Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
3.
J Immunol ; 200(1): 110-118, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29187589

ABSTRACT

Understanding the control of Ag restimulation-induced T cell death (RICD), especially in cancer immunotherapy, where highly proliferating T cells will encounter potentially large amounts of tumor Ags, is important now more than ever. It has been known that growth cytokines make T cells susceptible to RICD, but the precise molecular mediators that govern this in T cell subsets is unknown until now. STAT proteins are a family of transcription factors that regulate gene expression programs underlying key immunological processes. In particular, STAT5 is known to favor the generation and survival of memory T cells. In this study, we report an unexpected role for STAT5 signaling in the death of effector memory T (TEM) cells in mice and humans. TEM cell death was prevented with neutralizing anti-IL-2 Ab or STAT5/JAK3 inhibitors, indicating that STAT5 signaling drives RICD in TEM cells. Moreover, we identified a unique patient with a heterozygous missense mutation in the coiled-coil domain of STAT5B that presented with autoimmune lymphoproliferative syndrome-like features. Similar to Stat5b-/- mice, this patient exhibited increased CD4+ TEM cells in the peripheral blood. The mutant STAT5B protein dominantly interfered with STAT5-driven transcriptional activity, leading to global downregulation of STAT5-regulated genes in patient T cells upon IL-2 stimulation. Notably, CD4+ TEM cells from the patient were strikingly resistant to cell death by in vitro TCR restimulation, a finding that was recapitulated in Stat5b-/- mice. Hence, STAT5B is a crucial regulator of RICD in memory T cells in mice and humans.


Subject(s)
Apoptosis , Autoimmune Lymphoproliferative Syndrome/immunology , CD4-Positive T-Lymphocytes/immunology , Cell Survival , STAT5 Transcription Factor/metabolism , Animals , Antibodies, Neutralizing/metabolism , Autoimmune Lymphoproliferative Syndrome/genetics , Cells, Cultured , Female , Humans , Immunologic Memory , Interleukin-2/immunology , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation, Missense/genetics , STAT5 Transcription Factor/genetics , Signal Transduction , Transcription, Genetic
4.
Trends Immunol ; 35(7): 332-44, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24932518

ABSTRACT

Divalent cations of two alkaline earth metals Ca(2+) and Mg(2+) and the transition metal Zn(2+) play vital roles in the immune system, and several immune disorders are associated with disturbances of their function. Until recently only Ca(2+) was considered to serve as a second messenger. However, signaling roles for Mg(2+) and Zn(2+) have been recently described, leading to a reevaluation of their role as potential second messengers. We review here the roles of these cations as second messengers in light of recent advances in Ca(2+), Mg(2+), and Zn(2+) signaling in the immune system. Developing a better understanding of these signaling cations may lead to new therapeutic strategies for immune disorders.


Subject(s)
Cations, Divalent/metabolism , Immune System Diseases/metabolism , Immune System , Animals , Humans , Immune System Diseases/therapy , Molecular Targeted Therapy , Second Messenger Systems/immunology , Signal Transduction
5.
Nature ; 475(7357): 471-6, 2011 Jul 27.
Article in English | MEDLINE | ID: mdl-21796205

ABSTRACT

The magnesium ion, Mg(2+), is essential for all life as a cofactor for ATP, polyphosphates such as DNA and RNA, and metabolic enzymes, but whether it plays a part in intracellular signalling (as Ca(2+) does) is unknown. Here we identify mutations in the magnesium transporter gene, MAGT1, in a novel X-linked human immunodeficiency characterized by CD4 lymphopenia, severe chronic viral infections, and defective T-lymphocyte activation. We demonstrate that a rapid transient Mg(2+) influx is induced by antigen receptor stimulation in normal T cells and by growth factor stimulation in non-lymphoid cells. MAGT1 deficiency abrogates the Mg(2+) influx, leading to impaired responses to antigen receptor engagement, including defective activation of phospholipase Cγ1 and a markedly impaired Ca(2+) influx in T cells but not B cells. These observations reveal a role for Mg(2+) as an intracellular second messenger coupling cell-surface receptor activation to intracellular effectors and identify MAGT1 as a possible target for novel therapeutics.


Subject(s)
Magnesium/immunology , Second Messenger Systems/immunology , T-Lymphocytes/immunology , T-Lymphocytopenia, Idiopathic CD4-Positive/immunology , Calcium/immunology , Cation Transport Proteins/genetics , Female , Gene Knockdown Techniques , HEK293 Cells , Humans , Male , Phospholipase C gamma/genetics , Phospholipase C gamma/metabolism , T-Lymphocytopenia, Idiopathic CD4-Positive/genetics
6.
Blood ; 123(14): 2148-52, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24550228

ABSTRACT

Epstein-Barr virus (EBV) is an oncogenic gammaherpesvirus that infects and persists in 95% of adults worldwide and has the potential to cause fatal disease, especially lymphoma, in immunocompromised hosts. Primary immunodeficiencies (PIDs) that predispose to EBV-associated malignancies have provided novel insights into the molecular mechanisms of immune defense against EBV. We have recently characterized a novel PID now named "X-linked immunodeficiency with magnesium defect, EBV infection, and neoplasia" (XMEN) disease characterized by loss-of-function mutations in the gene encoding magnesium transporter 1 (MAGT1), chronic high-level EBV with increased EBV-infected B cells, and heightened susceptibility to EBV-associated lymphomas. The genetic etiology of XMEN disease has revealed an unexpected quantitative role for intracellular free magnesium in immune functions and has led to novel diagnostic and therapeutic strategies. Here, we review the clinical presentation, genetic mutation spectrum, molecular mechanisms of pathogenesis, and diagnostic and therapeutic considerations for this previously unrecognized disease.


Subject(s)
Epstein-Barr Virus Infections/complications , Herpesvirus 4, Human/immunology , Immunity, Innate/drug effects , Magnesium Deficiency/complications , Magnesium/pharmacology , Neoplasms/complications , X-Linked Combined Immunodeficiency Diseases/complications , Adult , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/genetics , Humans , Magnesium Deficiency/diagnosis , Magnesium Deficiency/genetics , Neoplasms/diagnosis , Neoplasms/genetics , Syndrome , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/genetics
7.
J Clin Immunol ; 35(2): 112-8, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25504528

ABSTRACT

XMEN disease (X-linked immunodeficiency with Magnesium defect, Epstein-Barr virus infection and Neoplasia) is a novel primary immune deficiency caused by mutations in MAGT1 and characterised by chronic infection with Epstein-Barr virus (EBV), EBV-driven lymphoma, CD4 T-cell lymphopenia, and dysgammaglobulinemia [1]. Functional studies have demonstrated roles for magnesium as a second messenger in T-cell receptor signalling [1], and for NKG2D expression and consequently NK- and CD8 T-cell cytotoxicity [2]. 7 patients have been described in the literature; the oldest died at 45 years and was diagnosed posthumously [1-3]. We present the case of a 58-year-old Caucasian gentleman with a novel mutation in MAGT1 with the aim of adding to the phenotype of this newly described disease by detailing his clinical course over more than 20 years.


Subject(s)
Cation Transport Proteins/genetics , Leukoencephalopathy, Progressive Multifocal/diagnosis , Leukoencephalopathy, Progressive Multifocal/etiology , Mutation , X-Linked Combined Immunodeficiency Diseases/complications , X-Linked Combined Immunodeficiency Diseases/genetics , Brain/pathology , DNA Mutational Analysis , Fluorodeoxyglucose F18 , Humans , Immunophenotyping , Lymph Nodes/pathology , Magnetic Resonance Imaging , Male , Middle Aged , Phenotype , Positron-Emission Tomography , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Tomography, X-Ray Computed , X-Linked Combined Immunodeficiency Diseases/diagnosis
8.
PLoS Biol ; 9(6): e1001090, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21713032

ABSTRACT

Patients affected by chronic inflammatory disorders display high amounts of soluble CD95L. This homotrimeric ligand arises from the cleavage by metalloproteases of its membrane-bound counterpart, a strong apoptotic inducer. In contrast, the naturally processed CD95L is viewed as an apoptotic antagonist competing with its membrane counterpart for binding to CD95. Recent reports pinpointed that activation of CD95 may attract myeloid and tumoral cells, which display resistance to the CD95-mediated apoptotic signal. However, all these studies were performed using chimeric CD95Ls (oligomerized forms), which behave as the membrane-bound ligand and not as the naturally processed CD95L. Herein, we examine the biological effects of the metalloprotease-cleaved CD95L on CD95-sensitive activated T-lymphocytes. We demonstrate that cleaved CD95L (cl-CD95L), found increased in sera of systemic lupus erythematosus (SLE) patients as compared to that of healthy individuals, promotes the formation of migrating pseudopods at the leading edge of which the death receptor CD95 is capped (confocal microscopy). Using different migration assays (wound healing/Boyden Chamber/endothelial transmigration), we uncover that cl-CD95L promotes cell migration through a c-yes/Ca²âº/PI3K-driven signaling pathway, which relies on the formation of a CD95-containing complex designated the MISC for Motility-Inducing Signaling Complex. These findings revisit the role of the metalloprotease-cleaved CD95L and emphasize that the increase in cl-CD95L observed in patients affected by chronic inflammatory disorders may fuel the local or systemic tissue damage by promoting tissue-filtration of immune cells.


Subject(s)
Cell Movement/immunology , Fas Ligand Protein/physiology , Phosphatidylinositol 3-Kinases/physiology , Apoptosis/drug effects , Cell Movement/drug effects , HEK293 Cells , Humans , Lupus Erythematosus, Systemic/blood , Pseudopodia/physiology , Signal Transduction , Transendothelial and Transepithelial Migration/physiology , fas Receptor/immunology , fas Receptor/metabolism , src-Family Kinases/physiology
9.
Curr Opin Pediatr ; 26(6): 713-9, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25313976

ABSTRACT

PURPOSE OF REVIEW: To describe the role of the magnesium transporter 1 (MAGT1) in the pathogenesis of 'X-linked immunodeficiency with magnesium defect, Epstein-Barr virus (EBV) infection, and neoplasia' (XMEN) disease and its clinical implications. RECENT FINDINGS: The magnesium transporter protein MAGT1 participates in the intracellular magnesium ion (Mg) homeostasis and facilitates a transient Mg influx induced by the activation of the T-cell receptor. Loss-of-function mutations in MAGT1 cause an immunodeficiency named 'XMEN syndrome', characterized by CD4 lymphopenia, chronic EBV infection, and EBV-related lymphoproliferative disorders. Patients with XMEN disease have impaired T-cell activation and decreased cytolytic function of natural killer (NK) and CD8 T cells because of decreased expression of the NK stimulatory receptor 'natural-killer group 2, member D' (NKG2D). Patients may have defective specific antibody responses secondary to T cell dysfunction, but B cells have not been shown to be directly affected by mutations in MAGT1. SUMMARY: XMEN disease has revealed a novel role for free intracellular magnesium in the immune system. Further understanding of the MAGT1 signaling pathway may lead to new diagnostic and therapeutic approaches.


Subject(s)
Epstein-Barr Virus Infections/complications , Magnesium Deficiency/complications , Neoplasms/complications , X-Linked Combined Immunodeficiency Diseases/complications , Cation Transport Proteins , Child , Epstein-Barr Virus Infections/diagnosis , Epstein-Barr Virus Infections/therapy , Humans , Magnesium Deficiency/diagnosis , Magnesium Deficiency/therapy , Male , Neoplasms/diagnosis , Neoplasms/therapy , Syndrome , X-Linked Combined Immunodeficiency Diseases/diagnosis , X-Linked Combined Immunodeficiency Diseases/therapy
10.
Proc Natl Acad Sci U S A ; 108(47): 19072-7, 2011 11 22.
Article in English | MEDLINE | ID: mdl-22065776

ABSTRACT

The death receptor CD95 plays a pivotal role in immune surveillance and immune tolerance. Binding of CD95L to CD95 leads to recruitment of the adaptor protein Fas-associated death domain protein (FADD), which in turn aggregates caspase-8 and caspase-10. Efficient formation of the CD95/FADD/caspase complex, known as the death-inducing signaling complex (DISC), culminates in the induction of apoptosis. We show that cells exposed to CD95L undergo a reorganization of the plasma membrane in which the Ca(2+) release-activated Ca(2+) channel Orai1 and the endoplasmic reticulum-resident activator stromal interaction molecule 1 colocalize with CD95 into a micrometer-sized cluster in which the channel elicits a polarized entry of calcium. Orai1 knockdown and expression of a dominant negative construct (Orai1E106A) reveal that on CD95 engagement, the Orai1-driven localized Ca(2+) influx is fundamental to recruiting the Ca(2+)-dependent protein kinase C (PKC) ß2 to the DISC. PKCß2 in turn transiently holds the complex in an inactive status, preventing caspase activation and transmission of the apoptotic signal. This study identifies a biological role of Ca(2+) and the Orai1 channel that drives a transient negative feedback loop, introducing a lag phase in the early steps of the CD95 signal. We suggest that these localized events provide a time of decision to prevent accidental cell death.


Subject(s)
Apoptosis/physiology , Calcium Channels/metabolism , Calcium/metabolism , Multiprotein Complexes/metabolism , Protein Kinase C/metabolism , fas Receptor/metabolism , Blotting, Western , Caspase 10/metabolism , Caspase 8/metabolism , Cell Line , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/metabolism , Flow Cytometry , Humans , Immunoprecipitation , Microscopy, Confocal , ORAI1 Protein , Protein Kinase C beta , Statistics, Nonparametric
11.
Eur J Immunol ; 41(8): 2368-78, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21557211

ABSTRACT

The immune system eliminates infected or transformed cells through the activation of the death receptor CD95. CD95 engagement drives the recruitment of the adaptor protein Fas-associated death domain protein (FADD), which in turn aggregates and activates initiator caspases-8 and -10. The CD95-mediated apoptotic signal relies on the capacity to form the CD95/FADD/caspases complex termed the death-inducing signalling complex (DISC). Cells are classified according to the magnitude of DISC formation as either type I (efficient DISC formation) or type II (inefficient). CD95 localised to lipid rafts in type I cells, whereas the death receptor was excluded from these domains in type II cells. Here, we show that inhibition of both PI3K class IA and serine-threonine kinase Akt in type II cells promoted the redistribution of CD95 into lipid rafts, DISC formation and the initiation of the apoptotic signal. Strikingly, these molecular events took place independently of CD95L and the actin cytoskeleton. Overall, these findings highlight that the oncogenic PI3K/Akt signalling pathway participates in maintaining cells in a type II phenotype by excluding CD95 from lipid rafts.


Subject(s)
Actins/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , fas Receptor/metabolism , Androstadienes/pharmacology , Apoptosis/drug effects , Blotting, Western , Caspases/metabolism , Cell Line , Cell Line, Tumor , Cell Membrane/metabolism , Chromones/pharmacology , Fas Ligand Protein/metabolism , Fas-Associated Death Domain Protein/metabolism , Flow Cytometry , Humans , Jurkat Cells , Membrane Microdomains/metabolism , Morpholines/pharmacology , Multiprotein Complexes/metabolism , Mutation , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/genetics , Wortmannin
12.
J Cell Sci ; 122(Pt 23): 4311-8, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19887587

ABSTRACT

Podosomes are specialized plasma-membrane actin-based microdomains that combine adhesive and proteolytic activities to spatially restrict sites of matrix degradation in in vitro assays, but the physiological relevance of these observations remain unknown. Inducible rings of podosomes (podosome rosettes) form in cultured aortic cells exposed to the inflammatory cytokine TGFbeta. In an attempt to prove the existence of podosomes in living tissues, we developed an ex vivo endothelium observation model. This system enabled us to visualize podosome rosettes in the endothelium of native arterial vessel exposed to biologically active TGFbeta. Podosomes induced in the vessel appear similar to those formed in cultured cells in terms of molecular composition, but in contrast to the latter, arrange in a protruding structure that is similar to invadopodia. Local degradation of the basement membrane scaffold protein collagen-IV, is observed underneath the structures. Our results reveal for the first time the presence of podosome rosettes in the native endothelium and provide evidence for their capacity to degrade the basement membrane, opening up new avenues to study their role in vascular pathophysiology. We propose that podosome rosettes are involved in arterial vessel remodeling.


Subject(s)
Basement Membrane/metabolism , Collagen/metabolism , Coronary Vessels/drug effects , Coronary Vessels/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Transforming Growth Factor beta/pharmacology , Animals , Basement Membrane/drug effects , Cells, Cultured , Endothelium/drug effects , Endothelium/metabolism , In Vitro Techniques , Mice , Mice, Inbred C57BL , Microscopy, Fluorescence
13.
Virol J ; 8: 219, 2011 May 11.
Article in English | MEDLINE | ID: mdl-21569376

ABSTRACT

The two major cytopathic factors in human immunodeficiency virus type 1 (HIV-1), the accessory proteins viral infectivity factor (Vif) and viral protein R (Vpr), inhibit cell-cycle progression at the G2 phase of the cell cycle. Although Vpr-induced blockade and the associated T-cell death have been well studied, the molecular mechanism of G2 arrest by Vif remains undefined. To elucidate how Vif induces arrest, we infected synchronized Jurkat T-cells and examined the effect of Vif on the activation of Cdk1 and CyclinB1, the chief cell-cycle factors for the G2 to M phase transition. We found that the characteristic dephosphorylation of an inhibitory phosphate on Cdk1 did not occur in infected cells expressing Vif. In addition, the nuclear translocation of Cdk1 and CyclinB1 was disregulated. Finally, Vif-induced cell cycle arrest was correlated with proviral expression of Vif. Taken together, our results suggest that Vif impairs mitotic entry by interfering with Cdk1-CyclinB1 activation.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Cycle , Cyclin B1/metabolism , Virulence Factors/metabolism , vif Gene Products, Human Immunodeficiency Virus/metabolism , CDC2 Protein Kinase/antagonists & inhibitors , Cyclin B1/antagonists & inhibitors , HIV-1 , Humans , Jurkat Cells , T-Lymphocytes/physiology , T-Lymphocytes/virology
14.
J Immunol ; 181(11): 7630-8, 2008 Dec 01.
Article in English | MEDLINE | ID: mdl-19017951

ABSTRACT

Mycophenolate mofetil (MMF) is an immunosuppressive agent used in transplantation. Over the last decade, MMF has also emerged as an alternative therapeutic regimen for autoimmune diseases, mainly for patients refractory to other therapies. The active compound of MMF, mycophenolic acid (MPA), depletes the intracellular pool of guanosine tri-phosphate through inosine monophosphate dehydrogenase blockade. The molecular mechanism involved in the elimination of T and B lymphocytes upon inhibition of inosine monophosphate dehydrogenase remains elusive. In this study, we showed that in contrast to the immunosuppressors azathioprine, cyclosporin A, and tacrolimus, MPA killed lymphocytes through the activation of a caspase-independent necrotic signal. Furthermore, the MPA-mediated necrotic signal relied on the transmission of a novel intracellular signal involving Rho-GTPase Cdc42 activity and actin polymerization. In addition to its medical interest, this study sheds light on a novel and atypical molecular mechanism leading to necrotic cell death.


Subject(s)
Actins/immunology , B-Lymphocytes/immunology , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/drug effects , Mycophenolic Acid/pharmacology , Signal Transduction/drug effects , T-Lymphocytes/immunology , Actins/metabolism , Adenosine Triphosphatases/immunology , Adenosine Triphosphatases/metabolism , B-Lymphocytes/enzymology , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Guanosine Triphosphate/immunology , Guanosine Triphosphate/metabolism , Humans , IMP Dehydrogenase/immunology , IMP Dehydrogenase/metabolism , Immunosuppressive Agents/therapeutic use , Jurkat Cells , Lymphocyte Activation/immunology , Mycophenolic Acid/therapeutic use , Necrosis/chemically induced , Necrosis/immunology , Organ Transplantation , Signal Transduction/immunology , T-Lymphocytes/enzymology , cdc42 GTP-Binding Protein/immunology , cdc42 GTP-Binding Protein/metabolism
15.
Mol Cancer Res ; 6(4): 604-13, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18403639

ABSTRACT

Activation of the phosphatidylinositol 3-kinase (PI3K) signaling pathway is known to protect tumor cells from apoptosis and more specifically from the Fas-mediated apoptotic signal. The antitumoral agent edelfosine sensitizes leukemic cells to death by inducing the redistribution of the apoptotic receptor Fas into plasma membrane subdomains called lipid rafts. Herein, we show that inhibition of the PI3K signal by edelfosine triggers a Fas-mediated apoptotic signal independently of the Fas/FasL interaction. Furthermore, similarly to edelfosine, blockade of the PI3K activity, using specific inhibitors LY294002 and wortmannin, leads to the clustering of Fas whose supramolecular complex is colocalized within the lipid rafts. These findings indicate that the antitumoral agent edelfosine down-modulates the PI3K signal to sensitize tumor cells to death through the redistribution of Fas into large platform of membrane rafts.


Subject(s)
Membrane Microdomains/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction , fas Receptor/metabolism , Cell Death/drug effects , Cell Line, Tumor , Down-Regulation , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Fas Ligand Protein/metabolism , Humans , Membrane Microdomains/drug effects , Phosphoinositide-3 Kinase Inhibitors , Phospholipid Ethers/pharmacology , Protein Transport/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects
16.
Am J Ophthalmol ; 207: 319-325, 2019 11.
Article in English | MEDLINE | ID: mdl-31323199

ABSTRACT

PURPOSE: To investigate the potential of utilizing the expression of genes for glucocorticoid receptor (GR) and mitogen-activated protein kinase phosphatase-1 (MKP-1) as biomarkers of corticosteroid (CS) refractoriness and disease activity in patients with Vogt-Koyanagi-Harada (VKH) disease. DESIGN: Prospective cohort study. METHODS: Twenty VKH patients receiving their first cycle of CS treatment in the absence of additional systemic immunosuppressive therapy and a control group of fifteen healthy volunteers were recruited from the University of Chile (Santiago, Chile) and US National Institutes of Health (Bethesda, United States). Intraocular inflammation was clinically quantified at enrolment and all follow-up visits. CS refractoriness was defined as an ocular reactivation of VKH upon CS withdrawal at a daily oral prednisone dose of 10 mg or more. Quantitative Reverse transcription polymerase chain reaction (qRT-PCR) was performed to measure the mRNA levels of the alpha (α) and beta (ß) isoforms of GR and MKP-1 in peripheral blood mononuclear cells (PBMC) after in vitro stimulation with either anti-CD3/anti-CD28 antibodies, lipopolysaccharide (LPS), or phytohemagglutinin (PHA), in the presence or absence of dexamethasone (Dex). RESULTS: After 6 hours of stimulation in the presence of Dex, PBMC from CS-refractory VKH patients had an impaired elevation in GRα expression (P = .03). Furthermore, inactive patients showed a significant Dex-induced upregulation of MKP-1 (P = .005). CONCLUSIONS: In this pilot study, the expression of GR isoforms and MKP-1 corresponded with patients' clinical response to systemic CS treatment and disease activity, respectively. Hence, these candidate biomarkers have potential clinical utility in the early identification of CS refractoriness and subclinical inflammation in patients with VKH disease.


Subject(s)
Biomarkers/metabolism , Dual Specificity Phosphatase 1/metabolism , Glucocorticoids/therapeutic use , Leukocytes, Mononuclear/metabolism , Receptors, Glucocorticoid/metabolism , Uveomeningoencephalitic Syndrome/blood , Uveomeningoencephalitic Syndrome/drug therapy , Adult , Dual Specificity Phosphatase 1/genetics , Female , Gene Expression Regulation/physiology , Humans , Male , Middle Aged , Pilot Projects , Prednisone/therapeutic use , Prospective Studies , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptors, Glucocorticoid/genetics
17.
Arch Immunol Ther Exp (Warsz) ; 56(1): 9-14, 2008.
Article in English | MEDLINE | ID: mdl-18250974

ABSTRACT

Fas (CD95/APO-1) belongs to the tumor necrosis factor receptor family and its signaling pathway has been extensively studied over the past 15 years. Blockade of the Fas-mediated apoptotic signal leads to abusive lymphoproliferation, auto-immunity, and an increased risk of developing lymphoma and leukemia. Fas engagement drives the formation of a complex termed DISC (death-inducing signaling complex), which contains the adaptor molecule Fas-associated protein, two members of the caspase family caspase-8 and -10, and a pseudo-caspase termed c-FLIP. According to different authors, DISC formation relies either on the redistribution of Fas into the lipid rafts or the recruitment of the actin cytoskeleton and receptor endocytosis or the production of ceramide. However, the accurate molecular ordering upstream from the formation of DISC remains very puzzling and is highly debated. Herein we review some of the factors that would potentially facilitate or limit the formation of the DISC.


Subject(s)
Apoptosis/physiology , Death Domain Receptor Signaling Adaptor Proteins/physiology , Membrane Microdomains/metabolism , Signal Transduction/physiology , fas Receptor/physiology , Animals , Apoptosis/drug effects , Ceramides/biosynthesis , Cytoskeleton/metabolism , Endocytosis/physiology , Fas Ligand Protein/physiology , Humans , Lymphoproliferative Disorders/etiology , Lymphoproliferative Disorders/metabolism , Models, Immunological , Receptor Aggregation , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
18.
PLoS One ; 13(10): e0205291, 2018.
Article in English | MEDLINE | ID: mdl-30379838

ABSTRACT

New cytometric techniques continue to push the boundaries of multi-parameter quantitative data acquisition at the single-cell level particularly in immunology and medicine. Sophisticated analysis methods for such ever higher dimensional datasets are rapidly emerging, with advanced data representations and dimensional reduction approaches. However, these are not yet standardized and clinical scientists and cell biologists are not yet experienced in their interpretation. More fundamentally their range of statistical validity is not yet fully established. We therefore propose a new method for the automated and unbiased analysis of high-dimensional single cell datasets that is simple and robust, with the goal of reducing this complex information into a familiar 2D scatter plot representation that is of immediate utility to a range of biomedical and clinical settings. Using publicly available flow cytometry and mass cytometry datasets we demonstrate that this method (termed CytoBinning), recapitulates the results of traditional manual cytometric analyses and leads to new and testable hypotheses.


Subject(s)
Aging/immunology , Flow Cytometry/statistics & numerical data , Image Cytometry/statistics & numerical data , Pattern Recognition, Automated/statistics & numerical data , Single-Cell Analysis/statistics & numerical data , Biomarkers/analysis , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8 Antigens/genetics , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/immunology , Datasets as Topic , Female , Gene Expression , Humans , Immunity, Innate , Male , Receptors, CCR7/genetics , Receptors, CCR7/immunology , Single-Cell Analysis/methods
19.
Invest Ophthalmol Vis Sci ; 58(9): 3600-3607, 2017 07 01.
Article in English | MEDLINE | ID: mdl-28715846

ABSTRACT

Purpose: Autoimmune retinopathy (AIR) is a retinopathy associated with unexplained vision loss presumably linked to circulating antiretinal antibodies; currently, however, there are no standardized criteria regarding the diagnosis, treatment strategy, or pathogenesis of this disease. The importance of B-lymphocyte immunophenotyping in the classification of AIR is unknown. Methods: We utilized 15-color multiparametric flow cytometry to identify aberrations in B cell subsets that may contribute to the pathophysiology of AIR. Luminex cytokine analysis was also performed on plasma samples from AIR patients. Results: Significant differences in AIR patients compared to individuals with other inflammatory conditions or healthy donors were found in the B cell memory compartment, including an increase in naïve B cells and a decrease in switched and unswitched memory B cells, which correlated with alterations in immunoglobulin secretion. Conclusions: These findings suggest that the maturation process of B cells may be impaired and that B cell immunophenotyping may help in understanding disease process in AIR.


Subject(s)
Autoimmune Diseases/immunology , B-Lymphocyte Subsets/immunology , Retinal Diseases/immunology , Adult , Aged , Aged, 80 and over , Autoantibodies/blood , Autoantigens/immunology , Autoimmune Diseases/pathology , B-Lymphocyte Subsets/pathology , Cytokines/blood , Electroretinography , Female , Flow Cytometry , Healthy Volunteers , Humans , Immunophenotyping , Male , Middle Aged , Panuveitis/blood , Retina/immunology , Retinal Diseases/pathology , Uveitis, Posterior/blood , Vision Disorders/diagnosis , Visual Fields
20.
Invest Ophthalmol Vis Sci ; 57(4): 1765-72, 2016 Apr.
Article in English | MEDLINE | ID: mdl-27070110

ABSTRACT

PURPOSE: To test the association between elevated proportions of CD1c+ myeloid dendritic cells (mDCs) and disease activation/reactivation in noninfectious uveitis. METHODS: Noninfectious uveitis patients (n = 89) and healthy controls (n = 111) were recruited. The proportion of CD1c+ mDCs in the total dendritic cell (DC) population of peripheral blood was measured by flow cytometry (CD1c+ mDCs gated on Lineage 1+HLADR+ DCs). Disease activity was assessed per Standardization of Uveitis Nomenclature criteria. Uveitis reactivation was ascribed to clinically quiescent patients who developed reactivation of intraocular inflammation within 6 months. RESULTS: The proportions of CD1c+ mDCs were increased in noninfectious uveitis patients, especially in active disease, compared to healthy controls. This CD1c+ mDC elevation was not associated with underlying systemic diseases, anatomic locations of uveitis, medications, or demographic factors. Longitudinal data showed that the dynamics of CD1c+ mDC levels were correlated with disease activity. The average proportion of CD1c+ mDCs in active uveitis patients was 60% so we set this as the cutoff between high and low CD1c+ mDC levels. Although 74% of quiescent patients had low proportions of CD1c+ mDCs, 26% still had high proportions. Quiescent patients with high CD1c+ mDC proportions showed increased risk of disease reactivation, compared to quiescent patients with low CD1c+ mDC proportions. CONCLUSIONS: Increased proportions of CD1c+ mDCs were associated with clinical activity, and quiescent patients with elevated CD1c+ mDCs were more likely to undergo reactivation. This suggests that CD1c+ mDC proportion may be a potential biomarker for assessing clinical activation and reactivation in noninfectious uveitis.


Subject(s)
Dendritic Cells/metabolism , Immunity, Cellular , Myeloid Cells/metabolism , Uveitis/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Antigens, CD1 , Biomarkers/metabolism , Cell Count/methods , Child , Dendritic Cells/immunology , Dendritic Cells/pathology , Disease Progression , Female , Flow Cytometry , Follow-Up Studies , Glycoproteins , Humans , Male , Middle Aged , Myeloid Cells/immunology , Myeloid Cells/pathology , Recurrence , Retrospective Studies , Uveitis/immunology , Uveitis/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL