Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Cell Calcium ; 47(1): 19-28, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19959227

ABSTRACT

Systemic lupus erythematosus (SLE) T cells exhibit several activation signaling anomalies including defective Ca(2+) response and increased NF-AT nuclear translocation. The duration of the Ca(2+) signal is critical in the activation of specific transcription factors and a sustained Ca(2+) response activates NF-AT. Yet, the distribution of Ca(2+) responses in SLE T cells is not known. Furthermore, the mechanisms responsible for Ca(2+) alterations are not fully understood. Kv1.3 channels control Ca(2+) homeostasis in T cells. We reported a defect in Kv1.3 trafficking to the immunological synapse (IS) of SLE T cells that might contribute to the Ca(2+) defect. The present study compares single T cell quantitative Ca(2+) responses upon formation of the IS in SLE, normal, and rheumatoid arthritis (RA) donors. Also, we correlated cytosolic Ca(2+) concentrations and Kv1.3 trafficking in the IS by two-photon microscopy. We found that sustained [Ca(2+)](i) elevations constitute the predominant response to antigen stimulation of SLE T cells. This defect is selective to SLE as it was not observed in RA T cells. Further, we observed that in normal T cells termination of Ca(2+) influx is accompanied by Kv1.3 permanence in the IS, while Kv1.3 premature exit from the IS correlates with sustained Ca(2+) responses in SLE T cells. Thus, we propose that Kv1.3 trafficking abnormalities contribute to the altered distribution in Ca(2+) signaling in SLE T cells. Overall these defects may explain in part the T cell hyperactivity and dysfunction documented in SLE patients.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Calcium Signaling/immunology , Kv1.3 Potassium Channel/metabolism , Lupus Erythematosus, Systemic/immunology , T-Lymphocyte Subsets/metabolism , Adult , Arthritis, Rheumatoid/immunology , Arthritis, Rheumatoid/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , Calcium Signaling/drug effects , Cell Line, Transformed , Female , Humans , Immunological Synapses/immunology , Kv1.3 Potassium Channel/immunology , Lupus Erythematosus, Systemic/metabolism , Lymphocyte Activation/drug effects , Lymphocyte Activation/physiology , Male , Middle Aged , Potassium Channel Blockers/pharmacology , Protein Transport/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/pathology
2.
J Immunol ; 179(1): 346-56, 2007 Jul 01.
Article in English | MEDLINE | ID: mdl-17579055

ABSTRACT

Aberrant T cell responses during T cell activation and immunological synapse (IS) formation have been described in systemic lupus erythematosus (SLE). Kv1.3 potassium channels are expressed in T cells where they compartmentalize at the IS and play a key role in T cell activation by modulating Ca(2+) influx. Although Kv1.3 channels have such an important role in T cell function, their potential involvement in the etiology and progression of SLE remains unknown. This study compares the K channel phenotype and the dynamics of Kv1.3 compartmentalization in the IS of normal and SLE human T cells. IS formation was induced by 1-30 min exposure to either anti-CD3/CD28 Ab-coated beads or EBV-infected B cells. We found that although the level of Kv1.3 channel expression and their activity in SLE T cells is similar to normal resting T cells, the kinetics of Kv1.3 compartmentalization in the IS are markedly different. In healthy resting T cells, Kv1.3 channels are progressively recruited and maintained in the IS for at least 30 min from synapse formation. In contrast, SLE, but not rheumatoid arthritis, T cells show faster kinetics with maximum Kv1.3 recruitment at 1 min and movement out of the IS by 15 min after activation. These kinetics resemble preactivated healthy T cells, but the K channel phenotype of SLE T cells is identical to resting T cells, where Kv1.3 constitutes the dominant K conductance. The defective temporal and spatial Kv1.3 distribution that we observed may contribute to the abnormal functions of SLE T cells.


Subject(s)
Antigen-Presenting Cells/immunology , Cell Communication/immunology , Kv1.3 Potassium Channel/metabolism , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/metabolism , T-Lymphocyte Subsets/immunology , Adult , Aged , Antigen-Presenting Cells/metabolism , Antigen-Presenting Cells/pathology , Calcium Signaling/immunology , Female , Gene Rearrangement, T-Lymphocyte , Homeostasis/immunology , Humans , Immunophenotyping , Kinetics , Kv1.3 Potassium Channel/biosynthesis , Kv1.3 Potassium Channel/physiology , Lupus Erythematosus, Systemic/pathology , Lymphocyte Activation/immunology , Male , Middle Aged , Protein Binding/immunology , Protein Transport/immunology , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Resting Phase, Cell Cycle/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocyte Subsets/pathology
3.
Blood ; 109(1): 383-5, 2007 Jan 01.
Article in English | MEDLINE | ID: mdl-16990602

ABSTRACT

Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome is a rare, fatal autoimmune disorder caused by mutations in the FOXP3 gene leading to the disruption of signaling pathways involved in regulatory T-lymphocyte function. Lifelong multiagent immunosuppression is necessary to control debilitating autoimmune manifestations such as colitis and food allergies. Allogeneic hematopoietic stem cell transplantation (HSCT) can restore T-cell regulatory function but has been previously associated with poor outcome. We describe successful HSCT in 4 patients with IPEX syndrome using a novel reduced-intensity conditioning regimen that resulted in stable donor engraftment, reconstitution of FOXP3+ T regulatory CD4+ cells, and amelioration of gastrointestinal symptoms.


Subject(s)
Bone Marrow Transplantation , Endocrine System Diseases/surgery , Ichthyosis, X-Linked/surgery , Immunologic Deficiency Syndromes/surgery , Transplantation Conditioning/methods , Alemtuzumab , Antibodies, Monoclonal , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm , Child , Child, Preschool , Colitis/genetics , Endocrine System Diseases/genetics , Food Hypersensitivity/genetics , Forkhead Transcription Factors/deficiency , Genes, X-Linked , Graft Survival , Humans , Ichthyosis, X-Linked/genetics , Immunologic Deficiency Syndromes/genetics , Infant , Leukocyte Count , Melphalan , Postoperative Complications , Reoperation , Syndrome , Transplantation, Homologous , Vidarabine/analogs & derivatives
4.
J Pediatr ; 149(1): 134-7, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16860143

ABSTRACT

Mutations of the perforin gene (PRF1) are present in a proportion of patients with hemophagocytic lymphohistiocytosis (HLH). We found that all identified infants with HLH of African descent (17 from USA, 4 from Europe) have 50delT-PRF1 (16 homozygotes, 5 compound heterozygotes), accounting for the most frequently observed PRF1 mutation. Two additional patients with HLH, self-reporting as Hispanic, carried 50delT, but no Caucasians were identified with 50delT. To test the hypothesis that this mutation represents a single haplotype, DNA from 23 patients with HLH and 30 African-American control subjects was sequenced for the PRF1 gene, including portions of the intron containing known single nucleotide polymorphisms (SNPs). The same groups were genotyped at 3 microsatellites proximal to PRF1. The SNP profiles of patients with 50delT-PRF1 were identical, and 5 novel SNPs were identified among African-American control subjects. Patients with 50delT-PRF1 were also found to have had an earlier age of disease onset than patients with other PRF1 mutations. Extent of haplotype sharing and variability of microsatellite alleles in 50delT-PRF1 chromosomes suggest that this mutation arose approximately 1000 to 4000 years ago and is restricted to patients of African descent.


Subject(s)
Black People/genetics , Haplotypes , Lymphohistiocytosis, Hemophagocytic/genetics , Membrane Glycoproteins/genetics , Mutation , Case-Control Studies , Child , Child, Preschool , Europe , Hispanic or Latino/genetics , Humans , Infant , Lymphohistiocytosis, Hemophagocytic/ethnology , Microsatellite Repeats , Ohio , Perforin , Pore Forming Cytotoxic Proteins , Sequence Analysis, DNA
5.
J Physiol ; 573(Pt 2): 357-70, 2006 Jun 01.
Article in English | MEDLINE | ID: mdl-16600997

ABSTRACT

T lymphocytes encounter hypoxia when they migrate to pathological sites such as tumours and wounds. The inability of T cells to provide an efficient defence at these sites can in part be explained by the hypoxic environment. Kv 1.3 channels, important components of the T cell activation process are inhibited by hypoxia and their inhibition accounts for a hypoxia-induced decrease in T cell proliferation. Although Kv 1.3 channels play a key role in T cell O(2) sensing, the signalling mechanisms mediating their response to hypoxia are still not understood. In this study, we show that the src-protein tyrosine kinase p56Lck (Lck) is required for Kv 1.3 channel response to hypoxia. Pre-exposure to the src inhibitor PP2 abolished the hypoxia-induced inhibition of Kv 1.3 channels in primary human T lymphocytes. Moreover, Kv 1.3 channel sensitivity to hypoxia was lost in Lck-deficient Jurkat T cells. Further studies with recombinant Kv 1.3 channels showed that Kv 1.3 channels lack intrinsic O(2) sensitivity, but delivery of Lck into the cells and transfection of a constitutively active Lck (Y505FLck) restored their sensitivity to hypoxia. Although Lck is necessary for the Kv 1.3 channel response to hypoxia, it does not directly inhibit Kv 1.3 channels. Indeed, under normal oxygen tension, delivery of active Lck into L929 cells and overexpression of Y505FLck did not decrease recombinant Kv 1.3 currents. On the contrary, activation of endogenous src kinases increased wild-type Kv 1.3 currents in T lymphocytes. Our findings indicate that Lck is required for the acute response to hypoxia of human T lymphocytes as it is necessary to confer O(2) sensitivity on Kv 1.3 channels.


Subject(s)
Kv1.3 Potassium Channel/physiology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/physiology , Oxygen/physiology , Signal Transduction/physiology , T-Lymphocytes/physiology , Cell Hypoxia/physiology , Humans , Male , T-Lymphocytes/enzymology
6.
J Physiol ; 564(Pt 1): 131-43, 2005 Apr 01.
Article in English | MEDLINE | ID: mdl-15677684

ABSTRACT

T lymphocytes are exposed to hypoxia during their development and when they migrate to hypoxic pathological sites. Although it has been shown that hypoxia inhibits Kv1.3 channels and proliferation in human T cells, the mechanisms by which hypoxia regulates T cell activation are not fully understood. Herein we test the hypothesis that hypoxic inhibition of Kv1.3 channels induces membrane depolarization, thus modulating the increase in cytoplasmic Ca2+ that occurs during activation. Hypoxia causes membrane depolarization in human CD3+ T cells, as measured by fluorescence-activated cell sorting (FACS) with the voltage-sensitive dye DiBAC4(3). Similar depolarization is produced by the selective Kv1.3 channel blockers ShK-Dap22 and margatoxin. Furthermore, pre-exposure to such blockers prevents any further depolarization by hypoxia. Since membrane depolarization is unfavourable to the influx of Ca2+ through the CRAC channels (necessary to drive many events in T cell activation such as cytokine production and proliferation), the effect of hypoxia on T cell receptor-mediated increase in cytoplasmic Ca2+ was determined using fura-2. Hypoxia depresses the increase in Ca2+ induced by anti-CD3/CD28 antibodies in approximately 50% of lymphocytes. In the remaining cells, hypoxia either did not elicit any change or produced a small increase in cytoplasmic Ca2+. Similar effects were observed in resting and pre-activated CD3+ cells and were mimicked by ShK-Dap22. These effects appear to be mediated solely by Kv1.3 channels, as we find no influence of hypoxia on IKCa1 and CRAC channels. Our findings indicate that hypoxia modulates Ca2+ homeostasis in T cells via Kv1.3 channel inhibition and membrane depolarization.


Subject(s)
Lymphocyte Activation/physiology , Potassium Channels, Voltage-Gated/physiology , Receptors, Antigen, T-Cell/physiology , T-Lymphocytes/metabolism , Cell Hypoxia/physiology , Humans , Kv1.3 Potassium Channel , Membrane Potentials/physiology , Signal Transduction/immunology , T-Lymphocytes/physiology , Time Factors
7.
Blood ; 105(8): 3066-71, 2005 Apr 15.
Article in English | MEDLINE | ID: mdl-15632210

ABSTRACT

Mutations in the SH2D1A gene have been described in most patients with the clinical syndrome of X-linked lymphoproliferative disease (XLP). The diagnosis of XLP is still difficult given its clinical heterogeneity and the lack of a readily available rapid diagnostic laboratory test, particularly in patients without a family history of XLP. XLP should always be a consideration in males with Epstein-Barr virus-associated hemophagocytic lymphohistiocytosis (EBV-HLH). Four-color flow cytometric analysis was used to establish normal patterns of SH2D1A protein expression in lymphocyte subsets for healthy subjects. Three of 4 patients with XLP, as confirmed by the detection of mutations in the SH2D1A gene, had minimal intracellular SH2D1A protein in all cytotoxic cell types. The remaining patient lacked intracellular SH2D1A protein in CD56+ natural killer (NK) and T lymphocytes and had an abnormal bimodal pattern in CD8+ T cells. Carriers of SH2D1A mutations had decreased SH2D1A protein staining patterns compared with healthy controls. Eleven males with clinical syndromes consistent with XLP, predominantly EBV-HLH, had patterns of SH2D1A protein expression similar to those of healthy controls. Four-color flow cytometry provides diagnostic information that may speed the identification of this fatal disease, differentiating it from other causes of EBV-HLH.


Subject(s)
Flow Cytometry/methods , Intracellular Signaling Peptides and Proteins/metabolism , Lymphoproliferative Disorders/diagnosis , Lymphoproliferative Disorders/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Adolescent , Adult , Child , Child, Preschool , Family , Female , Genetic Testing , Humans , Immunoblotting , Infant , Intracellular Signaling Peptides and Proteins/genetics , Lymphoproliferative Disorders/genetics , Male , Mutation , Reverse Transcriptase Polymerase Chain Reaction , Signaling Lymphocytic Activation Molecule Associated Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...