Higher levels of monocytic S1PR3 were associated with efficient intracellular bactericidal activity, better immune status, and preferable outcomes. CONCLUSIONS: S1PR3 signaling drives bacterial killing and is essential for survival in bacterial sepsis. Interventions targeting S1PR3 signaling could have translational implications for manipulating the innate immune response to combat pathogens.


Subject(s)
Cell Death/immunology , Receptors, Lysosphingolipid/genetics , Receptors, Lysosphingolipid/immunology , Sepsis/immunology , Signal Transduction/immunology , Animals , Cell Death/genetics , Disease Models, Animal , Disease-Free Survival , Humans , Mice , Signal Transduction/genetics , Sphingosine-1-Phosphate Receptors , Up-Regulation/genetics , Up-Regulation/immunology
14.
Gastroenterol Clin North Am ; 46(3): 627-644, 2017 09.
Article in English | MEDLINE | ID: mdl-28838419

ABSTRACT

There is an ongoing, unmet need for effective therapies for Crohn's disease. Treatments for Crohn's disease continue to evolve from the traditional biologics to novel small molecules, with targeted mechanisms directed toward pathways that are dysregulated in Crohn's disease. There are multiple emerging mechanisms of action, including Janus kinase inhibition, Smad7 inhibition, and sphingosine-1-phosphate receptor modulators, that are administered as oral medications, and small molecules represent the next generation of therapies for Crohn's disease.


Subject(s)
Crohn Disease/drug therapy , Indans/therapeutic use , Janus Kinase Inhibitors/therapeutic use , Oligonucleotides/therapeutic use , Oxadiazoles/therapeutic use , Piperidines/therapeutic use , Pyridines/therapeutic use , Pyrimidines/therapeutic use , Pyrroles/therapeutic use , Triazoles/therapeutic use , B-Lymphocytes/immunology , Colitis, Ulcerative/drug therapy , Crohn Disease/immunology , Humans , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 1/immunology , Receptors, Lysosphingolipid/immunology , Smad7 Protein/antagonists & inhibitors , T-Lymphocytes/immunology
15.
Biochem Biophys Res Commun ; 490(4): 1274-1281, 2017 09 02.
Article in English | MEDLINE | ID: mdl-28687489

ABSTRACT

Enhanced turnover of subchondral trabecular bone is a hallmark of rheumatoid arthritis (RA) and it results from an imbalance between bone resorption and bone formation activities. To investigate the formation and activation of osteoclasts which mediate bone resorption, a Fas-deficient MRL/lpr mouse model which spontaneously develops autoimmune arthritis and exhibits decreased bone mass was studied. Various assays were performed on subchondral trabecular bone of the temporomandibular joint (TMJ) from MRL/lpr mice and MRL+/+ mice. Initially, greater osteoclast production was observed in vitro from bone marrow macrophages obtained from MRL/lpr mice due to enhanced phosphorylation of NF-κB, as well as Akt and MAPK, to receptor activator of nuclear factor-κB ligand (RANKL). Expression of sphingosine 1-phosphate receptor 1 (S1P1) was also significantly upregulated in the condylar cartilage. S1P1 was found to be required for S1P-induced migration of osteoclast precursor cells and downstream signaling via Rac1. When SN50, a synthetic NF-κB-inhibitory peptide, was applied to the MRL/lpr mice, subchondral trabecular bone loss was reduced and both production of osteoclastogenesis markers and sphingosine kinase (Sphk) 1/S1P1 signaling were reduced. Thus, the present results suggest that Fas/S1P1 signaling via activation of NF-κB in osteoclast precursor cells is a key factor in the pathogenesis of RA in the TMJ.


Subject(s)
Arthritis, Rheumatoid/immunology , Bone Resorption/immunology , NF-kappa B/immunology , Osteoclasts/drug effects , Receptors, Lysosphingolipid/immunology , Temporomandibular Joint/immunology , fas Receptor/immunology , Animals , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Autoimmunity , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Bone Marrow Cells/pathology , Bone Resorption/genetics , Bone Resorption/pathology , Bone Resorption/prevention & control , Cell Differentiation , Disease Models, Animal , Female , Gene Expression Regulation , Lysophospholipids/immunology , Macrophages/drug effects , Macrophages/immunology , Macrophages/pathology , Mice , Mice, Inbred MRL lpr , Mitogen-Activated Protein Kinases/genetics , Mitogen-Activated Protein Kinases/immunology , NF-kappa B/antagonists & inhibitors , NF-kappa B/genetics , Neuropeptides/genetics , Neuropeptides/immunology , Osteoclasts/immunology , Osteoclasts/pathology , Osteogenesis/drug effects , Osteogenesis/immunology , Peptides/pharmacology , Phosphotransferases (Alcohol Group Acceptor)/genetics , Phosphotransferases (Alcohol Group Acceptor)/immunology , Primary Cell Culture , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/immunology , RANK Ligand/genetics , RANK Ligand/immunology , Receptors, Lysosphingolipid/genetics , Signal Transduction , Sphingosine/analogs & derivatives , Sphingosine/immunology , Temporomandibular Joint/drug effects , Temporomandibular Joint/pathology , fas Receptor/genetics , rac1 GTP-Binding Protein/genetics , rac1 GTP-Binding Protein/immunology
16.
Nat Immunol ; 18(7): 771-779, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28530714

ABSTRACT

TCRαß+CD4-CD8α+CD8ß- intestinal intraepithelial lymphocytes (CD8αα IELs) are an abundant population of thymus-derived T cells that protect the gut barrier surface. We sought to better define the thymic IEL precursor (IELp) through analysis of its maturation, localization and emigration. We defined two precursor populations among TCRß+CD4-CD8- thymocytes by dependence on the kinase TAK1 and rigorous lineage-exclusion criteria. Those IELp populations included a nascent PD-1+ population and a T-bet+ population that accumulated with age. Both gave rise to intestinal CD8αα IELs after adoptive transfer. The PD-1+ IELp population included more strongly self-reactive clones and was largely restricted by classical major histocompatibility complex (MHC) molecules. Those cells localized to the cortex and efficiently emigrated in a manner dependent on the receptor S1PR1. The T-bet+ IELp population localized to the medulla, included cells restricted by non-classical MHC molecules and expressed the receptor NK1.1, the integrin CD103 and the chemokine receptor CXCR3. The two IELp populations further differed in their use of the T cell antigen receptor (TCR) α-chain variable region (Vα) and ß-chain variable region (Vß). These data provide a foundation for understanding the biology of CD8αα IELs.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Intestinal Mucosa/immunology , Precursor Cells, T-Lymphoid/immunology , Thymocytes/immunology , Adaptive Immunity/immunology , Adoptive Transfer , Animals , Antigens, CD , Antigens, Ly/immunology , CD8 Antigens/immunology , Cell Lineage , Cell Movement/immunology , Flow Cytometry , Fluorescent Antibody Technique , Histocompatibility Antigens/immunology , Immunity, Mucosal/immunology , Integrin alpha Chains , Intestinal Mucosa/cytology , Lymphocytes , Mice , NK Cell Lectin-Like Receptor Subfamily B/immunology , Phenotype , Programmed Cell Death 1 Receptor/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , Receptors, CXCR3 , Receptors, Lysosphingolipid/immunology , Sphingosine-1-Phosphate Receptors , T-Box Domain Proteins/immunology , Thymocytes/cytology , Thymus Gland/cytology
17.
JCI Insight ; 2(7): e88533, 2017 04 06.
Article in English | MEDLINE | ID: mdl-28405607

ABSTRACT

HIV vaginal transmission accounts for the majority of newly acquired heterosexual infections. However, the mechanism by which HIV spreads from the initial site of viral entry at the mucosal surface of the female genital tract to establish a systemic infection of lymphoid and peripheral tissues is not known. Once the virus exits the mucosa it rapidly spreads to all tissues, leading to CD4+ T cell depletion and the establishment of a viral reservoir that cannot be eliminated with current treatments. Understanding the molecular and cellular requirements for viral dissemination from the genital tract is therefore of great importance, as it could reveal new strategies to lengthen the window of opportunity to target the virus at its entry site in the mucosa where it is the most vulnerable and thus prevent systemic infection. Using HIV vaginal infection of humanized mice as a model of heterosexual transmission, we demonstrate that blocking the ability of leukocytes to respond to chemoattractants prevented HIV from leaving the female genital tract. Furthermore, blocking lymphocyte egress from lymph nodes prevented viremia and infection of the gut. Leukocyte trafficking therefore plays a major role in viral dissemination, and targeting the chemoattractant molecules involved can prevent the establishment of a systemic infection.


Subject(s)
CD4-Positive T-Lymphocytes/virology , HIV Infections/transmission , HIV-1/pathogenicity , Receptors, CCR7/immunology , Receptors, Lysosphingolipid/immunology , Vagina/virology , Animals , CD4-Positive T-Lymphocytes/immunology , Cell Movement , Chemokine CCL19/immunology , Chemokine CCL21/immunology , Disease Models, Animal , Female , Humans , Leukocytes/immunology , Mice , Mice, Inbred NOD , Mice, SCID , Sphingosine-1-Phosphate Receptors , Vagina/immunology
18.
Cancer Immunol Immunother ; 66(5): 593-603, 2017 May.
Article in English | MEDLINE | ID: mdl-28224210

ABSTRACT

Regulatory T cells (Treg) hamper anti-tumor T-cell responses resulting in reduced survival and failure of cancer immunotherapy. Among lymphoid organs, the bone marrow (BM) is a major site of Treg residence and recirculation. However, the process governing the emigration of Treg from BM into the circulation remains elusive. We here show that breast cancer patients harbour reduced Treg frequencies in the BM as compared to healthy individuals or the blood. This was particularly the case for tumor antigen-specific Treg which were quantified by MHCII tumor peptide loaded tetramers. We further demonstrate that decreased Treg distribution in the BM correlated with increased Treg redistribution to tumor tissue, suggesting that TCR triggering induces a translocation of Treg from the BM into tumor tissue. Sphingosine-1-phosphate receptor 1 (S1P1)-which is known to mediate exit of immune cells from lymphoid organs was selectively expressed by tumor antigen-specific BM Treg. S1P1 expression could be induced in Treg by BM-resident antigen-presenting cells (BMAPCs) in conjunction with TCR stimulation, but not by TCR stimulation or BMAPCs alone and triggered the migration of Treg but not conventional T cells (Tcon) to its ligand Sphingosine-1-phosphate (S1P). Interestingly, we detected marked S1P gradients between PB and BM in breast cancer patients but not in healthy individuals. Taken together, our data suggest a role for S1P1 in mediating the selective mobilization of tumor specific Treg from the BM of breast cancer patients and their translocation into tumor tissue.


Subject(s)
Bone Marrow Cells/immunology , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Receptors, Lysosphingolipid/immunology , T-Lymphocytes, Regulatory/immunology , Adult , Aged , Female , Humans , Middle Aged , Up-Regulation
19.
Immunity ; 45(5): 1078-1092, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27851911

ABSTRACT

Th17 cells are most abundant in the gut, where their presence depends on the intestinal microbiota. Here, we examined whether intestinal Th17 cells contribute to extra-intestinal Th17 responses in autoimmune kidney disease. We found high frequencies of Th17 cells in the kidneys of patients with antineutrophil cytoplasmatic antibody (ANCA)-associated glomerulonephritis. We utilized photoconversion of intestinal cells in Kaede mice to track intestinal T cell mobilization upon glomerulonephritis induction, and we found that Th17 cells egress from the gut in a S1P-receptor-1-dependent fashion and subsequently migrate to the kidney via the CCL20/CCR6 axis. Depletion of intestinal Th17 cells in germ-free and antibiotic-treated mice ameliorated renal disease, whereas expansion of these cells upon Citrobacter rodentium infection exacerbated pathology. Thus, in some autoimmune settings, intestinal Th17 cells migrate into target organs, where they contribute to pathology. Targeting the intestinal Th17 cell "reservoir" may present a therapeutic strategy for these autoimmune disorders.


Subject(s)
Autoimmune Diseases/immunology , Chemotaxis, Leukocyte/immunology , Glomerulonephritis/immunology , Receptors, Lysosphingolipid/immunology , Th17 Cells/immunology , Animals , Citrobacter rodentium , Disease Models, Animal , Enterobacteriaceae Infections/immunology , Flow Cytometry , Humans , Intestines/immunology , Kidney/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Real-Time Polymerase Chain Reaction , Sphingosine-1-Phosphate Receptors
SELECTION OF CITATIONS
SEARCH DETAIL
...