Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
2.
Front Immunol ; 11: 603428, 2020.
Article in English | MEDLINE | ID: mdl-33329599

ABSTRACT

In this work we present the case of SARS-CoV-2 infection in a 1.5-year-old boy affected by severe Wiskott-Aldrich Syndrome with previous history of autoinflammatory disease, occurring 5 months after treatment with gene therapy. Before SARS-CoV-2 infection, the patient had obtained engraftment of gene corrected cells, resulting in WASP expression restoration and early immune reconstitution. The patient produced specific immunoglobulins to SARS-CoV-2 at high titer with neutralizing capacity and experienced a mild course of infection, with limited inflammatory complications, despite pre-gene therapy clinical phenotype.


Subject(s)
Antibodies, Neutralizing , Antibodies, Viral , COVID-19 , Genetic Therapy , SARS-CoV-2 , Wiskott-Aldrich Syndrome , Antibodies, Neutralizing/blood , Antibodies, Neutralizing/immunology , Antibodies, Viral/blood , Antibodies, Viral/immunology , COVID-19/blood , COVID-19/immunology , COVID-19/therapy , Humans , Infant , Male , SARS-CoV-2/immunology , SARS-CoV-2/metabolism , Wiskott-Aldrich Syndrome/blood , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/therapy , Wiskott-Aldrich Syndrome Protein/biosynthesis , Wiskott-Aldrich Syndrome Protein/immunology
3.
Front Immunol ; 10: 2262, 2019.
Article in English | MEDLINE | ID: mdl-31608063

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is a rare X-linked primary immunodeficiency characterized by recurrent infections, micro thrombocytopenia, eczema, and a high incidence of autoimmunity and malignancy. A defect in the T cell compartment is thought to be a major cause of immunodeficiency in patients with WAS; However, whether the antigen specific T memory cell is altered has not been extensively studied. Here, we examined the expansion/contraction kinetics of CD8+ memory T cells and their maintenance in WASp-/- mice. The results showed that WAS protein (WASp) is not required for differentiation of CD8+ effector T cells; however, CD8+ T cells from WASp-/- mice were hyperactive, resulting in increased cytokine production. The number of CD8+ T memory cells decreased as mice aged, and CD8+ T cell recall responses and protective immunity were impaired. WASp-deficient CD8+ T cells in bone marrow chimeric mice underwent clonal expansion, but the resulting effector cells failed to survive and differentiate into CD8+ memory T cells. Taken together, these findings indicate that WASp plays an intrinsic role in differentiation of CD8+ memory T cells.


Subject(s)
Apoptosis/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Sialic Acid Binding Ig-like Lectin 3/immunology , Wiskott-Aldrich Syndrome Protein/immunology , Animals , Apoptosis/genetics , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/virology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation/genetics , Cytokines/immunology , Cytokines/metabolism , Humans , Immunologic Memory/genetics , Lymphocytic Choriomeningitis/genetics , Lymphocytic Choriomeningitis/immunology , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Lymphocytic choriomeningitis virus/physiology , Mice , Mice, Knockout , Sialic Acid Binding Ig-like Lectin 3/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/metabolism
4.
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
5.
Sci Immunol ; 4(33)2019 03 22.
Article in English | MEDLINE | ID: mdl-30902904

ABSTRACT

Cytotoxic T lymphocytes (CTLs) kill by forming immunological synapses with target cells and secreting toxic proteases and the pore-forming protein perforin into the intercellular space. Immunological synapses are highly dynamic structures that boost perforin activity by applying mechanical force against the target cell. Here, we used high-resolution imaging and microfabrication to investigate how CTLs exert synaptic forces and coordinate their mechanical output with perforin secretion. Using micropatterned stimulatory substrates that enable synapse growth in three dimensions, we found that perforin release occurs at the base of actin-rich protrusions that extend from central and intermediate locations within the synapse. These protrusions, which depended on the cytoskeletal regulator WASP and the Arp2/3 actin nucleation complex, were required for synaptic force exertion and efficient killing. They also mediated physical deformation of the target cell surface during CTL-target cell interactions. Our results reveal the mechanical basis of cellular cytotoxicity and highlight the functional importance of dynamic, three-dimensional architecture in immune cell-cell interfaces.


Subject(s)
Immunological Synapses/immunology , Perforin/immunology , T-Lymphocytes, Cytotoxic/immunology , Actin-Related Protein 2-3 Complex/immunology , Actins/immunology , Animals , Mice , Wiskott-Aldrich Syndrome Protein/immunology
7.
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
8.
J Cell Biol ; 216(12): 4073-4090, 2017 12 04.
Article in English | MEDLINE | ID: mdl-29150539

ABSTRACT

The antimicrobial defense activity of neutrophils partly depends on their ability to form neutrophil extracellular traps (NETs), but the underlying mechanism controlling NET formation remains unclear. We demonstrate that inhibiting cytoskeletal dynamics with pharmacological agents or by genetic manipulation prevents the degranulation of neutrophils and mitochondrial DNA release required for NET formation. Wiskott-Aldrich syndrome protein-deficient neutrophils are unable to polymerize actin and exhibit a block in both degranulation and DNA release. Similarly, neutrophils with a genetic defect in NADPH oxidase fail to induce either actin and tubulin polymerization or NET formation on activation. Moreover, neutrophils deficient in glutaredoxin 1 (Grx1), an enzyme required for deglutathionylation of actin and tubulin, are unable to polymerize either cytoskeletal network and fail to degranulate or release DNA. Collectively, cytoskeletal dynamics are achieved as a balance between reactive oxygen species-regulated effects on polymerization and glutathionylation on the one hand and the Grx1-mediated deglutathionylation that is required for NET formation on the other.


Subject(s)
Cytoskeleton/immunology , Extracellular Traps/immunology , Glutathione/immunology , Neutrophils/immunology , Reactive Oxygen Species/immunology , Actins/genetics , Actins/immunology , Animals , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cytoskeleton/ultrastructure , DNA, Mitochondrial/immunology , DNA, Mitochondrial/metabolism , Extracellular Traps/chemistry , Extracellular Traps/drug effects , Gene Expression Regulation , Glutaredoxins/genetics , Glutaredoxins/immunology , Glutathione/metabolism , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Homeodomain Proteins/immunology , Humans , Mice , Mice, Transgenic , NADPH Oxidases/genetics , NADPH Oxidases/immunology , Neutrophils/cytology , Neutrophils/drug effects , Oxidation-Reduction , Primary Cell Culture , Reactive Oxygen Species/metabolism , Signal Transduction , Tubulin/genetics , Tubulin/immunology , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/immunology
9.
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
10.
Hum Immunol ; 78(9): 565-573, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28535968

ABSTRACT

Wiskott-Aldrich syndrome (WAS) is a severe and rare primary immunodeficiency. Several studies show that WAS protein (WASp) plays a key role in the function of certain lymphocyte subsets. So far, no study has described distinct immunophenotypic abnormalities associated with WAS; thus the prognostic significance of any such abnormalities is unclear. This study examined many differences in the percentage/absolute numbers of distinct lymphocyte subsets in 20 WAS patients and 20 age/sex-matched healthy controls, and analyzed the association between these abnormalities and clinical disease scores. The results showed that the numbers of CD4+ T cells, B cells, and CD8+ naïve T cells were significantly lower in WAS patients; furthermore, the numbers in WASp-negative patients were lower than those in WASp-positive patients. WAS patients showed a selective reduction in expression of CD19 by naïve and transitional B cells. There was a negative association between the number of B cells and the WAS clinical scores. Also, CD8+ naïve T cell numbers in patients with a score of 3-5A were lower than those in patients with a score of 2. The absence of WASp leads to a reduction in the population of specific lymphocyte subsets; therefore, these findings may help future management of patients with WAS.


Subject(s)
B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Lymphocyte Subsets/immunology , Wiskott-Aldrich Syndrome/immunology , Antigens, CD19/metabolism , Cells, Cultured , Child, Preschool , Humans , Immunologic Memory , Immunophenotyping , Infant , Infant, Newborn , Lymphocyte Count , Wiskott-Aldrich Syndrome/diagnosis , Wiskott-Aldrich Syndrome Protein/immunology
11.
Cell Death Dis ; 7: e2301, 2016 07 21.
Article in English | MEDLINE | ID: mdl-27441653

ABSTRACT

Natural killer (NK) cells are important effector cells of the innate immune system to kill certain virus-infected and transformed cells. Wiskott-Aldrich Syndrome protein (WASP) and SCAR homolog (WASH) has been identified as a member of WASP family proteins implicated in regulating the cytoskeletal reorganization, yet little is known about its function in lymphocytes. Here we demonstrate that WASH is crucial for NK cell cytotoxicity. WASH was found to colocalize with lytic granules upon NK cell activation. Knockdown of WASH expression substantially inhibited polarization and release of lytic granules to the immune synapse, resulting in the impairment of NK cell cytotoxicity. More importantly, our data also define a previously unappreciated mechanism for WASH function, in which Src family kinase Lck can interact with WASH and induce WASH phosphorylation. Mutation of tyrosine residue Y141, identified here as the major site of WASH phosphorylation, partially blocked WASH tyrosine phosphorylation and NK cell cytotoxicity. Taken together, these observations suggest that WASH has a pivotal role for regulation of NK cell cytotoxicity through Lck-mediated Y141 tyrosine phosphorylation.


Subject(s)
Cytotoxicity, Immunologic/genetics , Immunological Synapses , Killer Cells, Natural/immunology , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/immunology , Microfilament Proteins/genetics , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Line , Coculture Techniques , Cytoplasmic Granules/immunology , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Killer Cells, Natural/cytology , Lymphocyte Activation , Lymphocyte Specific Protein Tyrosine Kinase p56(lck)/genetics , Microfilament Proteins/antagonists & inhibitors , Microfilament Proteins/immunology , Mutation , Phosphorylation , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Tyrosine/immunology , Tyrosine/metabolism , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome Protein/immunology
12.
Sci Rep ; 6: 25629, 2016 05 16.
Article in English | MEDLINE | ID: mdl-27181049

ABSTRACT

CD23 has been implicated as a negative regulator of IgE and IgG antibody responses. However, whether CD23 has any role in B-cell activation remains unclear. We examined the expression of CD23 in different subsets of peripheral B cells and the impact of CD23 expression on the early events of B-cell receptor (BCR) activation using CD23 knockout (KO) mice. We found that in addition to marginal zone B cells, mature follicular B cells significantly down regulate the surface expression level of CD23 after undergoing isotype switch and memory B-cell differentiation. Upon stimulation with membrane-associated antigen, CD23 KO causes significant increases in the area of B cells contacting the antigen-presenting membrane and the magnitude of BCR clustering. This enhanced cell spreading and BCR clustering is concurrent with increases in the levels of phosphorylation of tyrosine and Btk, as well as the levels of F-actin and phosphorylated Wiskott Aldrich syndrome protein, an actin nucleation promoting factor, in the contract zone of CD23 KO B cells. These results reveal a role of CD23 in the negative regulation of BCR signaling in the absence of IgE immune complex and suggest that CD23 down-regulates BCR signaling by influencing actin-mediated BCR clustering and B-cell morphological changes.


Subject(s)
B-Lymphocytes/immunology , Receptors, Antigen, B-Cell/immunology , Receptors, IgE/immunology , Signal Transduction/immunology , Actins/immunology , Actins/metabolism , Animals , B-Lymphocytes/metabolism , Immunoglobulin Class Switching/genetics , Immunoglobulin Class Switching/immunology , Immunologic Memory/genetics , Immunologic Memory/immunology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation/immunology , Receptors, Antigen, B-Cell/metabolism , Receptors, IgE/genetics , Receptors, IgE/metabolism , Signal Transduction/genetics , Wiskott-Aldrich Syndrome Protein/immunology , Wiskott-Aldrich Syndrome Protein/metabolism
13.
Asian Pac J Allergy Immunol ; 33(3): 189-95, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26342115

ABSTRACT

BACKGROUND: The engagement of the T cell receptor (TCR)-CD3 complex induces the formation of multiple signalling complexes, which are required for actin cytoskeletal rearrangement. The Wiskott-Aldrich syndrome protein (WASp) is a key regulator of actin polymerization that is recruited to the TCR activation site. Since WASp is a binding partner of adaptor protein Nck, which is recruited directly to the TCR CD3? subunit upon TCR ligation, therefore we proposed that the direct recruitment of Nck to TCR-CD3 may also bring WASp directly to TCR-CD3. OBJECTIVE: The aim of this present study was to assess the distribution of WASp, in relation to Nck, to the TCR-CD3ε complex. METHODS: Jurkat T cells were stimulated with anti-TCR antibody and then the cell lysates were immunoprecipitated with anti-CD3 antibody before immunoblotting with antibodies specific to WASp, Nck1, Nck2, SLP-76 and CD3ε molecules. RESULTS: WASp was recruited to SLP-76 and also directly to the TCR-CD3 complex upon TCR triggering. The inducible recruitment of WASp to the TCR-CD3 complex is partially dependent of tyrosine phosphorylation. CONCLUSIONS: The present findings provide an alternative mechanism of WASp recruitment to the site of TCR activation that may be involved in recruitment of Nck.


Subject(s)
CD3 Complex/metabolism , Receptor-CD3 Complex, Antigen, T-Cell/metabolism , T-Lymphocytes/metabolism , Wiskott-Aldrich Syndrome Protein/metabolism , Adaptor Proteins, Signal Transducing/metabolism , CD3 Complex/immunology , Humans , Jurkat Cells , Lymphocyte Activation , Oncogene Proteins/metabolism , Phosphoproteins/metabolism , Phosphorylation , Protein Binding , Protein Conformation , Protein Transport , Receptor-CD3 Complex, Antigen, T-Cell/immunology , Signal Transduction , T-Lymphocytes/immunology , Tyrosine , Wiskott-Aldrich Syndrome Protein/immunology
14.
Hum Immunol ; 76(6): 406-13, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25862925

ABSTRACT

A spontaneous reversion that restores Wiskott-Aldrich syndrome protein (WASP) expression was reported recently. However, the genetic mechanism underlying the reversion event was unclear. In the present study, a WAS patient with a nonsense mutation (155 C>T, R41X) was followed during a three-year period. No expression of WASP was detected in peripheral blood mononucleated cells (PBMCs) in 2009 and a small population of intracellular WASP positive lymphocytes was detected during the following three years. The increasing trend of the revertant genotype was significant. WASP-expressing cells were present mainly CD56+ NK cells and CD8+ T cells. Sorted WASP+ cells were analyzed, indicating that the population of CD3+ T cells increased from 36.81% to 99.8%. Although the revertant cells in vivo may have a growth advantage, the patient presented a persistent autoimmune disease, thrombocytopenia, and died from extensive pulmonary fibrosis. The results suggest that the clinical consequences of T-cell mosaicism in WAS remain difficult to predict and is not sufficient to fully normalize immune functions in patients with WAS.


Subject(s)
Pulmonary Fibrosis/genetics , Thrombocytopenia/genetics , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/genetics , Adolescent , Asian People , Base Sequence , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Fatal Outcome , Gene Expression , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/pathology , Male , Molecular Sequence Data , Mosaicism , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Thrombocytopenia/immunology , Thrombocytopenia/pathology , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/immunology
15.
Rev. bras. anestesiol ; 65(2): 130-135, Mar-Apr/2015. tab, graf
Article in English | LILACS | ID: lil-741712

ABSTRACT

BACKGROUND AND OBJECTIVE: To investigate the influence of intraoperative and preoperative positive pressure in the time of extubation in patients undergoing bariatric surgery. METHOD: Randomized clinical trial, in which 40 individuals with a body mass index between 40 and 55 kg/m2, age between 25 and 55 years, nonsmokers, underwent bariatric surgery type Roux-en-Y gastric bypass by laparotomy and with normal preoperative pulmonary function were randomized into the following groups: G-pre (n = 10): individuals who received treatment with noninvasive positive pressure before surgery for 1 h; G-intra (n = 10): individuals who received positive end-expiratory pressure of 10 cm H2O throughout the surgical procedure; and G-control (n = 20): not received any preoperative or intraoperative intervention. Following were recorded: time between induction of anesthesia and extubation, between the end of anesthesia and extubation, duration of mechanical ventilation, and time between extubation and discharge from the post-anesthetic recovery. RESULTS: There was no statistical difference between groups. However, when applied to the Cohen coefficient, the use of positive end-expiratory pressure of 10 cm H2O during surgery showed a large effect on the time between the end of anesthesia and extubation. About this same time, the treatment performed preoperatively showed moderate effect. CONCLUSION: The use of positive end-expiratory pressure of 10 cm H2O in the intraoperative and positive pressure preoperatively, influenced the time of extubation of patients undergoing bariatric surgery. .


JUSTIFICATIVA E OBJETIVO: investigar a influência do uso da pressão positiva nas vias aéreas intraoperatória e pré-operatória no tempo de extubação de pacientes submetidos à cirurgia bariátrica. MÉTODO: Trata-se de ensaio clínico randomizado, no qual 40 indivíduos com índice de massa corporal entre 40 e 55 kg/m2, idade entre 25 e 55 anos, não tabagistas, submetidos à cirurgia bariátrica do tipo derivação gástrica em Y de Roux por laparotomia e com prova de função pulmonar pré-operatória dentro da normalidade foram randomizados nos seguintes grupos: G-pré (n = 10): indivíduos que receberam tratamento com pressão positiva não invasiva antes da cirurgia, durante uma hora, G-intra (n = 10): indivíduos que receberam Positive End-expiratory Pressure de 10 cm H2O durante todo o procedimento cirúrgico e G-controle (n = 20): não receberam qualquer tipo de intervenção pré ou intraoperatória. foram anotados os seguintes tempos: tempo decorrido entre a indução anestésica e a extubação, entre o término da anestesia e extubação, tempo de ventilação mecânica, e tempo entre a extubação e a alta da Recuperação Pós-Anestésica. RESULTADOS: Não houve diferença estatística entre os grupos, porém quando aplicado ao Coeficiente de Cohen, o uso da Positive End-expiratory Pressure de 10 cm H2O no intraoperatório mostrou um efeito grande sobre o tempo entre o término da anestesia e a extubação. Sobre este mesmo tempo, o tratamento realizado no pré-operatório apresentou efeito moderado. CONCLUSÃO: O uso da Positive End-expiratory Pressure de 10 cm H2O no intraoperatório e da pressão positiva no pré-operatório, pode influenciar o tempo de extubação de pacientes submetidos à cirurgia bariátrica. .


JUSTIFICACIÓN Y OBJETIVO: Investigar la influencia del uso de la presión positiva en las vías aéreas intraoperatoria y preoperatoria en el tiempo de extubación de pacientes sometidos a la cirugía bariátrica. MÉTODO: Se trata de un ensayo clínico aleatorizado, en el cual 40 individuos con IMC entre 40 y 55 kg/m2, edad entre 25 y 55 años, no fumadores, sometidos a cirugía bariátrica del tipo derivación gástrica en Y de Roux por laparotomía y con prueba de función pulmonar preoperatoria dentro de la normalidad fueron aleatorizados en los siguientes grupos: G-pre (n = 10): individuos que recibieron tratamiento con presión positiva no invasiva antes de la cirugía durante una hora; G-intra (n = 10): individuos que recibieron PEEP de 10 cm H2O durante todo el procedimiento quirúrgico y G-control (n = 20): no recibieron ningún tipo de intervención pre- o intraoperatoria. Fueron anotados los siguientes tiempos: tiempo trascurrido entre la inducción anestésica y la extubación, entre el fin de la anestesia y la extubación, tiempo de ventilación mecánica, y tiempo entre la extubación y el alta de la sala de recuperación postanestésica. RESULTADOS: No hubo diferencia estadística entre los grupos, sin embargo cuando se aplicó el coeficiente de Cohen, el uso de la PEEP de 10 cm H2O en el intraoperatorio mostró un efecto importante sobre el tiempo entre el término de la anestesia y la extubación. Sobre ese mismo tiempo, el tratamiento realizado en el preoperatorio presentó un efecto moderado. CONCLUSIÓN: El uso de la PEEP de 10 cm H2O en el intraoperatorio y de la presión positiva en el preoperatorio puede influir en el tiempo de extubación de pacientes sometidos a cirugía bariátrica. .


Subject(s)
Animals , Female , Humans , Male , Mice , Arthritis, Experimental/immunology , B-Lymphocyte Subsets/immunology , Wiskott-Aldrich Syndrome Protein/immunology , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocyte Subsets/pathology , /genetics , /immunology , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , /immunology , /pathology , Wiskott-Aldrich Syndrome Protein/genetics
16.
Scand J Immunol ; 81(6): 483-93, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25728049

ABSTRACT

The Wiskott-Aldrich syndrome protein (WASp) is an important regulator of the actin cytoskeleton and is required for immune cell function. WASp deficiency causes a marked reduction in major mature peripheral B cell subsets, particularly marginal zone (MZ) B cells. We hypothesized that WASp deficiency may also lead to a reduction of regulatory B cells (known as B10 cells) belonging to a novel subset of B cells. And in consideration of the key role of B10 cells play in maintaining peripheral tolerance, we conjectured that a deficit of these cells could contribute to the autoimmunity in patients with Wiskott-Aldrich syndrome (WAS). The effects of WASp deficiency on B10 cells have been reported by only one group, which used an antigen-induced arthritis model. To add more information, we measured the percentage of B10 cells, regulatory T cells (Tregs) and Th1 cells in WASp knockout (WASp KO) mice. We also measured the percentage of B10 cells in patients with WAS by flow cytometry. Importantly, we used the non-induced autoimmune WASp KO mouse model to investigate the association between B10 cell frequency and the Treg/Th1 balance. We found that the percentage of B10 cells was reduced in both mice (steady state and inflammatory state) and in humans and that the lower B10 population correlated with an imbalance in the Treg/Th1 ratio in old WASp KO mice with autoimmune colitis. These findings suggest that WASp plays a crucial role in B10 cell development and that WASp-deficient B10 cells may contribute to autoimmunity in WAS.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Interleukin-10/immunology , Wiskott-Aldrich Syndrome Protein/immunology , Wiskott-Aldrich Syndrome/immunology , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , B-Lymphocytes, Regulatory/metabolism , Child , Child, Preschool , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Flow Cytometry , Humans , Infant , Interleukin-10/metabolism , Lymphocyte Count , Mice, Inbred C57BL , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/metabolism , Wiskott-Aldrich Syndrome Protein/deficiency , Wiskott-Aldrich Syndrome Protein/genetics
17.
J Allergy Clin Immunol ; 136(3): 692-702.e2, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25792466

ABSTRACT

BACKGROUND: Wiskott-Aldrich syndrome (WAS) is a severe X-linked immunodeficiency characterized by microthrombocytopenia, eczema, recurrent infections, and susceptibility to autoimmunity and lymphomas. Hematopoietic stem cell transplantation is the treatment of choice; however, administration of WAS gene-corrected autologous hematopoietic stem cells has been demonstrated as a feasible alternative therapeutic approach. OBJECTIVE: Because B-cell homeostasis is perturbed in patients with WAS and restoration of immune competence is one of the main therapeutic goals, we have evaluated reconstitution of the B-cell compartment in 4 patients who received autologous hematopoietic stem cells transduced with lentiviral vector after a reduced-intensity conditioning regimen combined with anti-CD20 administration. METHODS: We evaluated B-cell counts, B-cell subset distribution, B cell-activating factor and immunoglobulin levels, and autoantibody production before and after gene therapy (GT). WAS gene transfer in B cells was assessed by measuring vector copy numbers and expression of Wiskott-Aldrich syndrome protein. RESULTS: After lentiviral vector-mediated GT, the number of transduced B cells progressively increased in the peripheral blood of all patients. Lentiviral vector-transduced progenitor cells were able to repopulate the B-cell compartment with a normal distribution of B-cell subsets both in bone marrow and the periphery, showing a WAS protein expression profile similar to that of healthy donors. In addition, after GT, we observed a normalized frequency of autoimmune-associated CD19(+)CD21(-)CD35(-) and CD21(low) B cells and a reduction in B cell-activating factor levels. Immunoglobulin serum levels and autoantibody production improved in all treated patients. CONCLUSIONS: We provide evidence that lentiviral vector-mediated GT induces transgene expression in the B-cell compartment, resulting in ameliorated B-cell development and functionality and contributing to immunologic improvement in patients with WAS.


Subject(s)
B-Lymphocyte Subsets/metabolism , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/therapy , Autoantibodies/biosynthesis , B-Cell Activating Factor/genetics , B-Cell Activating Factor/metabolism , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/pathology , Bone Marrow/immunology , Bone Marrow/metabolism , Bone Marrow/pathology , Child , Child, Preschool , Gene Expression , Gene Expression Profiling , Genetic Vectors , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Immunoglobulins/biosynthesis , Immunophenotyping , Infant , Lentivirus/genetics , Male , Recombinant Fusion Proteins/therapeutic use , Transduction, Genetic , Transplantation Conditioning , Transplantation, Autologous , Wiskott-Aldrich Syndrome/genetics , Wiskott-Aldrich Syndrome/immunology , Wiskott-Aldrich Syndrome/pathology , Wiskott-Aldrich Syndrome Protein/immunology
18.
Biochem Biophys Res Commun ; 458(1): 28-33, 2015 Feb 27.
Article in English | MEDLINE | ID: mdl-25634698

ABSTRACT

Wiskott-Aldrich syndrome protein (WASP) is an adaptor molecule in immune cells. Recently, we showed that the WASP N-terminal domain interacted with the SH3 domain of Bruton's tyrosine kinase (Btk), and that the complex formed by WASP and Btk was important for TLR2 and TLR4 signaling in macrophages. Several other studies have shown that Btk played important roles in modulating innate immune responses through TLRs in immune cells. Here, we evaluated the significance of the interaction between WASP and Btk in TLR3, TLR7, and TLR9 signaling. We established bone marrow-derived macrophage cell lines from transgenic (Tg) mice that expressed intracellular antibodies (intrabodies) that specifically targeted the WASP N-terminal domain. One intrabody comprised the single-chain variable fragment and the other comprised the light-chain variable region single domain of an anti-WASP N-terminal monoclonal antibody. Both intrabodies inhibited the specific interaction between WASP and Btk, which impaired the expression of TNF-α, IL-6, and IL-1ß in response to TLR3, TLR7, or TLR9 stimulation. Furthermore, the intrabodies inhibited the phosphorylation of both nuclear factor (NF)-κB and WASP in response to TLR3, TLR7, or TLR9 stimulation, in the Tg bone marrow-derived macrophages. These results suggested that WASP plays important roles in TLR3, TLR7, and TLR9 signaling by associating with Btk in macrophages.


Subject(s)
Antibodies/immunology , Inflammation/drug therapy , Membrane Glycoproteins/metabolism , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 7/metabolism , Toll-Like Receptor 9/metabolism , Wiskott-Aldrich Syndrome Protein/immunology , Aminoquinolines/pharmacology , Animals , Antibodies/genetics , Antibodies/metabolism , Bone Marrow Cells/cytology , Cytokines/metabolism , Imiquimod , Inflammation/immunology , Inflammation/metabolism , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Membrane Glycoproteins/immunology , Mice, Inbred C57BL , Mice, Transgenic , NF-kappa B/metabolism , Phosphorylation/drug effects , Poly I-C/pharmacology , Single-Chain Antibodies/genetics , Single-Chain Antibodies/immunology , Single-Chain Antibodies/metabolism , Toll-Like Receptor 3/immunology , Toll-Like Receptor 7/immunology , Toll-Like Receptor 9/immunology , Wiskott-Aldrich Syndrome Protein/genetics
19.
Eur J Immunol ; 44(9): 2692-702, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24945741

ABSTRACT

Patients deficient in the cytoskeletal regulator Wiskott-Aldrich syndrome protein (WASp) are predisposed to varied autoimmunity, suggesting it has an important controlling role in participating cells. IL-10-producing regulatory B (Breg) cells are emerging as important mediators of immunosuppressive activity. In experimental, antigen-induced arthritis WASp-deficient (WASp knockout [WAS KO]) mice developed exacerbated disease associated with decreased Breg cells and regulatory T (Treg) cells, but increased Th17 cells in knee-draining LNs. Arthritic WAS KO mice showed increased serum levels of B-cell-activating factor, while their B cells were unresponsive in terms of B-cell-activating factor induced survival and IL-10 production. Adoptive transfer of WT Breg cells ameliorated arthritis in WAS KO recipients and restored a normal balance of Treg and Th17 cells. Mice with B-cell-restricted WASp deficiency, however, did not develop exacerbated arthritis, despite exhibiting reduced Breg- and Treg-cell numbers during active disease, and Th17 cells were not increased over equivalent WT levels. These findings support a contributory role for defective Breg cells in the development of WAS-related autoimmunity, but demonstrate that functional competence in other regulatory populations can be compensatory. A properly regulated cytoskeleton is therefore important for normal Breg-cell activity and complementation of defects in this lineage is likely to have important therapeutic benefits.


Subject(s)
Arthritis, Experimental/immunology , B-Lymphocyte Subsets/immunology , Wiskott-Aldrich Syndrome Protein/immunology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Autoimmune Diseases/pathology , B-Lymphocyte Subsets/pathology , Female , Humans , Interleukin-10/genetics , Interleukin-10/immunology , Male , Mice , Mice, Knockout , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Th17 Cells/immunology , Th17 Cells/pathology , Wiskott-Aldrich Syndrome Protein/genetics
20.
J Immunol ; 193(1): 150-60, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24872192

ABSTRACT

Defects in Wiskott-Aldrich Syndrome protein (WASp) underlie development of WAS, an X-linked immunodeficiency and autoimmunity disorder of childhood. Nucleation-promoting factors (NPFs) of the WASp family generate F-actin in the cytosol via the VCA (verprolin-homology, cofilin-homology, and acidic) domain and support RNA polymerase II-dependent transcription in the nucleus. Whether nuclear-WASp requires the integration of its actin-related protein (ARP)2/3-dependent cytoplasmic function to reprogram gene transcription, however, remains unresolved. Using the model of human TH cell differentiation, we find that WASp has a functional nuclear localizing and nuclear exit sequences, and accordingly, its effects on transcription are controlled mainly at the level of its nuclear entry and exit via the nuclear pore. Human WASp does not use its VCA-dependent, ARP2/3-driven, cytoplasmic effector mechanisms to support histone H3K4 methyltransferase activity in the nucleus of TH1-skewed cells. Accordingly, an isolated deficiency of nuclear-WASp is sufficient to impair the transcriptional reprogramming of TBX21 and IFNG promoters in TH1-skewed cells, whereas an isolated deficiency of cytosolic-WASp does not impair this process. In contrast, nuclear presence of WASp in TH2-skewed cells is small, and its loss does not impair transcriptional reprogramming of GATA3 and IL4 promoters. Our study unveils an ARP2/3:VCA-independent function of nuclear-WASp in TH1 gene activation that is uncoupled from its cytoplasmic role in actin polymerization.


Subject(s)
Actin-Related Protein 2-3 Complex/immunology , Actins/immunology , Cell Nucleus/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Transcription, Genetic/immunology , Wiskott-Aldrich Syndrome Protein/immunology , Actin-Related Protein 2-3 Complex/genetics , Actins/genetics , Cell Nucleus/genetics , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , HeLa Cells , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-4/genetics , Interleukin-4/immunology , Jurkat Cells , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Transcription, Genetic/genetics , Wiskott-Aldrich Syndrome Protein/genetics
SELECTION OF CITATIONS
SEARCH DETAIL