Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 12 de 12
1.
Front Immunol ; 12: 649112, 2021.
Article En | MEDLINE | ID: mdl-33981304

Selective IgA deficiency (SIgAD), characterized by a serum IgA level below 0.07 mg/ml, while displaying normal serum levels of IgM and IgG antibodies, is the most frequently occurring primary immunodeficiency that reveals itself after the first four years after birth. These individuals with SIgAD are for the majority healthy and even when they are identified they are usually not investigated further or followed up. However, recent studies show that newborns and young infants already display clinical manifestations of this condition due to aberrancies in their immune defense. Interestingly, there is a huge heterogeneity in the clinical symptoms of the affected individuals. More than 50% of the affected individuals do not have clinical symptoms, while the individuals that do show clinical symptoms can suffer from mild to severe infections, allergies and autoimmune diseases. However, the reason for this heterogeneity in the manifestation of clinical symptoms of the individuals with SIgAD is unknown. Therefore, this review focusses on the characteristics of innate immune system driving T-cell independent IgA production and providing a mechanism underlying the development of SIgAD. Thereby, we focus on some important genes, including TNFRSF13B (encoding TACI), associated with SIgAD and the involvement of epigenetics, which will cover the methylation degree of TNFRSF13B, and environmental factors, including the gut microbiota, in the development of SIgAD. Currently, no specific treatment for SIgAD exists and novel therapeutic strategies could be developed based on the discussed information.


IgA Deficiency/immunology , Immunity, Innate/immunology , Immunoglobulin A/immunology , Transmembrane Activator and CAML Interactor Protein/immunology , Epigenesis, Genetic/genetics , Epigenesis, Genetic/immunology , Gastrointestinal Microbiome/immunology , Humans , IgA Deficiency/genetics , IgA Deficiency/metabolism , Immunity, Innate/genetics , Immunoglobulin A/blood , Infant , Infant, Newborn , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transmembrane Activator and CAML Interactor Protein/genetics , Transmembrane Activator and CAML Interactor Protein/metabolism
2.
Front Immunol ; 12: 634544, 2021.
Article En | MEDLINE | ID: mdl-33679786

B cell differentiation and memory are controlled by the transmembrane activator and CAML interactor (TACI), a receptor encoded by TNFRSF13B. TNFRSF13B mutations are frequently found in common variable immunodeficiency (CVID) and in IgA -deficiency; yet, ~98% of those with mutant TNFRSF13B are healthy. Indeed, TNFRSF13B is among the 5% most polymorphic genes in man. Other mammals evidence polymorphism at comparable loci. We hypothesize that TNFRSF13B diversity might promote rather than detract from well-being by controlling key elements of innate immunity. We shall discuss how extraordinary diversity of TNFRSF13B could have evolved and persisted across diverse species of mammals by controlling innate and adaptive B cell responses in apparently paradoxical ways.


Adaptive Immunity/genetics , B-Lymphocytes/immunology , Common Variable Immunodeficiency/genetics , Evolution, Molecular , IgA Deficiency/genetics , Immunity, Innate/genetics , Mutation , Transmembrane Activator and CAML Interactor Protein/genetics , Animals , Antibodies/metabolism , B-Lymphocytes/metabolism , Common Variable Immunodeficiency/immunology , Common Variable Immunodeficiency/metabolism , Genetic Predisposition to Disease , Humans , IgA Deficiency/immunology , IgA Deficiency/metabolism , Phenotype , Transmembrane Activator and CAML Interactor Protein/metabolism
3.
Front Immunol ; 10: 403, 2019.
Article En | MEDLINE | ID: mdl-30936864

Objectives: It has recently been shown that individuals with selective IgA deficiency (sIgAD) have defective B cell responses both to T cell dependent and independent mimicking stimulations. The complex intracellular signaling pathways from different stimuli leading to IgA isotype switching have not been fully elucidated. Thus, the main objective of this study was to delineate these pathways and their potential role in the immunopathology linked to sIgAD. Materials and Methods: PBMCs from 10 individuals with sIgAD and 10 healthy controls (HC) were activated in vitro via either a T cell dependent or independent mimicking stimulation. Intracellular phosphorylation of pSTAT3, pSTAT5, pSTAT6, and as pERK1/2 was evaluated in T and B cells using phosphoflow cytometry. Results: By evaluating T cell dependent cytokine driven pathways linked to IgA isotype induction we identified a defect involving an IL-21 driven STAT3 activation isolated to B cells in sIgAD individuals. However, all other signaling pathways studied were found to be normal compared to HC. In T cell dependent cytokine driven stimulations linked to IgA isotype induction the following patterns emerged: (i) IL-10 led to significant STAT3 activation in both T- and B cells; (ii) IL-4 stimulation was predominantly confined to STAT6 activation in both T- and B cells, with some effects on STAT3 activation in T-cells; (iii) as expected, of tested stimuli, IL-2 alone activated STAT5 and some STAT3 activation though in both cases only in T-cells; (iv) IL-21 induced significant activation of STAT3 in both T- and B cells, with some effects on STAT5 activation in T-cells; and finally (v) synergistic effects were noted of IL-4+IL-10 on STAT5 activation in T-cells, and possibly STAT6 in both T- and B cells. On the other hand, CPG induced T cell independent activation was confined to ERK1/2 activation in B cells. Conclusion: Our results indicate a diminished STAT3 phosphorylation following IL-21 stimulation solely in B cells from sIgAD individuals. This can represent aberrant germinal center reactions or developmental halt. Thus, our work provides further insight into the unraveling of the previously hypothesized role of IL-21 to reconstitute immunoglobulin production in primary antibody deficiencies.


B-Lymphocytes/immunology , IgA Deficiency/immunology , Interleukins/immunology , Lymphocyte Activation/immunology , STAT3 Transcription Factor/immunology , B-Lymphocytes/metabolism , Humans , IgA Deficiency/metabolism , Phosphorylation , STAT3 Transcription Factor/metabolism , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
4.
Int Arch Allergy Immunol ; 169(2): 130-4, 2016.
Article En | MEDLINE | ID: mdl-27073906

BACKGROUND: Immunoglobulin (Ig) A deficiency is a primary immunodeficiency in which autoimmunity is frequently observed. Thirty to fifty percent of patients with spontaneous chronic urticaria have autoantibodies that are able to cross-link FcεRI on mast cells and basophils. METHODS: We investigated whether spontaneous chronic urticaria in patients with IgA deficiency meets the criteria for autoimmunity. Four patients were screened for positivity to a skin prick test and an autologous serum skin test and for the presence of other autoimmune diseases. Patient sera were tested for the ability to activate basophils and mast cells in vitro by measuring surface CD63 expression and ß-hexosaminidase release, respectively. RESULTS: The autologous serum test was positive in all patients, and patient sera were found to induce CD63 upregulation on basophils and degranulation of an LAD2 mast cell line. Moreover, all patients were affected by other autoimmune disorders. CONCLUSION: For the first time, these data point out chronic autoimmune urticaria in subjects with an IgA deficiency and confirm that different autoimmune disorders are common among patients with an IgA deficiency. Patients with chronic autoimmune spontaneous urticaria should be screened for IgA deficiency, especially if they are affected by other autoimmune disorders. Thus, spontaneous urticaria could mirror more complex systemic diseases, such as immune deficiency.


Autoantibodies/immunology , Autoimmune Diseases/complications , Autoimmune Diseases/immunology , IgA Deficiency/complications , IgA Deficiency/immunology , Urticaria/complications , Urticaria/immunology , Adolescent , Adult , Autoimmune Diseases/metabolism , Basophils/immunology , Basophils/metabolism , Cell Degranulation/immunology , Female , Humans , IgA Deficiency/metabolism , Immunoglobulin E/blood , Immunoglobulin E/immunology , Male , Mast Cells/immunology , Mast Cells/metabolism , Middle Aged , Receptors, IgE/metabolism , Urticaria/metabolism , Young Adult
5.
J Cutan Pathol ; 40(12): 1035-41, 2013 Dec.
Article En | MEDLINE | ID: mdl-24274426

Multiple lymphoma subtypes occurring within one patient is rare in the context of B-cell lymphoma, and only few such cases have been reported in association with primary cutaneous marginal zone lymphoma (PCMZL). We herein describe the case of a 43-year-old patient who was diagnosed with PCMZL and subsequently developed a clonally unrelated nodal marginal zone lymphoma (MZL). At the time of diagnosis of PCMZL, multiple skin lesions were present. The atypical lymphoid infiltrate showed monotypic expression of immunoglobulin light chain lambda and heavy chain (IgM) on immunohistochemistry and an identical B-cell clone. No sign of systemic lymphoma was present in staging examinations. Complete remission was achieved utilizing rituximab. After a 3-year clinical course of repetitive cutaneous relapses and remissions, the patient additionally developed nodal lymphoma involvement by MZL which, however, harbored an immunophenotype and a genetic clone distinct from the cutaneous lymphoma counterpart. Therefore, the rare occurrence of two different types of MZL with sequential evolution was diagnosed. In this uncommon case, we hypothesize that selective immunoglobulin A deficiency may play a promoting role for the metachronous development of the two MZL that occurred in our patient.


IgA Deficiency/pathology , Lymphoma, B-Cell, Marginal Zone/pathology , Skin Neoplasms/pathology , Adult , Female , Humans , IgA Deficiency/complications , IgA Deficiency/metabolism , IgA Deficiency/therapy , Lymphoma, B-Cell, Marginal Zone/metabolism , Lymphoma, B-Cell, Marginal Zone/therapy , Skin Neoplasms/metabolism , Skin Neoplasms/therapy
6.
Ann Clin Biochem ; 44(Pt 2): 131-9, 2007 Mar.
Article En | MEDLINE | ID: mdl-17362578

IgA deficiency is the most common primary immunoglobulin deficiency. The prevalence in Caucasians is around one in 500, whereas in some Asian populations it is very uncommon. Most individuals with IgA deficiency are clinically asymptomatic. Those with symptoms of immunodeficiency have predominantly sinopulmonary or gastrointestinal infections, which are more severe when associated with IgG2, IgG4 or specific antibody deficiency. IgA deficiency is believed to be one end of a spectrum of immunodeficiency with common variable immunodeficiency at the most severe end. Although primary IgA deficiency is the most commonly encountered form, secondary deficiencies due to drugs or viral infections are recognized. IgA deficiencies can be partial or transient. Primary IgA deficiency is caused by a defect of terminal lymphocyte differentiation, which leads to underproduction of serum and mucosal IgA; affected individuals have normal IgA genes. A number of non-immunoglobulin genes have been implicated in IgA deficiency. There have been many diseases reported in association with IgA deficiency, particularly autoimmune diseases. The most common association is with coeliac disease (CD), which has special significance since CD is usually diagnosed by detection of specific IgA antibodies that are obviously lacking in IgA deficiency. There is no specific treatment for patients with symptomatic IgA deficiency. Antibiotics are prescribed in those with acute infections. A significant proportion of IgA-deficient individuals are reported to have anti-IgA antibodies in their serum. Although blood or blood products given to IgA-deficient individuals can lead to severe, even fatal, transfusion reactions, such reactions are rare.


IgA Deficiency/pathology , Immunoglobulin A/immunology , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Celiac Disease/complications , Celiac Disease/immunology , Celiac Disease/metabolism , Humans , Hypersensitivity/immunology , Hypersensitivity/metabolism , IgA Deficiency/complications , IgA Deficiency/metabolism
7.
Pathol Int ; 57(1): 37-42, 2007 Jan.
Article En | MEDLINE | ID: mdl-17199741

A combined chromosomal abberation trisomy of the short arm of chromosome 10 associated with translocation of 10q to chromosome 4p was found in a 14-month-old boy, who died after repeated bouts of pneumonia. The translocation involved the target region 4p16.3 of Wolf-Hirschhorn syndrome and/or Pitt-Rogers-Danks syndrome. The karyotype was 46,XY,der(4)t(4;10)(p16;q11.2),i(10)(p10),ish der(4)t(4;10)(p16.3;q11.2) (D4S96+,D4Z1+),i(10) (pter ++). In addition to growth retardation and external as well as internal dysmorphism, the patient had abnormalities of the immune system, such as thymic involution, generalized lymph node enlargement, unusual distribution of T cells in lymphoid follicles, and selective IgA deficiency. The IgA-producing cells were rarely found in lymph nodes but normally in intestinal mucosa. In contrast, in the lymph nodes, the paracortical T-lymphocytes were hyperplastic, but they rarely entered the primary follicles. It is assumed that the chromosomal abnormality may lead to the dysfunction of T lymphocytes and, further, to the dysgenesis of IgA-producing cells in lymph nodes but not in intestinal mucosa. This suggests that the thymus may differentially control the subsets of IgA-producing cells in lymph nodes and intestinal mucosa.


Chromosomes, Human, Pair 10/genetics , Immunoglobulin A/metabolism , Lymphoid Tissue/pathology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology , Translocation, Genetic/genetics , Trisomy/genetics , Autopsy , Humans , IgA Deficiency/diagnosis , IgA Deficiency/metabolism , IgA Deficiency/pathology , Infant , Intestinal Mucosa/immunology , Intestinal Mucosa/pathology , Lymphoid Tissue/metabolism , Male , Phenotype , Trisomy/diagnosis
8.
Acta Otorhinolaryngol Belg ; 54(3): 373-90, 2000.
Article En | MEDLINE | ID: mdl-11082774

Adequate surface protection of the upper airway tract depends on intimate co-operation between natural non specific defence mechanisms such as ciliary function and acquired adaptative immunity. The latter is mediated by specific antibodies mainly belonging to secretory immunoglobulin A (SIgA) and to lesser extent secretory IgM (SigM) as well as by serum derived and locally produced IgG. Immunoglobulin's deficiency may exist in a significant percentage of patients with chronic or recurrent infections of the upper respiratory tract. So an immunologic screening should be extended to patients who have persistent sinus infection, despite normal mucocilary and ventilation patterns, inadequate response to antimicrobial therapy, culture of unusual pathogens from the upper respiratory tract or history of infection at other side, mainly bronchopulmonary. The therapeutic aspects include immunoglobulin replacement or mucosal immunogens whose efficacy is related with both an unspecific and a specific way. In the future, cytokines will probably become the strongest therapy of the immunoglobulin's deficiency.


IgA Deficiency/immunology , IgG Deficiency/immunology , Immunoglobulin M/immunology , Immunoglobulins, Intravenous/therapeutic use , Respiratory System/immunology , Respiratory Tract Infections , Agammaglobulinemia/genetics , Agammaglobulinemia/immunology , Albumins/metabolism , Antibody Formation/immunology , Child , Child, Preschool , Chronic Disease , Cilia/physiology , Ciliary Motility Disorders/diagnosis , Ciliary Motility Disorders/physiopathology , Humans , IgA Deficiency/metabolism , IgG Deficiency/metabolism , Immunoglobulin M/deficiency , Immunoglobulin M/metabolism , Infant , Nasal Mucosa/immunology , Nasal Mucosa/metabolism , Recurrence , Respiratory Mucosa/physiology , Respiratory Tract Infections/diagnosis , Respiratory Tract Infections/drug therapy , Respiratory Tract Infections/immunology , X Chromosome/genetics
9.
J Immunol ; 163(10): 5367-73, 1999 Nov 15.
Article En | MEDLINE | ID: mdl-10553061

We generated mouse lacking exon 2 of polymeric Ig receptor (pIgR) gene by a gene-targeting strategy (pIgR-deficient mouse; pIgR-/- mouse) to define the physiological role of pIgR in the transcytosis of Igs. pIgR-/- mice were born at the expected ratio from a cross between pIgR+/- mice, indicating that disruption of the pIgR gene in mice is not lethal. pIgR and secretory component proteins were not detected in pIgR-/- mice by Western blot analysis. Moreover, immunohistochemical analysis showed that pIgR protein is not expressed in jejunal and colonic epithelial cells of pIgR-/- mice, whereas IgA+ cells are present in the intestinal mucosa of pIgR-/- mice as well as wild-type littermates. Disruption of the pIgR gene caused a remarkable increase in serum IgA concentration and a slight increment of serum IgG and IgE levels, leaving serum IgM level unaltered. In contrast, IgA was much reduced but not negligible in the bile, feces, and intestinal contents of pIgR-/- mice. Additionally, IgA with a molecular mass of 280 kDa preferentially accumulated in the serum of pIgR-/- mice, suggesting that transepithelial transport of dIgA is severely blocked in pIgR-/- mice. These results demonstrate that dIgA is mainly transported by pIgR on the epithelial cells of intestine and hepatocytes, but a small quantity of IgA may be secreted via other pathways.


IgA Deficiency/genetics , Immunoglobulin A, Secretory/metabolism , Receptors, Polymeric Immunoglobulin/deficiency , Receptors, Polymeric Immunoglobulin/genetics , Animals , Bile/chemistry , Dimerization , Exons/genetics , Genes, Immunoglobulin/genetics , Genetic Vectors/chemical synthesis , IgA Deficiency/blood , IgA Deficiency/metabolism , Immunoglobulin A/blood , Immunoglobulin A/metabolism , Immunoglobulin A, Secretory/analysis , Intestinal Secretions/chemistry , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Recombination, Genetic , Secretory Component/analysis , Secretory Component/metabolism , Sequence Deletion
10.
J Allergy Clin Immunol ; 100(4): 556-62, 1997 Oct.
Article En | MEDLINE | ID: mdl-9338552

BACKGROUND: Transient hypogammaglobulinemia of infancy and isolated IgA deficiency are characterized by normal numbers of circulating B lymphocytes. It is likely that no single abnormality, but rather different factors, may be relevant for the delayed onset of IgG synthesis in transient hypogammaglobulinemia or for the differentiation defect of B cells in IgA deficiency. These factors may include defective production of cytokines or an abnormal response of B cells to various mediators. Alternatively, some cytokines may act as inhibitory factors of B-cell function. METHODS: The ability of peripheral blood mononuclear cells from children with proved or probable transient hypogammaglobulinemia (30 patients) and IgA deficiency (15 patients) to secrete several cytokines on stimulation with phytohemagglutinin in vitro was analyzed. RESULTS: An enhanced production of tumor necrosis factor (TNF)-alpha, TNF-beta, and IL-10 was observed in transient hypogammaglobulinemia; whereas secretion of IL-1, IL-4, and IL-6 was essentially similar in the control and patient groups. Increased frequency of mononuclear cells secreting TNF-alpha was seen in the patient groups. Apart from elevated production of TNF-alpha, no other abnormalities in cytokine synthesis in selective IgA deficiency were observed. In vitro observations showed that exogenously added TNF-alpha and TNF-beta inhibited IgG and IgA secretion by pokeweed mitogen-stimulated mononuclear cells. During follow-up of 10 children, normalization of serum IgG level was associated with a decrease in previously elevated TNF-alpha and TNF-beta production, but IL-10 production remained unchanged. CONCLUSION: These results suggest that TNF may be involved in the regulation of IgG and IgA production and can be associated with an arrest of IgG and IgA switch of B cells in hypogammaglobulinemia. The balance between TNF and IL-10 may be important for the normal development of IgG-secreting B cells.


Agammaglobulinemia/metabolism , Cytokines/biosynthesis , IgA Deficiency/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cells, Cultured , Child , Child, Preschool , Female , Humans , Immunoglobulin A/biosynthesis , Immunoglobulin A/blood , Immunoglobulin G/biosynthesis , Immunoglobulin G/blood , Infant , Interleukin-10/biosynthesis , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , Longitudinal Studies , Male , Phytohemagglutinins/pharmacology , Pokeweed Mitogens/pharmacology , Stimulation, Chemical , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/pharmacology
11.
Infect Immun ; 64(7): 2794-8, 1996 Jul.
Article En | MEDLINE | ID: mdl-8698510

Most Escherichia coli isolates can express type 1 fimbriae with mannose-specific adhesins. These adhesins bind to the oligosaccharide chains of secretory immunoglobulin A (IgA). Thus, in addition to specific antibody activity, secretory IgA possesses a broad reactivity with bacteria expressing type 1 fimbriae. The absence of secretory IgA in colonic secretions, as seen in IgA deficiency, might therefore alter the ability of type 1-fimbriated E. coli to colonize the large intestines of these individuals. In the present study, 10 E. coli isolates from each of 17 IgA-deficient and 17 age-matched control individuals were assessed for the carriage of the fim gene cluster by DNA-DNA hybridization and for the expression of type 1 fimbriae by hemagglutination of guinea pig erythrocytes. The contribution of type 1-fimbria-mediated adherence to HT-29 colonic cells was also analyzed. The proportion of fim+ E. coli isolates was lower in IgA-deficient than in control individuals (74 versus 94%, P < 0.05), as was the proportion of isolates expressing type 1 fimbriae in vitro (69% versus 85%, P < 0.05). The median mannose-sensitive adherence to HT-29 cells was lower for isolates from IgA-deficient individuals than from the controls (9 versus 26 bacteria per cell, P < 0.05). Isolates expressing type 1 fimbriae showed lower adherence to HT-29 cells when they were derived from IgA-deficient individuals than when they were derived from control individuals (15 versus 27 bacteria per cell, P < 0.05). The results suggest that the interaction of type 1 fimbriae with secretory IgA contributes to the large intestinal colonization by these bacteria.


Adhesins, Bacterial/metabolism , Colon/microbiology , Escherichia coli/metabolism , IgA Deficiency/metabolism , IgA Deficiency/microbiology , Mannose/metabolism , Adhesins, Bacterial/genetics , Adult , Aged , Binding Sites , Case-Control Studies , Enterobacteriaceae/isolation & purification , Escherichia coli/genetics , Escherichia coli/isolation & purification , Female , Fimbriae, Bacterial/genetics , Fimbriae, Bacterial/metabolism , Genes, Bacterial , Genotype , Humans , Male , Middle Aged
12.
Exp Clin Immunogenet ; 13(1): 1-6, 1996.
Article En | MEDLINE | ID: mdl-8854082

The aim of the study was to investigate the production of IgG from alternative alleles of IGHCG1, IGHCG2 and IGHCG3, closely related to IGHCA1 and IGHCA2 on chromosome 14, in IgA deficient (IgAD; serum IgA levels < 0.05 g/l) for individuals. Sixty-two IgAD individuals were included in the study and sera were investigated with the sensitive competitive indirect ELISA for measuring serum concentrations of the Gm allotypes G1m(a), G1m(f), G2m(n) and G3m(b), performed with specific monoclonal antisera and purified myeloma proteins in combination with IgG subclass quantitation. The known 'compensatorily increased' serum levels of IgG1 and IgG3 were recognized with significantly increased G1m(a) and G1m(f) from IGHCG1 and significantly increased G1m(g) and G3m(b) from IGHCG3. The quotients of G1m(a)/G1m(f) from IGHCG1 and G3m(g)/G3m(b) from IGHCG3 were also normal. Instead, the levels of G2m(n) from IGHCG2 were selectively decreased in combination with normal or increased levels of G2m(") from the same IGHCG2. The quotient G2m(n)/G2m(") was also significantly decreased. As the selectively decreased G2m(n) allotype expression from IGHCG2 was situated close to the non-expressing IGHCA1, the origin of most serum IgA could be the result of a defective common regulating factor. The selectively decreased G2m(n) allotype levels from IGHCG2 must also be discussed with a view to Gm allotype suppression described in mice. The selectively decreased G2m(n) allotype levels in G2m(n,") heterozygous IgAD individuals could be the result of a preferential allelic exclusion of G2m(n) favoring G2m(") on IGHCG2 in many cells.


IgA Deficiency/metabolism , Immunoglobulin G/biosynthesis , Immunoglobulin G/classification , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Heavy Chains/genetics , Alleles , Humans , Immunoglobulin G/analysis , Immunoglobulin G/genetics , Immunoglobulin Heavy Chains/analysis
...