Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 452
Filter
Add more filters

Publication year range
1.
Eur J Immunol ; 54(5): e2350450, 2024 May.
Article in English | MEDLINE | ID: mdl-38356202

ABSTRACT

The Wiskott-Aldrich syndrome protein (WASp) regulates actin cytoskeletal dynamics and function of hematopoietic cells. Mutations in the WAS gene lead to two different syndromes; Wiskott-Aldrich syndrome (WAS) caused by loss-of-function mutations, and X-linked neutropenia (XLN) caused by gain-of-function mutations. We previously showed that WASp-deficient mice have a decreased number of regulatory T (Treg) cells in the thymus and the periphery. We here evaluated the impact of WASp mutations on Treg cells in the thymus of WAS and XLN mouse models. Using in vitro Treg differentiation assays, WAS CD4 single-positive thymocytes have decreased differentiation to Treg cells, despite normal early signaling upon IL-2 and TGF-ß stimulation. They failed to proliferate and express CD25 at high levels, leading to poor survival and a lower number of Foxp3+ Treg cells. Conversely, XLN CD4 single-positive thymocytes efficiently differentiate into Foxp3+ Treg cells following a high proliferative response to IL-2 and TGF-ß, associated with high CD25 expression when compared with WT cells. Altogether, these results show that specific mutations of WASp affect Treg cell development differently, demonstrating a critical role of WASp activity in supporting Treg cell development and expansion.


Subject(s)
Cell Differentiation , Cell Proliferation , T-Lymphocytes, Regulatory , Thymus Gland , Wiskott-Aldrich Syndrome Protein , Animals , T-Lymphocytes, Regulatory/immunology , Cell Differentiation/immunology , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism , Mice , Thymus Gland/immunology , Thymus Gland/cytology , Forkhead Transcription Factors/metabolism , Forkhead Transcription Factors/genetics , Interleukin-2/metabolism , Interleukin-2/immunology , Mutation , Transforming Growth Factor beta/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-2 Receptor alpha Subunit/genetics , Mice, Knockout , Mice, Inbred C57BL
2.
EMBO J ; 39(5): e102783, 2020 03 02.
Article in English | MEDLINE | ID: mdl-31894880

ABSTRACT

When migratory T cells encounter antigen-presenting cells (APCs), they arrest and form radially symmetric, stable intercellular junctions termed immunological synapses which facilitate exchange of crucial biochemical information and are critical for T-cell immunity. While the cellular processes underlying synapse formation have been well characterized, those that maintain the symmetry, and thereby the stability of the synapse, remain unknown. Here we identify an antigen-triggered mechanism that actively promotes T-cell synapse symmetry by generating cytoskeletal tension in the plane of the synapse through focal nucleation of actin via Wiskott-Aldrich syndrome protein (WASP), and contraction of the resultant actin filaments by myosin II. Following T-cell activation, WASP is degraded, leading to cytoskeletal unraveling and tension decay, which result in synapse breaking. Thus, our study identifies and characterizes a mechanical program within otherwise highly motile T cells that sustains the symmetry and stability of the T cell-APC synaptic contact.


Subject(s)
Antigen-Presenting Cells/metabolism , Immunological Synapses/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/metabolism , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Antigen-Presenting Cells/immunology , Cell Movement , Cytoskeleton/metabolism , Humans , Lymphocyte Activation , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein/genetics
3.
Scand J Immunol ; 95(1): e13115, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34758123

ABSTRACT

Wiskott-Aldrich syndrome (WAS) also called the eczema-thrombocytopenia-immunodeficiency syndrome is a primary immunodeficiency disease with X-linked recessive inheritance caused by mutations in the WAS protein (WASp) gene and characterized by thrombocytopenia with reduced platelet volume, eczema, immunodeficiency, and increased risk of malignant tumours. The mutations will lead to separate WAS severity which can be typical severe 'classical' WAS or less severe 'non-classical' WAS. This article will review and analyse clinical and immune characteristics of five unrelated Chinese families harbouring classical and non-classical WAS. The expression of WASp was detected in the peripheral blood monocytes (PBMC) by flow cytometry, and five mutations were found by WAS gene sequencing, one of which had not been reported in the literature, namely frameshift mutation c.1240_1247delCCACTCCC (p. P414Sfs*41).


Subject(s)
Leukocytes, Mononuclear/metabolism , Mutation/genetics , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/immunology , China , DNA Mutational Analysis , Eczema , Family , Female , Humans , Infant , Leukocytes, Mononuclear/immunology , Male , Mean Platelet Volume , Thrombocytopenia , Wiskott-Aldrich Syndrome/genetics
4.
J Allergy Clin Immunol ; 145(1): 324-334, 2020 01.
Article in English | MEDLINE | ID: mdl-31604087

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is an X-linked primary immune deficiency disorder resulting from Wiskott-Aldrich syndrome protein (WASp) deficiency. Lymphocytes from patients with WAS manifest increased DNA damage and lymphopenia from cell death, yet how WASp influences DNA damage-linked cell survival is unknown. A recently described mechanism promoting cell survival after ionizing radiation (IR)-induced DNA damage involves fragmentation and dispersal of the Golgi apparatus, known as the Golgi-dispersal response (GDR), which uses the Golgi phosphoprotein 3 (GOLPH3)-DNA-dependent protein kinase (DNA-PK)-myosin XVIIIA-F-actin signaling pathway. OBJECTIVE: We sought to define WASp's role in the DNA damage-induced GDR and its disruption as a contributor to the development of radiosensitivity-linked immunodeficiency in patients with WAS. METHODS: In human TH and B-cell culture systems, DNA damage-induced GDR elicited by IR or radiomimetic chemotherapy was monitored in the presence or absence of WASp or GOLPH3 alone or both together. RESULTS: WASp deficiency completely prevents the development of IR-induced GDR in human TH and B cells, despite the high DNA damage load. Loss of WASp impedes nuclear translocation of GOLPH3 and its colocalization with the DNA-dependent protein kinase catalytic subunit (DNA-PKcs). Surprisingly, however, depletion of GOLPH3 alone or depolymerization of F-actin in WASp-sufficient TH cells still allows development of robust GDR, suggesting that WASp, but not GOLPH3, is essential for GDR and cell survival after IR-induced DNA-damage in human lymphocytes. CONCLUSION: The study identifies WASp as a novel effector of the nucleus-to-Golgi cell-survival pathway triggered by IR-induced DNA damage in cells of the hematolymphoid lineage and proposes an impaired GDR as a new cause for development of a "radiosensitive" form of immune dysregulation in patients with WAS.


Subject(s)
B-Lymphocytes/immunology , DNA Damage/immunology , Golgi Apparatus/immunology , Signal Transduction/immunology , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein Family/immunology , DNA Damage/genetics , DNA-Activated Protein Kinase/genetics , DNA-Activated Protein Kinase/immunology , Golgi Apparatus/genetics , Humans , Membrane Proteins/genetics , Membrane Proteins/immunology , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein Family/genetics
5.
Clin Immunol ; 219: 108573, 2020 10.
Article in English | MEDLINE | ID: mdl-32814211

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency disorder (PID) characterized by microthrombocytopenia, bloody diarrhea, eczema, recurrent infections, and a high incidence of autoimmunity and malignancy. OBJECTIVE: To investigate the mechanism of thrombocytopenia and infections in four boys of consanguineous parents from Lebanon. METHODS: Patient gDNA was studied using Next Generation Sequencing and Sanger Sequencing. Protein expression was determined by immunoblotting, and mRNA expression by semi-quantitative RT-PCR. F-actin polymerization and cellular proliferation were assayed by flow cytometry. RESULTS: We identified a threonine to a methionine change at position 45 (T45M) of the WAS protein (WASp) that abolished protein expression and disturbed F-actin polymerization and T cell proliferation, but not B cell proliferation. In addition, the levels of the WAS-interacting protein (WIP) were significantly decreased in the patients. CONCLUSION: The mutation identified severely destabilizes WASp and affects the downstream signaling events important for T cell function, but not B cell function. It was previously known that the stability of WASp depends on WIP. In this manuscript, we report that the stability of WIP also depends on WASp. Finally, it is important to suspect X-linked PIDs even in consanguineous families. CLINICAL IMPLICATIONS: The patients are above the optimal age for transplant in WAS, and it is difficult to identify one or more donors for four patients, therefore, they represent ideal candidates for gene therapy or interleukin-2 therapy.


Subject(s)
Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/genetics , X-Linked Combined Immunodeficiency Diseases/genetics , B-Lymphocytes/immunology , Child , Child, Preschool , Consanguinity , Humans , Lebanon , Male , Mutation , Siblings , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome/immunology , X-Linked Combined Immunodeficiency Diseases/immunology
6.
J Clin Immunol ; 39(8): 786-794, 2019 11.
Article in English | MEDLINE | ID: mdl-31620947

ABSTRACT

BACKGROUND: We undertook a study to determine the impact of Wiskott Aldrich Syndrome (WAS) and X-linked thrombocytopenia (XLT) and their therapies upon the health-related quality of life (HRQOL) of patients and their families. MATERIALS AND METHODS: We undertook a survey of patients and their families, who self-identified as having either WAS or XLT. We assessed the PedsQL™ 4.0, the parent proxy form, and the family impact module. These results were compared with normative data from previously published reports. RESULTS: Sixty-eight patients (29 patients completed both the PedsQL™ 4.0 and the parent proxy form; 21 completed only the PedsQL™ 4.0; and 18 completed only the parent proxy form) were included. In contrast to patient-reported outcomes, parents of patients who had a bone marrow transplant (BMT) reported that their children had better QOL scores compared with those who did not (82.6 vs. 73.3, p = 0.023). The QOL of patients vs. previously published normative data showed decreases in patient scores for psychosocial health (72.62 vs. 86.58, p = < 0.001), emotional functioning (69.91 vs. 82.64, p = < 0.001), social functioning (77.55 vs. 91.56, p = < 0.001), and school functioning (70.46 vs. 85.67, p = < 0.001). The family impact study revealed deficits in emotional, social, and cognitive functioning, communication, and worry. CONCLUSION: These results show that patients with WAS/XLT are significantly impacted with respect to QOL. BMT offered a better QOL for patients according to parents, but not as reported by the patients. Future studies should incorporate QOL to provide more data and a better understanding of outcomes for long-term survivors and decision-making regarding BMT.


Subject(s)
Genetic Diseases, X-Linked/psychology , Parents/psychology , Patient Reported Outcome Measures , Quality of Life , Thrombocytopenia/psychology , Wiskott-Aldrich Syndrome/psychology , Adolescent , Bone Marrow Transplantation , Caregivers/psychology , Child , Child, Preschool , Decision Making , Genetic Diseases, X-Linked/complications , Genetic Diseases, X-Linked/immunology , Genetic Diseases, X-Linked/therapy , Humans , Male , Surveys and Questionnaires/statistics & numerical data , Survivors/psychology , Thrombocytopenia/complications , Thrombocytopenia/immunology , Thrombocytopenia/therapy , Wiskott-Aldrich Syndrome/complications , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/therapy , Young Adult
7.
Cell Immunol ; 338: 43-50, 2019 04.
Article in English | MEDLINE | ID: mdl-30981413

ABSTRACT

Wiskott-Aldrich syndrome (WAS) patients are characterized by immunodeficiency and viral infections. T cells derived from WAS patients and WAS protein (WASP)-deficient mice have various defects. However, whether WASP plays a role in immune control of cytomegalovirus (CMV) infection remains unclear. We analyzed the distribution of CD8+ T subsets and the pathological damage to various organs and tissues in MCMV infected Was knockout (KO) mice. A relatively high number of MCMV-specific cytotoxic T cells (CTLs) were observed in the spleen of Was KO mice. In MCMV infected Was KO mice, the late differentiated CD8+ T subset (CD27-CD28-) decreased in lungs, compared with those in the spleen and peripheral blood. Additionally, we found that the most severe pathological lesions occurred in the lungs, the main target organ of MCMV infection. By stimulating the spleen-derived CD8+ T lymphocytes of Was KO mice, we found that IL-2 and granzyme B production declined compared with that in wild- type mice. Moreover, the number of apoptotic CD8+ T cells increased in Was KO mice compared with the number in wild-type mice. Therefore, our results demonstrate that WASP may be involved in regulating cytotoxic function and apoptosis in CD8+ T cells following MCMV infection, which is supported by the distribution and memory compartment of MCMV-specific T cells in MCMV infected WAS mice.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Herpesviridae Infections/immunology , Lung/pathology , Muromegalovirus/physiology , Wiskott-Aldrich Syndrome Protein/metabolism , Wiskott-Aldrich Syndrome/immunology , Animals , Apoptosis , Cells, Cultured , Cytotoxicity, Immunologic , Disease Models, Animal , Female , Granzymes/metabolism , Humans , Interleukin-2/metabolism , Male , Mice , Mice, Knockout , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome Protein/genetics
8.
Cell Immunol ; 341: 103919, 2019 07.
Article in English | MEDLINE | ID: mdl-31047647

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is a form of primary immunodeficiency (PIDs) resulting from mutations of the gene that encodes Wiskott-Aldrich syndrome protein (WASp). WASp is the first identified and most widely studied protein belonging to the actin nucleation-promoting factor family and plays significant role in integrating and transforming signals from critical receptors on the cell surface to actin remodeling. WASp functions in immune defense and homeostasis through the regulation of actin cytoskeleton-dependent cellular processes as well as processes uncoupled with actin polymerization like nuclear transcription programs. In this article, we review the mechanisms of WASp activation through an understanding of its structure. We further discuss the role of WASp in adaptive immunity, paying special attention to some recent findings on the crucial role of WASp in the formation of immunological synapse, the regulation of T follicular helper (Tfh) cells and in the prevention of autoimmunity.


Subject(s)
Actin Cytoskeleton/immunology , B-Lymphocytes/immunology , Homeostasis/immunology , T-Lymphocytes, Helper-Inducer/immunology , Wiskott-Aldrich Syndrome Protein/immunology , Wiskott-Aldrich Syndrome/immunology , Actin Cytoskeleton/genetics , Adaptive Immunity , Animals , Autoimmunity/genetics , B-Lymphocytes/pathology , Disease Models, Animal , Gene Expression Regulation , Homeostasis/genetics , Humans , Immunity, Innate , Immunological Synapses/genetics , Mice , Signal Transduction , T-Lymphocytes, Helper-Inducer/pathology , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/genetics
10.
J Allergy Clin Immunol ; 142(4): 1272-1284, 2018 10.
Article in English | MEDLINE | ID: mdl-29421274

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is an X-linked immunodeficiency characterized by eczema, infections, and susceptibility to autoimmunity and malignancies. Thrombocytopenia is a constant finding, but its pathogenesis remains elusive. OBJECTIVE: To dissect the basis of the WAS platelet defect, we used a novel conditional mouse model (CoWas) lacking Wiskott-Aldrich syndrome protein (WASp) only in the megakaryocytic lineage in the presence of a normal immunologic environment, and in parallel we analyzed samples obtained from patients with WAS. METHODS: Phenotypic and functional characterization of megakaryocytes and platelets in mutant CoWas mice and patients with WAS with and without autoantibodies was performed. Platelet antigen expression was examined through a protein expression profile and cluster proteomic interaction network. Platelet immunogenicity was tested by using ELISAs and B-cell and platelet cocultures. RESULTS: CoWas mice showed increased megakaryocyte numbers and normal thrombopoiesis in vitro, but WASp-deficient platelets had short lifespan and high expression of activation markers. Proteomic analysis identified signatures compatible with defects in cytoskeletal reorganization and metabolism yet surprisingly increased antigen-processing capabilities. In addition, WASp-deficient platelets expressed high levels of surface and soluble CD40 ligand and were capable of inducing B-cell activation in vitro. WASp-deficient platelets were highly immunostimulatory in mice and triggered the generation of antibodies specific for WASp-deficient platelets, even in the context of a normal immune system. Patients with WAS also showed platelet hyperactivation and increased plasma soluble CD40 ligand levels correlating with the presence of autoantibodies. CONCLUSION: Overall, these findings suggest that intrinsic defects in WASp-deficient platelets decrease their lifespan and dysregulate immune responses, corroborating the role of platelets as modulators of inflammation and immunity.


Subject(s)
Blood Platelets/immunology , Wiskott-Aldrich Syndrome/immunology , Adolescent , Adult , Animals , Autoimmunity , CD40 Ligand/immunology , Child , Child, Preschool , Female , Humans , Infant , Inflammation/blood , Inflammation/immunology , Mice, Inbred C57BL , Mice, Knockout , Platelet Count , Wiskott-Aldrich Syndrome/blood , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/immunology , Young Adult
11.
J Allergy Clin Immunol ; 142(5): 1605-1617.e4, 2018 11.
Article in English | MEDLINE | ID: mdl-29447842

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is a rare primary immunodeficiency caused by mutations in Wiskott-Aldrich syndrome protein (WASp), a key regulator of cytoskeletal dynamics in hematopoietic cells. A high proportion of patients experience autoimmunity caused by a breakdown in T- and B-cell tolerance. Moreover, excessive production of type I interferon (IFN-I) by plasmacytoid dendritic cells (pDCs) contributes to autoimmune signs; however, the factors that trigger excessive innate activation have not been defined. OBJECTIVE: Neutrophil extracellular traps (NETs) emerged as major initiating factors in patients with diseases such as systemic lupus erythematosus and rheumatoid arthritis. In this study we explored the possible involvement of aberrant neutrophil functions in patients with WAS. METHODS: We evaluated the expression of a set of granulocyte genes associated with NETs in a cohort of patients with WAS and the presence of NET inducers in sera. Using a mouse model of WAS, we analyzed NET release by WASp-null neutrophils and evaluated the composition and homeostasis of neutrophils in vivo. By using depletion experiments, we assessed the effect of neutrophils in promoting inflammation and reactivity against autoantigens. RESULTS: Transcripts of genes encoding neutrophil enzymes and antimicrobial peptides were increased in granulocytes of patients with WAS, and serum-soluble factors triggered NET release. WASp-null neutrophils showed increased spontaneous NETosis, induced IFN-I production by pDCs, and activated B cells through B-cell activating factor. Consistently, their depletion abolished constitutive pDC activation, normalized circulating IFN-I levels, and, importantly, abolished production of autoantibodies directed against double-stranded DNA, nucleosomes, and myeloperoxidase. CONCLUSIONS: These findings reveal that neutrophils are involved in the pathogenic loop that causes excessive activation of innate cells and autoreactive B cells, thus identifying novel mechanisms that contribute to the autoimmunity of WAS.


Subject(s)
Interferon Type I/immunology , Neutrophils/immunology , Wiskott-Aldrich Syndrome/immunology , Adolescent , Adult , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , Child, Preschool , Dendritic Cells/immunology , Extracellular Traps , Female , Gene Expression , Humans , Infant , Male , Mice, Inbred C57BL , Mice, Knockout , Wiskott-Aldrich Syndrome/genetics , Young Adult
12.
Eur J Immunol ; 47(11): 1857-1866, 2017 11.
Article in English | MEDLINE | ID: mdl-28805251

ABSTRACT

The Wiskott-Aldrich syndrome protein (WASP) participates in innate and adaptive immunity through regulation of actin cytoskeleton-dependent cellular processes, including immune synapse formation, cell signaling, migration and cytokine release. There is also emerging evidence for a direct role in nuclear transcription programmes uncoupled from actin polymerization. A deeper understanding of some of the more complex features of Wiskott Aldrich syndrome (WAS) itself, such as the associated autoimmunity and inflammation, has come from identification of defects in the number and function of anti-inflammatory myeloid cells and regulatory T and B cells, as well as defects in positive and negative B-cell selection. In this review we outline the cellular defects that have been characterized in both human WAS patients and murine models of the disease. We will emphasize in particular recent discoveries that provide a mechanistic insight into disease pathology, including lymphoid and myeloid cell homeostasis, immune synapse assembly and immune cell signaling.


Subject(s)
Wiskott-Aldrich Syndrome Protein/immunology , Animals , Humans , Mice , Wiskott-Aldrich Syndrome/immunology
13.
Blood ; 127(2): 216-20, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26468226

ABSTRACT

Mutations of the Wiskott-Aldrich syndrome gene (WAS) are responsible for Wiskott-Aldrich syndrome (WAS), a disease characterized by thrombocytopenia, eczema, immunodeficiency, and autoimmunity. Mice with conditional deficiency of Was in B lymphocytes (B/WcKO) have revealed a critical role for WAS protein (WASP) expression in B lymphocytes in the maintenance of immune homeostasis. Neural WASP (N-WASP) is a broadly expressed homolog of WASP, and regulates B-cell signaling by modulating B-cell receptor (BCR) clustering and internalization. We have generated a double conditional mouse lacking both WASP and N-WASP selectively in B lymphocytes (B/DcKO). Compared with B/WcKO mice, B/DcKO mice showed defective B-lymphocyte proliferation and impaired antibody responses to T-cell-dependent antigens, associated with decreased autoantibody production and lack of autoimmune kidney disease. These results demonstrate that N-WASP expression in B lymphocytes is required for the development of autoimmunity of WAS and may represent a novel therapeutic target in WAS.


Subject(s)
Autoimmunity/genetics , B-Lymphocytes/immunology , Wiskott-Aldrich Syndrome Protein, Neuronal/physiology , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/immunology , Animals , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Deletion , Mice , Mice, Knockout , Receptors, Antigen, B-Cell/metabolism , Signal Transduction/immunology , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein, Neuronal/genetics
14.
Blood ; 127(25): 3180-91, 2016 06 23.
Article in English | MEDLINE | ID: mdl-27170596

ABSTRACT

Wiskott-Aldrich syndrome protein (WASp) is a hematopoietic-specific regulator of actin nucleation. Wiskott-Aldrich syndrome (WAS) patients show immunodeficiencies, most of which have been attributed to defective T-cell functions. T follicular helper (Tfh) cells are the major CD4(+) T-cell subset with specialized B-cell helper capabilities. Aberrant Tfh cells activities are involved in immunopathologies such as autoimmunity, immunodeficiencies, and lymphomas. We found that in WAS patients, the number of circulating Tfh cells was significantly reduced due to reduced proliferation and increased apoptosis, and Tfh cells were Th2 and Th17 polarized. The expression of inducible costimulator (ICOS) in circulating Tfh cells was higher in WAS patients than in controls. BCL6 expression was decreased in total CD4(+) T and Tfh cells of WAS patients. Mirroring the results in patients, the frequency of Tfh cells in WAS knockout (KO) mice was decreased, as was the frequency of BCL6(+) Tfh cells, but the frequency of ICOS(+) Tfh cells was increased. Using WAS chimera mice, we found that the number of ICOS(+) Tfh cells was decreased in WAS chimera mice, indicating that the increase in ICOS(+) Tfh cells in WAS KO mice was cell extrinsic. The data from in vivo CD4(+) naive T-cell adoptive transfer mice as well as in vitro coculture of naive B and Tfh cells showed that the defective function of WASp-deficient Tfh cells was T-cell intrinsic. Consistent findings in both WAS patients and WAS KO mice suggested an essential role for WASp in the development and memory response of Tfh cells and that WASp deficiency causes a deficient differentiation defect in Tfh cells by downregulating the transcription level of BCL6.


Subject(s)
Germinal Center/pathology , T-Lymphocytes, Helper-Inducer/pathology , T-Lymphocytes, Helper-Inducer/physiology , Wiskott-Aldrich Syndrome/immunology , Animals , B-Lymphocytes , CD4-Positive T-Lymphocytes/pathology , CD4-Positive T-Lymphocytes/physiology , Case-Control Studies , Cell Differentiation , Cells, Cultured , Germinal Center/immunology , Humans , Inducible T-Cell Co-Stimulator Protein/metabolism , Interleukins/metabolism , Lymph Nodes/immunology , Lymph Nodes/pathology , Mice , Mice, Knockout , Positive Regulatory Domain I-Binding Factor 1 , Receptors, CXCR5/metabolism , Repressor Proteins/metabolism , Wiskott-Aldrich Syndrome/blood , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/genetics
15.
Blood ; 128(13): 1723-34, 2016 09 29.
Article in English | MEDLINE | ID: mdl-27330000

ABSTRACT

Wiskott-Aldrich syndrome (WAS) pediatric patients exhibit a deficiency in humoral immune memory. However, the mechanism by which Wiskott-Aldrich syndrome protein (WASP) regulates the differentiation and activation of memory B cells remains elusive. Here we examine the early activation events of memory B cells from the peripheral blood mononuclear cells of WAS patients and age-matched healthy controls (HCs) using total internal reflection fluorescence microscopy. In response to stimulation through the B-cell receptor (BCR), memory B cells from HCs showed significantly higher magnitudes of BCR clustering and cell spreading than naive B cells from the same individuals. This was associated with increases in CD19 recruitment to the BCR and the activation of its downstream signaling molecule Btk and decreases in FcγRIIB recruitment and the activation of its downstream molecule Src homology 2-containing inositol 5' phosphatase (SHIP). However, these enhanced signaling activities mediated by CD19 and Btk are blocked in memory B cells from WAS patients, whereas the activation of FcγRIIB and SHIP was increased. Although the expression levels of CD19, Btk, and FcγRIIB did not change between CD27(-) and CD27(+) B cells of HCs, the protein and mRNA levels of CD19 but not Btk and FcγRIIB were significantly reduced in both CD27(-) and CD27(+) B cells of WAS patients, compared with those of HCs. Overall, our study suggests that WASP is required for memory B-cell activation, promoting the activation by positive regulating CD19 transcription and CD19 recruitment to the BCR.


Subject(s)
Antigens, CD19/metabolism , B-Lymphocytes/immunology , Immunologic Memory , Wiskott-Aldrich Syndrome/immunology , Actin Cytoskeleton/metabolism , Antigens, CD19/genetics , B-Lymphocytes/metabolism , Case-Control Studies , Child, Preschool , Down-Regulation , Humans , Infant , Lymphocyte Activation , Mutation , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, IgG/metabolism , Signal Transduction , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome Protein/genetics
16.
Clin Immunol ; 179: 47-53, 2017 06.
Article in English | MEDLINE | ID: mdl-28232030

ABSTRACT

BACKGROUND: Low dose IL-2 can restore the function of T and NK cells from Wiskott-Aldrich (WAS) patients. However, the safety of in vivo IL-2 in WAS is unknown. OBJECTIVES: A phase-I study to assess safety of low dose IL-2 in WAS. METHODS: Patients received 5 daily subcutaneous IL-2 injections, every 2months, for three courses. A "3+3" dose escalation method was used. RESULTS: 6 patients received the 0.5millionunits/m2/day dose without serious adverse events. However, 2 of 3 patients receiving the 1millionunits/m2/day dose developed thrombocytopenia requiring platelet transfusions. A statistically significant platelet increase occurred in patients receiving the 0.5millionunits/m2/day dose. A trend toward higher T, B and NK cell numbers and higher T regulatory cell percentages was observed. CONCLUSION: We have identified a safe IL-2 dose for WAS patients. Additional trials are indicated to study the efficacy of this immunostimulant as a therapy for WAS.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Interleukin-2/administration & dosage , Wiskott-Aldrich Syndrome/drug therapy , Adjuvants, Immunologic/adverse effects , Adjuvants, Immunologic/therapeutic use , Adolescent , B-Lymphocytes/immunology , Child , Child, Preschool , Humans , Interleukin-2/adverse effects , Interleukin-2/therapeutic use , Killer Cells, Natural/immunology , Leukocyte Count , Platelet Count , T-Lymphocytes/immunology , Wiskott-Aldrich Syndrome/blood , Wiskott-Aldrich Syndrome/immunology
18.
Allergy ; 72(12): 1916-1924, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28600891

ABSTRACT

BACKGROUND: Food allergies are a growing health problem, and the development of therapies that prevent disease onset is limited by the lack of adjuvant-free experimental animal models. We compared allergic sensitization in patients with food allergy or Wiskott-Aldrich syndrome (WAS) and defined whether spontaneous disease in Was-/- mice recapitulates the pathology of a conventional disease model and/or human food allergy. METHODS: Comparative ImmunoCAP ISAC microarray was performed in patients with food allergy or WAS. Spontaneous food allergy in Was-/- mice was compared to an adjuvant-based model in wild-type mice (WT-OVA/alum). Intestinal and systemic anaphylaxis was assessed, and the role of the high-affinity IgE Fc receptor (FcεRI) in allergic sensitization was evaluated using Was-/- Fcer1a-/- mice. RESULTS: Polysensitization to food was detected in both WAS and food-allergic patients which was recapitulated in the Was-/- model. Oral administration of ovalbumin (OVA) in Was-/- mice induced low titers of OVA-specific IgE compared to the WT-OVA/alum model. Irrespectively, 79% of Was-/- mice developed allergic diarrhea following oral OVA challenge. Systemic anaphylaxis occurred in Was-/- mice (95%) with a mortality rate >50%. Spontaneous sensitization and intestinal allergy occurred independent of FcεRI expression on mast cells (MCs) and basophils. CONCLUSIONS: Was-/- mice provide a model of food allergy with the advantage of mimicking polysensitization and low food-antigen IgE titers as observed in humans with clinical food allergy. This model will facilitate studies on aberrant immune responses during spontaneous disease development. Our results imply that therapeutic targeting of the IgE/FcεRI activation cascade will not affect sensitization to food.


Subject(s)
Food Hypersensitivity/etiology , Food Hypersensitivity/metabolism , Mast Cells/immunology , Mast Cells/metabolism , Receptors, IgE/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Adult , Allergens/immunology , Anaphylaxis , Animals , Disease Models, Animal , Female , Food Hypersensitivity/diagnosis , Gene Expression , Humans , Immunization , Immunoglobulin E/immunology , Male , Mice , Mice, Knockout , Phenotype , Wiskott-Aldrich Syndrome/diagnosis , Wiskott-Aldrich Syndrome/immunology , Young Adult
19.
Immunol Rev ; 256(1): 282-99, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24117828

ABSTRACT

The importance of the cytoskeleton in mounting a successful immune response is evident from the wide range of defects that occur in actin-related primary immunodeficiencies (PIDs). Studies of these PIDs have revealed a pivotal role for the actin cytoskeleton in almost all stages of immune system function, from hematopoiesis and immune cell development, through to recruitment, migration, intercellular and intracellular signaling, and activation of both innate and adaptive immune responses. The major focus of this review is the immune defects that result from mutations in the Wiskott-Aldrich syndrome gene (WAS), which have a broad impact on many different processes and give rise to clinically heterogeneous immunodeficiencies. We also discuss other related genetic defects and the possibility of identifying new genetic causes of cytoskeletal immunodeficiency.


Subject(s)
Actin Cytoskeleton , Immunologic Deficiency Syndromes/etiology , Actin Cytoskeleton/genetics , Actin Cytoskeleton/immunology , Actin Cytoskeleton/metabolism , Actins/metabolism , Animals , Humans , Immune System/cytology , Immune System/immunology , Immune System/metabolism , Mutation , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome Protein Family/chemistry , Wiskott-Aldrich Syndrome Protein Family/genetics , Wiskott-Aldrich Syndrome Protein Family/metabolism
20.
Eur J Immunol ; 45(10): 2773-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26256668

ABSTRACT

Patients with Wiskott-Aldrich syndrome (WAS) exhibit prominent defects in splenic marginal zone (MZ), resulting in abnormal T-cell-independent antibody responses and increased bacterial infections. B-cell-intrinsic deletion of the affected gene WAS protein (WASp) markedly reduces splenic MZ B cells, without impacting the rate of MZ B-cell development, suggesting that abnormal B-cell retention within the MZ accounts for MZ defects in WAS. Since WASp regulates integrin-dependent actin cytoskeletal rearrangement, we previously hypothesized that defective B-cell integrin function promotes MZ depletion. In contrast, we now report that B-cell-intrinsic deletion of the TLR signaling adaptor MyD88 is sufficient to restore the MZ in WAS. We further identify TLR7, an endosomal single-stranded RNA (ssRNA) receptor, as the MyD88-dependent receptor responsible for WAS MZ depletion. These findings implicate spontaneous activation of MZ B cells by ssRNA-containing self-ligands (likely derived from circulating apoptotic material) as the mechanism underlying MZ depletion in WAS. Together, these data suggest a previously unappreciated role for B-cell intrinsic TLR signals in MZ homeostasis, of relevance to both pathogen responses and to the development of systemic autoimmunity.


Subject(s)
B-Lymphocytes/immunology , Membrane Glycoproteins/immunology , Signal Transduction/immunology , Spleen/immunology , Toll-Like Receptor 7/immunology , Wiskott-Aldrich Syndrome/immunology , Animals , B-Lymphocytes/pathology , Cytoskeleton/genetics , Cytoskeleton/immunology , Disease Models, Animal , Integrins/genetics , Integrins/immunology , Lymphocyte Depletion , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/immunology , Signal Transduction/genetics , Spleen/pathology , Toll-Like Receptor 7/genetics , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/pathology
SELECTION OF CITATIONS
SEARCH DETAIL