Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 140
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Clin Immunol ; 44(6): 143, 2024 Jun 07.
Article in English | MEDLINE | ID: mdl-38847936

ABSTRACT

Despite advancements in genetic and functional studies, the timely diagnosis of common variable immunodeficiency (CVID) remains a significant challenge. This exploratory study was designed to assess the diagnostic performance of a novel panel of biomarkers for CVID, incorporating the sum of κ+λ light chains, soluble B-cell maturation antigen (sBCMA) levels, switched memory B cells (smB) and the VISUAL score. Comparative analyses utilizing logistic regression were performed against established gold-standard tests, specifically antibody responses. Our research encompassed 88 subjects, comprising 27 CVID, 23 selective IgA deficiency (SIgAD), 20 secondary immunodeficiency (SID) patients and 18 healthy controls. We established the diagnostic accuracy of sBCMA and the sum κ+λ, achieving sensitivity (Se) and specificity (Spe) of 89% and 89%, and 90% and 99%, respectively. Importantly, sBCMA showed strong correlations with all evaluated biomarkers (sum κ+λ, smB cell and VISUAL), whereas the sum κ+λ was uniquely independent from smB cells or VISUAL, suggesting its additional diagnostic value. Through a multivariate tree decision model, specific antibody responses and the sum κ+λ emerged as independent, signature biomarkers for CVID, with the model showcasing an area under the curve (AUC) of 0.946, Se 0.85, and Spe 0.95. This tree-decision model promises to enhance diagnostic efficiency for CVID, underscoring the sum κ+λ as a superior CVID classifier and potential diagnostic criterion within the panel.


Subject(s)
Biomarkers , Common Variable Immunodeficiency , Humans , Common Variable Immunodeficiency/diagnosis , Common Variable Immunodeficiency/immunology , Male , Female , Adult , Middle Aged , Logistic Models , Young Adult , Adolescent , Aged , Immunoglobulin kappa-Chains/blood , Immunoglobulin kappa-Chains/genetics , Sensitivity and Specificity , B-Lymphocytes/immunology , Immunoglobulin lambda-Chains , Memory B Cells/immunology
2.
Exp Dermatol ; 33(8): e15146, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39075828

ABSTRACT

Patients with recessive dystrophic epidermolysis bullosa (RDEB) experience numerous complications, which are exacerbated by inflammatory dysregulation and infection. Understanding the immunological mechanisms is crucial for selecting medications that balance inflammation control and immunocompetence. In this cross-sectional study, aiming to identify potential immunotherapeutic targets and inflammatory biomarkers, we delved into the interrelationship between clinical severity and systemic inflammatory parameters in a representative RDEB cohort. Encompassing 84 patients aged 1-67 and spanning all three Epidermolysis Bullosa Disease Activity and Scarring Index (EBDASI) severity categories, we analysed the interrelationship of infection history, standard inflammatory markers, systemic cytokines and Ig levels to elucidate their roles in RDEB pathophysiology. Our findings identify C-reactive protein as an excellent biomarker for disease severity in RDEB. A type 2 inflammatory profile prevails among moderate and severe RDEB patients, correlating with dysregulated circulating IgA and IgG. These results underscore the IL4/IL13 pathways as potential evidence-based therapeutic targets. Moreover, the complete inflammatory scenario aligns with Staphylococcus aureus virulence mechanisms. Concurrently, abnormalities in IgG, IgE and IgM levels suggest an immunodeficiency state in a substantial number of the cohort's patients. Our results provide new insights into the interplay of infection and immunological factors in the pathogenesis of RDEB.


Subject(s)
Biomarkers , C-Reactive Protein , Epidermolysis Bullosa Dystrophica , Interleukin-13 , Interleukin-4 , Severity of Illness Index , Humans , Cross-Sectional Studies , Biomarkers/blood , Child , Child, Preschool , Interleukin-4/blood , Adolescent , C-Reactive Protein/metabolism , Adult , Young Adult , Female , Male , Infant , Middle Aged , Interleukin-13/blood , Interleukin-13/metabolism , Aged
3.
J Allergy Clin Immunol ; 152(4): 997-1009.e11, 2023 10.
Article in English | MEDLINE | ID: mdl-37301410

ABSTRACT

BACKGROUND: Inborn errors of immunity (IEI) are a group of monogenic diseases that confer susceptibility to infection, autoimmunity, and cancer. Despite the life-threatening consequences of some IEI, their genetic cause remains unknown in many patients. OBJECTIVE: We investigated a patient with an IEI of unknown genetic etiology. METHODS: Whole-exome sequencing identified a homozygous missense mutation of the gene encoding ezrin (EZR), substituting a threonine for an alanine at position 129. RESULTS: Ezrin is one of the subunits of the ezrin, radixin, and moesin (ERM) complex. The ERM complex links the plasma membrane to the cytoskeleton and is crucial for the assembly of an efficient immune response. The A129T mutation abolishes basal phosphorylation and decreases calcium signaling, leading to complete loss of function. Consistent with the pleiotropic function of ezrin in myriad immune cells, multidimensional immunophenotyping by mass and flow cytometry revealed that in addition to hypogammaglobulinemia, the patient had low frequencies of switched memory B cells, CD4+ and CD8+ T cells, MAIT, γδ T cells, and centralnaive CD4+ cells. CONCLUSIONS: Autosomal-recessive human ezrin deficiency is a newly recognized genetic cause of B-cell deficiency affecting cellular and humoral immunity.


Subject(s)
CD8-Positive T-Lymphocytes , Cytoskeleton , Humans , Cytoskeleton/metabolism , Cell Membrane/metabolism , Immunity, Humoral
4.
J Clin Immunol ; 44(1): 13, 2023 12 22.
Article in English | MEDLINE | ID: mdl-38129623

ABSTRACT

Human BCL10 deficiency causes combined immunodeficiency with bone marrow transplantation as its only curative option. To date, there are four homozygous mutations described in the literature that were identified in four unrelated patients. Here, we describe a fifth patient with a novel mutation and summarize what we have learned about BCL10 deficiency. Due to the severity of the disease, accurate knowledge of its clinical and immunological characteristics is instrumental for early diagnosis and adequate clinical management of the patients.


Subject(s)
Immunologic Deficiency Syndromes , Humans , B-Cell CLL-Lymphoma 10 Protein/genetics , Bone Marrow Transplantation , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/therapy , Mutation/genetics
5.
J Clin Immunol ; 42(5): 1093-1105, 2022 07.
Article in English | MEDLINE | ID: mdl-35486340

ABSTRACT

Common variable immunodeficiency disorders (CVID), the most common primary immune deficiency, includes heterogeneous syndromes characterized by hypogammaglobulinemia and impaired antibody responses. CVID patients frequently suffer from recurrent infections and inflammatory conditions. Currently, immunoglobulin replacement therapy (IgRT) is the first-line treatment to prevent infections and aminorate immune alterations in CVID patients. Intravenous Immunoglobulin (IVIg), a preparation of highly purified poly-specific IgG, is used for treatment of immunodeficiencies as well as for autoimmune and inflammatory disorders, as IVIg exerts immunoregulatory and anti-inflammatory actions on innate and adaptive immune cells. To determine the mechanism of action of IVIg in CVID in vivo, we determined the effect of IVIg infusion on the transcriptome of peripheral blood mononuclear cells from CVID patients, and found that peripheral blood monocytes are primary targets of IVIg in vivo, and that IVIg triggers the acquisition of an anti-inflammatory gene profile in human monocytes. Moreover, IVIg altered the relative proportions of peripheral blood monocyte subsets and enhanced the proportion of CD14+ cells with a transcriptional, phenotypic, and functional profile that resembles that of monocytic myeloid-derived suppressor cells (MDSC). Therefore, our results indicate that CD14 + MDSC-like cells might contribute to the immunoregulatory effects of IVIg in CVID and other inflammatory disorders.


Subject(s)
Common Variable Immunodeficiency , Myeloid-Derived Suppressor Cells , Common Variable Immunodeficiency/drug therapy , Humans , Immunoglobulins, Intravenous , Leukocytes, Mononuclear , Monocytes
6.
J Allergy Clin Immunol ; 147(2): 520-531, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32980424

ABSTRACT

BACKGROUND: There is uncertainty about the impact of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in individuals with rare inborn errors of immunity (IEI), a population at risk of developing severe coronavirus disease 2019. This is relevant not only for these patients but also for the general population, because studies of IEIs can unveil key requirements for host defense. OBJECTIVE: We sought to describe the presentation, manifestations, and outcome of SARS-CoV-2 infection in IEI to inform physicians and enhance understanding of host defense against SARS-CoV-2. METHODS: An invitation to participate in a retrospective study was distributed globally to scientific, medical, and patient societies involved in the care and advocacy for patients with IEI. RESULTS: We gathered information on 94 patients with IEI with SARS-CoV-2 infection. Their median age was 25 to 34 years. Fifty-three patients (56%) suffered from primary antibody deficiency, 9 (9.6%) had immune dysregulation syndrome, 6 (6.4%) a phagocyte defect, 7 (7.4%) an autoinflammatory disorder, 14 (15%) a combined immunodeficiency, 3 (3%) an innate immune defect, and 2 (2%) bone marrow failure. Ten were asymptomatic, 25 were treated as outpatients, 28 required admission without intensive care or ventilation, 13 required noninvasive ventilation or oxygen administration, 18 were admitted to intensive care units, 12 required invasive ventilation, and 3 required extracorporeal membrane oxygenation. Nine patients (7 adults and 2 children) died. CONCLUSIONS: This study demonstrates that (1) more than 30% of patients with IEI had mild coronavirus disease 2019 (COVID-19) and (2) risk factors predisposing to severe disease/mortality in the general population also seemed to affect patients with IEI, including more younger patients. Further studies will identify pathways that are associated with increased risk of severe disease and are nonredundant or redundant for protection against SARS-CoV-2.


Subject(s)
COVID-19/epidemiology , Genetic Diseases, Inborn/epidemiology , Immunologic Deficiency Syndromes/epidemiology , SARS-CoV-2 , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Retrospective Studies , Risk Factors , Severity of Illness Index , Young Adult
7.
J Clin Immunol ; 40(2): 388-398, 2020 02.
Article in English | MEDLINE | ID: mdl-32008135

ABSTRACT

In 2014, a child with broad combined immunodeficiency (CID) who was homozygous for a private BCL10 allele was reported to have complete inherited human BCL10 deficiency. In the present study, we report a new BCL10 mutation in another child with CID who was homozygous for a BCL10 variant (R88X), previously reported as a rare allele in heterozygosis (minor allele frequency, 0.000003986). The mutant allele was a loss-of-expression and loss-of-function allele. As with the previously reported patient, this patient had complete BCL10 deficiency. The clinical phenotype shared features, such as respiratory infections, but differed from that of the previous patient that he did not develop significant gastroenteritis episodes or chronic colitis. Cellular and immunological phenotypes were similar to those of the previous patient. TLR4, TLR2/6, and Dectin-1 responses were found to depend on BCL10 in fibroblasts, and final maturation of T cell and B cell maturation into memory cells was affected. Autosomal-recessive BCL10 deficiency should therefore be considered in children with CID.


Subject(s)
B-Cell CLL-Lymphoma 10 Protein/genetics , B-Lymphocytes/immunology , Immunologic Deficiency Syndromes/genetics , Mutation/genetics , T-Lymphocytes/immunology , Cells, Cultured , Chromosome Disorders , Homozygote , Humans , Immunologic Memory , Infant , Lectins, C-Type/metabolism , Male , Respiratory Tract Infections , Toll-Like Receptors/metabolism
8.
J Immunol ; 201(1): 41-52, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29743313

ABSTRACT

IVIg is an approved therapy for immunodeficiency and for several autoimmune and inflammatory diseases. However, the molecular basis for the IVIg anti-inflammatory activity remains to be fully explained and cannot be extrapolated from studies on animal models of disease. We now report that IVIg impairs the generation of human monocyte-derived anti-inflammatory macrophages by inducing JNK activation and activin A production and limits proinflammatory macrophage differentiation by inhibiting GM-CSF-driven STAT5 activation. In vivo, IVIg provokes a rapid increase in peripheral blood activin A, CCL2, and IL-6 levels, an effect that can be recapitulated in vitro on human monocytes. On differentiating monocytes, IVIg promotes the acquisition of altered transcriptional and cytokine profiles, reduces TLR expression and signaling, and upregulates negative regulators of TLR-initiated intracellular signaling. In line with these effects, in vivo IVIg infusion induces a state tolerant toward subsequent stimuli that results in reduced inflammatory cytokine production after LPS challenge in human peripheral blood and significant protection from LPS-induced death in mice. Therefore, IVIg conditions human macrophages toward the acquisition of a state of cross-tolerance against inflammatory stimuli, an effect that correlates with the net anti-inflammatory action of IVIg in vivo.


Subject(s)
Anti-Inflammatory Agents/immunology , Immune Tolerance/immunology , Immunoglobulins, Intravenous/immunology , Immunoglobulins, Intravenous/pharmacology , Macrophages/immunology , STAT5 Transcription Factor/metabolism , Activins/blood , Animals , Cells, Cultured , Chemokine CCL2/blood , Enzyme Activation , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Inflammation/immunology , Interleukin-6/blood , JNK Mitogen-Activated Protein Kinases/metabolism , Lipopolysaccharides/immunology , Mice , Monocytes/cytology , Monocytes/immunology
10.
Eur J Immunol ; 48(1): 180-193, 2018 01.
Article in English | MEDLINE | ID: mdl-28799230

ABSTRACT

Recurrent respiratory tract infections (RRTIs) are the first leading cause of community- and nosocomial-acquired infections. Antibiotics remain the mainstay of treatment, enhancing the potential to develop antibiotic resistances. Therefore, the development of new alternative approaches to prevent and treat RRTIs is highly demanded. Daily sublingual administration of the whole heat-inactivated polybacterial preparation (PBP) MV130 significantly reduced the rate of respiratory infections in RRTIs patients, however, the immunological mechanisms of action remain unknown. Herein, we study the capacity of MV130 to immunomodulate the function of human dendritic cells (DCs) as a potential mechanism that contribute to the clinical benefits. We demonstrate that DCs from RRTIs patients and healthy controls display similar ex vivo immunological responses to MV130. By combining systems biology and functional immunological approaches we show that MV130 promotes the generation of Th1/Th17 responses via receptor-interacting serine/threonine-protein kinase-2 (RIPK2)- and myeloid-differentiation primary-response gene-88 (MyD88)-mediated signalling pathways under the control of IL-10. In vivo BALB/c mice sublingually immunized with MV130 display potent systemic Th1/Th17 and IL-10 responses against related and unrelated antigens. We elucidate immunological mechanisms underlying the potential way of action of MV130, which might help to design alternative treatments in other clinical conditions with high risk of recurrent infections.


Subject(s)
Bacterial Vaccines/immunology , Dendritic Cells/immunology , Interleukin-10/immunology , Myeloid Differentiation Factor 88/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Respiratory Tract Infections/prevention & control , Th1 Cells/immunology , Th17 Cells/immunology , Adult , Aged , Animals , Cells, Cultured , Female , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Respiratory Tract Infections/immunology , Respiratory Tract Infections/microbiology , Signal Transduction/immunology
11.
Eur J Haematol ; 102(6): 447-456, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30801785

ABSTRACT

OBJECTIVE: Despite long-standing safe and effective use of immunoglobulin replacement therapy (IgRT) in primary immunodeficiency, clinical data on IgRT in patients with secondary immunodeficiency (SID) due to B-cell lymphoproliferative diseases are limited. Here, we examine the correlation between approved IgRT indications, treatment recommendations, and clinical practice in SID. METHODS: An international online survey of 230 physicians responsible for the diagnosis of SID and the prescription of IgRT in patients with hematological malignancies was conducted. RESULTS: Serum immunoglobulin was measured in 83% of patients with multiple myeloma, 76% with chronic lymphocytic leukemia, and 69% with non-Hodgkin lymphoma. Most physicians (85%) prescribed IgRT after ≥2 severe infections. In Italy, Germany, Spain, and the United States, immunoglobulin use was above average in patients with hypogammaglobulinemia, while in the UK considerably fewer patients received IgRT. The use of subcutaneous immunoglobulin was highest in France (34%) and lowest in Spain (19%). Immunologists measured specific antibody responses, performed test immunization, implemented IgRT, and used subcutaneous immunoglobulin more frequently than physicians overall. CONCLUSIONS: The management of SID in hematological malignancies varied regionally. Clinical practice did not reflect treatment guidelines, highlighting the need for robust clinical studies on IgRT in this population and harmonization between countries and disciplines.


Subject(s)
Hematologic Neoplasms/complications , Hematologic Neoplasms/epidemiology , Immunologic Deficiency Syndromes/epidemiology , Immunologic Deficiency Syndromes/etiology , Disease Management , Global Health , Humans , Immunoglobulins/blood , Immunoglobulins/immunology , Immunoglobulins, Intravenous/therapeutic use , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/therapy , Infection Control , Infections/etiology , Public Health Surveillance , Treatment Outcome
12.
Int J Cancer ; 142(1): 133-144, 2018 01 01.
Article in English | MEDLINE | ID: mdl-28884480

ABSTRACT

Human blood monocytes are very potent to take up antigens. Like macrophages in tissue, they efficiently degrade exogenous protein and are less efficient than dendritic cells (DCs) at cross-presenting antigens to CD8+ T cells. Although it is generally accepted that DCs take up tissue antigens and then migrate to lymph nodes to prime T cells, the mechanisms of presentation of antigens taken up by monocytes are poorly documented so far. In the present work, we show that monocytes loaded in vitro with MelanA long peptides retain the capacity to stimulate antigen-specific CD8+ T cell clones after 5 days of differentiation into monocytes-derived dendritic cells (MoDCs). Tagged-long peptides can be visualized in electron-dense endocytic compartments distinct from lysosomes, suggesting that antigens can be protected from degradation for extended periods of time. To address the pathophysiological relevance of these findings, we screened blood monocytes from 18 metastatic melanoma patients and found that CD14+ monocytes from two patients effectively activate a MelanA-specific CD8 T cell clone after in vitro differentiation into MoDCs. This in vivo sampling of tumor antigen by circulating monocytes might alter the tumor-specific immune response and should be taken into account for cancer immunotherapy.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , MART-1 Antigen/immunology , Melanoma/immunology , Monocytes/immunology , Antigen Presentation/immunology , Cell Differentiation/immunology , Dendritic Cells/cytology , Humans , Monocytes/cytology
13.
Respir Res ; 19(1): 219, 2018 11 12.
Article in English | MEDLINE | ID: mdl-30419907

ABSTRACT

BACKGROUND: Pulmonary complications are common in primary immunodeficiency diseases (PID) and contribute to morbidity and mortality in these patients. However, their varied presentation and a general lack of awareness of PID in this setting make early diagnosis and treatment difficult. The aim of this study was to define the warning signs of PID in patients with respiratory manifestations, the necessary diagnostic tests, and the therapeutic management of both children and adults. METHODS: A review of the literature was performed, and 43 PID interdisciplinary specialists were consulted. RESULTS: This document identifies the pulmonary and extrapulmonary manifestations that should prompt a suspicion of PID, the immunological and respiratory tests that should be included in the diagnostic process according to the level of care, recommendations regarding the use of immunoglobulin replacement therapy according to the specific immunodeficiency, and the minimum recommended immunological and pulmonary monitoring in these patients. CONCLUSIONS: This document is the first to combine scientific evidence with the opinion of a broad panel of experts specializing in the treatment of patients with immunodeficiencies. It aims to provide a useful tool for all practitioners who are regularly involved in the management of these patients.


Subject(s)
Disease Management , Immunologic Deficiency Syndromes/diagnosis , Immunologic Deficiency Syndromes/therapy , Lung Diseases/diagnosis , Lung Diseases/therapy , Expert Testimony/methods , Expert Testimony/trends , Humans , Immunologic Deficiency Syndromes/epidemiology , Lung Diseases/epidemiology
15.
Clin Immunol ; 181: 43-50, 2017 08.
Article in English | MEDLINE | ID: mdl-28578025

ABSTRACT

Delay in the diagnosis of multiple sclerosis (MS) stems from the lack of specific clinical and analytical markers to assist in the early diagnosis and prediction of progressive course. We propose a decision-tree model that better defines early at onset MS patients and those with the progressive form by analysing a 12-biomarkers panel in serum and CSF samples of patients with MS, other neurological diseases (OND) and healthy contols. Thus, patients at onset of neurological disease were first classified by serum IL-7 levels <141pg/ml (OR=6.51, p<0.001). Combination of IL-7 and CXCL10 indicated risk for a specific MS clinical form, where IL-7<141 and CXCL10<570pg/ml were associated with the highest risk for PP-MS (OR=22, p=0.01). Unexpectedly, both PP-MS and RR-MS patients shared significantly decreased prototypical biomarkers of inflammation and tissue regeneration in CSF than OND suggesting a defective intrinsic immune response playing a role at the beginning of the disease.


Subject(s)
Multiple Sclerosis, Chronic Progressive/diagnosis , Multiple Sclerosis, Relapsing-Remitting/diagnosis , Area Under Curve , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Case-Control Studies , Chemokine CCL11 , Chemokine CCL2 , Chemokine CCL4 , Chemokine CCL5 , Chemokine CXCL10/blood , Chemokine CXCL10/cerebrospinal fluid , Chemokine CXCL9/blood , Chemokine CXCL9/cerebrospinal fluid , Decision Trees , Dipeptidyl Peptidase 4/blood , Dipeptidyl Peptidase 4/cerebrospinal fluid , Early Diagnosis , Epidermal Growth Factor , Fibroblast Growth Factor 2/blood , Fibroblast Growth Factor 2/cerebrospinal fluid , Hepatocyte Growth Factor , Humans , Interleukin 1 Receptor Antagonist Protein/blood , Interleukin 1 Receptor Antagonist Protein/cerebrospinal fluid , Interleukin-7/blood , Interleukin-7/cerebrospinal fluid , Multiple Sclerosis, Chronic Progressive/blood , Multiple Sclerosis, Chronic Progressive/cerebrospinal fluid , Multiple Sclerosis, Relapsing-Remitting/blood , Multiple Sclerosis, Relapsing-Remitting/cerebrospinal fluid , Multivariate Analysis , Nervous System Diseases/blood , Nervous System Diseases/cerebrospinal fluid , Nervous System Diseases/diagnosis , Prognosis , Risk Assessment
17.
Int J Mol Sci ; 18(6)2017 Jun 21.
Article in English | MEDLINE | ID: mdl-28635659

ABSTRACT

Asthma is an airway disease characterised by chronic inflammation with intermittent or permanent symptoms including wheezing, shortness of breath, chest tightness, and cough, which vary in terms of their occurrence, frequency, and intensity. The most common associated feature in the airways of patients with asthma is airway inflammation. In recent decades, efforts have been made to characterise the heterogeneous clinical nature of asthma. The interest in improving the definitions of asthma phenotypes and endotypes is growing, although these classifications do not always correlate with prognosis nor are always appropriate therapeutic approaches. Attempts have been made to identify the most relevant molecular and cellular biomarkers underlying the immunopathophysiological mechanisms of the disease. For almost 50 years, immunoglobulin E (IgE) has been identified as a central factor in allergic asthma, due to its allergen-specific nature. Many of the mechanisms of the inflammatory cascade underlying allergic asthma have already been elucidated, and IgE has been shown to play a fundamental role in the triggering, development, and chronicity of the inflammatory responses within the disease. Blocking IgE with monoclonal antibodies such as omalizumab have demonstrated their efficacy, effectiveness, and safety in treating allergic asthma. A better understanding of the multiple contributions of IgE to the inflammatory continuum of asthma could contribute to the development of novel therapeutic strategies for the disease.


Subject(s)
Anti-Asthmatic Agents/therapeutic use , Antibodies, Monoclonal/therapeutic use , Asthma/drug therapy , Immunoglobulin E/immunology , Omalizumab/therapeutic use , Animals , Asthma/immunology , Asthma/physiopathology , Humans , Inflammation/drug therapy , Inflammation/immunology , Inflammation/physiopathology
19.
Clin Immunol ; 173: 117-120, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27693481

ABSTRACT

Antibody deficiencies can be caused by a variety of defects that interfere with B-cell development, maturation, and/or function. Using whole-exome sequencing we found a PIK3R1 mutation in a patient with hypogammaglobulinemia and a narrow clinical phenotype of respiratory infections. Early diagnosis is crucial; careful analysis of B and T-cells followed by genetic analyses may help to distinguish activated PI3K-delta syndrome (APDS) from other, less severe, predominantly antibody deficiencies.


Subject(s)
Agammaglobulinemia/genetics , Phosphatidylinositol 3-Kinases/genetics , Respiratory Tract Infections/genetics , Agammaglobulinemia/immunology , B-Lymphocytes/immunology , Child , Class Ia Phosphatidylinositol 3-Kinase , Female , Humans , Mutation , Phenotype , Respiratory Tract Infections/immunology , T-Lymphocytes/immunology
SELECTION OF CITATIONS
SEARCH DETAIL