Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 96
Filtrer
1.
Clin Exp Immunol ; 215(2): 160-176, 2024 02 07.
Article de Anglais | MEDLINE | ID: mdl-37724703

RÉSUMÉ

Recombination activating genes (RAG)1 and RAG2 deficiency leads to combined T/B-cell deficiency with varying clinical presentations. This study aimed to define the clinical/laboratory spectrum of RAG1 and RAG2 deficiency. We retrospectively reviewed the clinical/laboratory data of 35 patients, grouped them as severe combined immunodeficiency (SCID), Omenn syndrome (OS), and delayed-onset combined immunodeficiency (CID) and reported nine novel mutations. The male/female ratio was 23/12. Median age of clinical manifestations was 1 months (mo) (0.5-2), 2 mo (1.25-5), and 14 mo (3.63-27), age at diagnosis was 4 mo (3-6), 4.5 mo (2.5-9.75), and 27 mo (14.5-70) in SCID (n = 25; 71.4%), OS (n = 5; 14.3%), and CID (n = 5; 14.3%) patients, respectively. Common clinical manifestations were recurrent sinopulmonary infections 82.9%, oral moniliasis 62.9%, diarrhea 51.4%, and eczema/dermatitis 42.9%. Autoimmune features were present in 31.4% of the patients; 80% were in CID patients. Lymphopenia was present in 92% of SCID, 80% of OS, and 80% of CID patients. All SCID and CID patients had low T (CD3, CD4, and CD8), low B, and increased NK cell numbers. Twenty-eight patients underwent hematopoietic stem cell transplantation (HSCT), whereas seven patients died before HSCT. Median age at HSCT was 7 mo (4-13.5). Survival differed in groups; maximum in SCID patients who had an HLA-matched family donor, minimum in OS. Totally 19 (54.3%) patients survived. Early molecular genetic studies will give both individualized therapy options, and a survival advantage because of timely diagnosis and treatment. Further improvement in therapeutic outcomes will be possible if clinicians gain time for HSCT.


Sujet(s)
Lymphopénie , Maladies d'immunodéficience primaire , Immunodéficience combinée grave , Humains , Mâle , Femelle , Nourrisson , Protéines à homéodomaine/génétique , Études rétrospectives , Immunodéficience combinée grave/génétique , Mutation , Protéines de liaison à l'ADN/génétique , Protéines nucléaires/génétique
2.
J Allergy Clin Immunol Pract ; 11(10): 3030-3039, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37391021

RÉSUMÉ

Cutaneous manifestations are common in patients with inborn errors of immunity (IEI)/primary immunodeficiency and could be due to infections, immune dysregulation, or lymphoproliferative/malign diseases. Immunologists accept some as warning signs for underlying IEI. Herein, we include noninfectious/infectious cutaneous manifestations that we come across in rare IEI cases in our clinic and provide a comprehensive literature review. For several skin diseases, the diagnosis is challenging and differential diagnosis is necessary. Detailed disease history and examination play a vital role in reaching a diagnosis, especially if there is a potential underlying IEI. A skin biopsy is sometimes necessary, especially if we need to rule out inflammatory, infectious, lymphoproliferative, and malignant conditions. Specific and immunohistochemical stainings are particularly important when diagnosing granuloma, amyloidosis, malignancies, and infections like human herpes virus-6, human herpes virus-8, human papillomavirus, and orf. Elucidation of mechanisms of IEIs has improved our understanding of their relation to cutaneous findings. In challenging cases, the immunological evaluation may lead the approach when there is a specific primary immunodeficiency diagnosis or at least help to reduce the number of differential diagnoses. Conversely, the response to therapy may provide conclusive evidence for some conditions. This review raises awareness of concomitant lesions and expands the scope of the differential diagnosis of IEI and the spectrum of skin disease therapy by highlighting frequent forms of IEI-associated cutaneous manifestations. The manifestations given here will guide clinicians to plan for alternative use of diverse therapeutics in a multidisciplinary way for skin diseases.


Sujet(s)
Herpèsvirus humain de type 6 , Maladies de la peau , Humains , Diagnostic différentiel , Établissements de soins ambulatoires , Biopsie , Maladies de la peau/diagnostic
3.
Scand J Immunol ; 96(5): e13213, 2022 Nov.
Article de Anglais | MEDLINE | ID: mdl-36808635

RÉSUMÉ

Isotype class-switch recombination (CSR), somatic hypermutation (SHM), B cell signalling and DNA repair mechanisms defects are responsible for high IgM. The hyperimmunoglobulin M (HIGM) phenotype and CSR-related defects are now classified under primary antibody defects, combined immunodeficiencies or syndromic immunodeficiencies groups. The aim of the study is to evaluate the diverse phenotypic/genotypic/laboratory characteristics and outcome of patients with CSR defects and HIGM-related defects. We enrolled 50 patients. The most common gene defect was Activation-induced cytidine deaminase (AID) deficiency (n = 18), followed by CD40 Ligand (CD40L) (n = 14) and CD40 (n = 3) deficiency. Median ages at first symptom and diagnosis were significantly lower in CD40L deficiency (8.5 and 30 months, respectively) than AID deficiency (30 and 114 months, respectively) (p = .001 and p = .008, respectively). Frequent clinical symptoms were recurrent (66%) and severe (14.9%) infections, and/or autoimmune/non-infectious inflammatory features (48.4%). Eosinophilia and neutropenia were at a higher rate in CD40L deficiency patients (77.8%, p = .002 and 77.8%, p = .002, respectively) when compared to AID deficiency. Median serum IgM level was low in 28.6% of CD40L deficiency patients. It was significantly lower when compared to AID deficiency (p < 0.001). Six patients (CD40L deficiency n = 4, CD40 deficiency n = 2) underwent hematopoietic stem cell transplantation. Five were alive at the last visit. Four patients two patients with CD40L deficiency, one with CD40 deficiency and one with AID deficiency had novel mutations. In conclusion; patients with CSR defects and HIGM phenotype may present with a wide range of clinical manifestations and laboratory findings. Low IgM, neutropenia and eosinophilia were prominent in patients with CD40L deficiency. Characterization of genetic defect-specific clinical and laboratory features may ease the diagnosis, prevent the underdiagnoses of patients and ameliorate the outcome.


Sujet(s)
Syndrome d'hyper-IgM , Neutropénie , Humains , Ligand de CD40/génétique , Commutation de classe des immunoglobulines , Syndrome d'hyper-IgM/génétique , Immunoglobuline M , Neutropénie/génétique , Cytidine deaminase
4.
Immunol Invest ; 51(4): 739-747, 2022 May.
Article de Anglais | MEDLINE | ID: mdl-33472463

RÉSUMÉ

BACKGROUND: We aimed to evaluate hematopoietic stem cell transplantation (HSCT) related outcomes of patients with severe combined immunodeficiency (SCID). METHODS: We retrospectively collected data from SCID patients who were diagnosed, followed up and survived at least 2 years after HSCT. RESULTS: Forty four SCID patients were included in the study. Median age of HSCT and follow-up period after HSCT were 7.1 months and 8.7 years, respectively. Human leukocyte antigen (HLA) identical donors were used in 77.3% (n = 34) of the patients (23 siblings, six fathers, two mothers, three extended family donors), HLA 1-2 mismatched family donors in 11.3% (n = 5), and haploidentical family donors in 11.3% (n = 5). CD3 and CD19 counts were normal in more than 90% and in 45.4% at last follow-up, respectively. Intravenous immunoglobulin (IVIG) could be stopped in 72.7% (n = 32) after HSCT. B+ SCID patients had better CD19 counts than B- (p < .001). T cell numbers, lymphocyte proliferation, IVIG need, immunoglobulin levels, antibody responses did not differ among B- and B+ immunophenotypes. Acute graft-versus-host disease (GVHD) was less in bone marrow transplanted patients (19.4%) than peripheral stem cell (58.3%) transplanted ones (p = .024). There was no correlation between age at transplantation and immune reconstitution. At the last follow-up, 70.2% and 78.3% of the patients had body weight and height above 3rd percentile, respectively. CONCLUSION: The immune reconstitution and the growth were normal in the majority of SCID patients after HSCT. It may be rational to use bone marrow instead of peripheral stem cell, as acute GVHD was less in bone marrow transplanted patients.


Sujet(s)
Maladie du greffon contre l'hôte , Transplantation de cellules souches hématopoïétiques , Immunodéficience combinée grave , Études de suivi , Maladie du greffon contre l'hôte/étiologie , Transplantation de cellules souches hématopoïétiques/effets indésirables , Humains , Immunoglobulines par voie veineuse , Études rétrospectives , Immunodéficience combinée grave/étiologie , Immunodéficience combinée grave/thérapie
5.
Immunol Invest ; 51(5): 1272-1283, 2022 Jul.
Article de Anglais | MEDLINE | ID: mdl-34098853

RÉSUMÉ

OBJECTIVE: Wiskott Aldrich Syndrome is an X-linked primary immunodeficiency disorder characterized by microthrombocytopenia, severe immunodeficiency, and eczema. To define clinical-laboratory features, genetic defects (known/novel) of 23 patients of Wiskott Aldrich Syndrome/X-linked Thrombocytopenia (WAS/XLT) cohort, establish relationships between molecular defects and clinical features if present, evaluate patients who underwent hematopoietic stem cell transplantation (HSCT) and did not. METHODS: Qualitative analysis from patients' hospital files and Sanger sequencing for molecular diagnosis was performed. Twenty-two WAS patients and one XLT patient were included in the study. RESULTS: The median age of diagnosis was 15 months (2.5-172 months). The most common symptom was otitis media and all patients had microthrombocytopenia. Autoimmune findings were detected in 34.7% (8 patients) of the patients; three patients (13%) had positive anti-nuclear antibody (ANA), three patients (13%) hemolytic anemia, one patient autoimmune neutropenia, two patients vasculitis, and one patient demyelinating polyneuropathy. Nine of the 23 (39,1%) patients had HSCT with nearly 90% success. We identified 13 different mutations in our cohort; seven were novel. CONCLUSIONS: HSCT is the only curative treatment for WAS. The study confirms that early diagnosis is very important for the success of therapy, so we must increase awareness in society and physicians to keep an eye out for clues. Our study cohort and follow-up period are not sufficient to establish phenotype-genotype correlation, so a larger cohort from various centers with longer follow-up will be more decisive.


Sujet(s)
Maladies génétiques liées au chromosome X , Thrombopénie , Syndrome de Wiskott-Aldrich , Maladies génétiques liées au chromosome X/génétique , Humains , Mutation , Thrombopénie/diagnostic , Thrombopénie/génétique , Syndrome de Wiskott-Aldrich/diagnostic , Syndrome de Wiskott-Aldrich/génétique , Syndrome de Wiskott-Aldrich/thérapie , Protéine du syndrome de Wiskott-Aldrich/génétique
6.
J Allergy Clin Immunol Pract ; 9(10): 3752-3766.e4, 2021 10.
Article de Anglais | MEDLINE | ID: mdl-34146746

RÉSUMÉ

BACKGROUND: Serine/threonine kinase-4 (STK4) deficiency is an autosomal recessive combined immunodeficiency. OBJECTIVE: We aimed to define characteristic clinical and laboratory features to aid the differential diagnosis and determine the most suitable therapy. METHODS: In addition to nine STK4 deficiency patients, we reviewed 15 patients from the medical literature. We compared B lymphocyte subgroups of the cohort with age-matched healthy controls. RESULTS: In the cohort, median age at symptom onset and age at diagnosis were 6 years 8 months (range, 6-248 months) and 7 years 5 months (range, 6-260 months), respectively. The main clinical findings were infections (in all nine patients [9 of 9]), autoimmune or inflammatory diseases (7 of 9), and atopy (4 of 9). CD4 lymphopenia (9 out 9), lymphopenia (7 out 9), intermittent eosinophilia (4 out 9), transient neutropenia (3 out 9), low IgM (4 out 9), and high IgE (4 out 9) were common. Decreased recent thymic emigrants, naive and central memory T cells, but increased effector memory T cells were present. The increase in plasmablasts (P = .003) and decrease in switched memory B cells (P = .022) were significant. When 24 patients are analyzed, cutaneous viral infections (n = 20), recurrent pneumonia (n = 18), Epstein Barr virus-associated lymphoproliferation (n = 11), atopic dermatitis (n = 10), autoimmune cytopenia (n = 7), and lymphoma (n = 6) were frequent. Lymphopenia, CD4 lymphopenia, high IgG, IgA, and IgE were common laboratory characteristics. CONCLUSIONS: The differential diagnosis with autosomal recessive hyper-IgE syndrome is crucial. Because, atopy and CD4 lymphopenia are common in both diseases. Immunoglobulins, antibacterial, and antiviral prophylaxis are the mainstays of treatment. Clinicians may use immunomodulatory therapies during inflammatory or autoimmune complications. However, more data are needed to recommend hematopoietic stem cell transplantation as a safe therapy.


Sujet(s)
Infections à virus Epstein-Barr , Lymphopénie , Herpèsvirus humain de type 4 , Humains , Protéines et peptides de signalisation intracellulaire , Lymphopénie/diagnostic , Protein-Serine-Threonine Kinases , Sérine , Thréonine
7.
J Clin Immunol ; 41(6): 1272-1290, 2021 08.
Article de Anglais | MEDLINE | ID: mdl-33929673

RÉSUMÉ

Biallelic inactivating mutations in IL21R causes a combined immunodeficiency that is often complicated by cryptosporidium infections. While eight IL-21R-deficient patients have been reported previously, the natural course, immune characteristics of disease, and response to hematopoietic stem cell transplantation (HSCT) remain to be comprehensively examined. In our study, we have collected clinical histories of 13 patients with IL-21R deficiency from eight families across seven centers worldwide, including five novel patients identified by exome or NGS panel sequencing. Eight unique mutations in IL21R were identified in these patients, including two novel mutations. Median age at disease onset was 2.5 years (0.5-7 years). The main clinical manifestations were recurrent bacterial (84.6%), fungal (46.2%), and viral (38.5%) infections; cryptosporidiosis-associated cholangitis (46.2%); and asthma (23.1%). Inflammatory skin diseases (15.3%) and recurrent anaphylaxis (7.9%) constitute novel phenotypes of this combined immunodeficiency. Most patients exhibited hypogammaglobulinemia and reduced proportions of memory B cells, circulating T follicular helper cells, MAIT cells and terminally differentiated NK cells. However, IgE levels were elevated in 50% of IL-21R-deficient patients. Overall survival following HSCT (6 patients, mean follow-up 1.8 year) was 33.3%, with pre-existing organ damage constituting a negative prognostic factor. Mortality of non-transplanted patients (n = 7) was 57.1%. Our detailed analysis of the largest cohort of IL-21R-deficient patients to date provides in-depth clinical, immunological and immunophenotypic features of these patients, thereby establishing critical non-redundant functions of IL-21/IL-21R signaling in lymphocyte differentiation, humoral immunity and host defense against infection, and mechanisms of disease pathogenesis due to IL-21R deficiency. Outcome following HSCT depends on prior chronic infections and organ damage, which should thus be considered as early as possible following molecular diagnosis.


Sujet(s)
Sous-unité alpha du récepteur à l'interleukine 21/déficit , Sous-unité alpha du récepteur à l'interleukine 21/génétique , Adolescent , Lymphocytes B/immunologie , Différenciation cellulaire/génétique , Différenciation cellulaire/immunologie , Enfant , Enfant d'âge préscolaire , Cryptosporidiose/génétique , Cryptosporidiose/immunologie , Cryptosporidium/immunologie , Femelle , Génomique/méthodes , Humains , Immunité humorale/génétique , Immunité humorale/immunologie , Nourrisson , Sous-unité alpha du récepteur à l'interleukine 21/immunologie , Activation des lymphocytes/génétique , Activation des lymphocytes/immunologie , Mâle , Cellules B mémoire/immunologie , Infection persistante/génétique , Infection persistante/immunologie , Phénotype , Transduction du signal/génétique , Transduction du signal/immunologie , Jeune adulte
8.
J Clin Immunol ; 39(7): 726-738, 2019 10.
Article de Anglais | MEDLINE | ID: mdl-31432443

RÉSUMÉ

INTRODUCTION: Autosomal recessively inherited lipopolysaccharide-responsive beige-like anchor (LRBA) protein deficiency was shown to be responsible for different types of inborn errors of immunity, such as common variable immunodeficiency (CVID) and autoimmune lymphoproliferative syndrome (ALPS). The aim of this study was to compare patients with LRBA-related ALPS and LRBA-related CVID, to describe their clinical and laboratory phenotypes, and to prepare an algorithm for their diagnosis and management. METHODS: Fifteen LRBA-deficient patients were identified among 31 CVID and 14 possible ALPS patients with Western blotting (WB), primary immunodeficiency disease (PIDD) gene, next-generation panel screening (NGS), and whole exome sequencing (WES). RESULTS: The median age on admission and age of diagnosis were 7 years (0.3-16.5) and 11 years (5-44), respectively. Splenomegaly was seen in 93.3% (14/15) of the patients on admission. Splenectomy was performed to 1/5. Recurrent upper respiratory tract infections (93.3% (14/15)), autoimmune cytopenia (80% (12/15)), chronic diarrhea (53.3% (8/15)), lower respiratory tract infections (53.3% (8/15)), lymphoma (26.6% (4/15)), Evans syndrome (26.6% (4/15)), and autoimmune thyroiditis (20% (3/15)) were common clinical findings and diseases. Lymphopenia (5/15), intermittant neutropenia (4/15), eosinophilia (4/15), and progressive hypogammaglobulinemia are recorded in given number of patients. Double negative T cells (TCRαß+CD4-CD8-) were increased in 80% (8/10) of the patients. B cell percentage/numbers were low in 60% (9/15) of the patients on admission. Decreased switched memory B cells, decreased naive and recent thymic emigrant (RTE) Thelper (Th) cells, markedly increased effector memory/effector memory RA+ (TEMRA) Th were documented. Large PD1+ population, increased memory, and enlarged follicular helper T cell population in the CD4+ T cell compartment was seen in one of the patients. Most of the deleterious missense mutations were located in the DUF1088 and BEACH domains. Interestingly, one of the two siblings with the same homozygous LRBA defect did not have any clinical symptom. Hematopoietic stem cell transplantation (HSCT) was performed to 7/15 (46.6%) of the patients. Transplanted patients are alive and well after a median of 2 years (1-3). In total, one patient died from sepsis during adulthood before HSCT. CONCLUSION: Patients with LRBA deficiency may initially be diagnosed as CVID or ALPS in the clinical practice. Progressive decrease in B cells as well as IgG in ALPS-like patients and addition of IBD symptoms in the follow-up should raise the suspicion for LRBA deficiency. Decreased switched memory B cells, decreased naive and recent thymic emigrant (RTE) Th cells, and markedly increased effector memory/effector memory RA+ Th cells (TEMRA Th) cells are important for the diagnosis of the patients in addition to clinical features. Analysis of protein by either WB or flow cytometry is required when the clinicians come across especially with missense LRBA variants of uncertain significance. High rate of malignancy shows the regulatory T cell's important role of immune surveillance. HSCT is curative and succesful in patients with HLA-matched family donor.


Sujet(s)
Protéines adaptatrices de la transduction du signal/déficit , Syndrome lymphoprolifératif avec auto-immunité/diagnostic , Syndrome lymphoprolifératif avec auto-immunité/étiologie , Déficit immunitaire commun variable/diagnostic , Déficit immunitaire commun variable/étiologie , Études d'associations génétiques , Prédisposition génétique à une maladie , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Adolescent , Adulte , Syndrome lymphoprolifératif avec auto-immunité/complications , Syndrome lymphoprolifératif avec auto-immunité/thérapie , Marqueurs biologiques , Enfant , Enfant d'âge préscolaire , Association thérapeutique , Déficit immunitaire commun variable/complications , Déficit immunitaire commun variable/thérapie , Maladies transmissibles/étiologie , Femelle , Études d'associations génétiques/méthodes , Locus génétiques , Transplantation de cellules souches hématopoïétiques , Humains , Immunophénotypage , Mâle , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Résultat thérapeutique , , Jeune adulte
9.
J Allergy Clin Immunol Pract ; 7(3): 848-855, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30391550

RÉSUMÉ

BACKGROUND: Biallelic variations in the dedicator of cytokinesis 8 (DOCK8) gene cause a combined immunodeficiency with eczema, recurrent bacterial and viral infections, and malignancy. Natural disease outcome is dismal, but allogeneic hematopoietic stem cell transplantation (HSCT) can cure the disease. OBJECTIVE: To determine outcome of HSCT for DOCK8 deficiency and define possible outcome variables. METHODS: We performed a retrospective study of the results of HSCT in a large international cohort of DOCK8-deficient patients. RESULTS: We identified 81 patients from 22 centers transplanted at a median age of 9.7 years (range, 0.7-27.2 years) between 1995 and 2015. After median follow-up of 26 months (range, 3-135 months), 68 (84%) patients are alive. Severe acute (III-IV) or chronic graft versus host disease occurred in 11% and 10%, respectively. Causes of death were infections (n = 5), graft versus host disease (5), multiorgan failure (2), and preexistent lymphoma (1). Survival after matched related (n = 40) or unrelated (35) HSCT was 89% and 81%, respectively. Reduced-toxicity conditioning based on either treosulfan or reduced-dose busulfan resulted in superior survival compared with fully myeloablative busulfan-based regimens (97% vs 78%; P = .049). Ninety-six percent of patients younger than 8 years at HSCT survived, compared with 78% of those 8 years and older (P = .06). Of the 73 patients with chimerism data available, 65 (89%) had more than 90% donor T-cell chimerism at last follow-up. Not all disease manifestations responded equally well to HSCT: eczema, infections, and mollusca resolved quicker than food allergies or failure to thrive. CONCLUSIONS: HSCT is curative in most DOCK8-deficient patients, confirming this approach as the treatment of choice. HSCT using a reduced-toxicity regimen may offer the best chance for survival.


Sujet(s)
Facteurs d'échange de nucléotides guanyliques/déficit , Transplantation de cellules souches hématopoïétiques , Déficits immunitaires/thérapie , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Femelle , Maladie du greffon contre l'hôte , Humains , Déficits immunitaires/mortalité , Nourrisson , Estimation de Kaplan-Meier , Mâle , Jeune adulte
10.
Scand J Immunol ; 88(4): e12709, 2018 Oct.
Article de Anglais | MEDLINE | ID: mdl-30152873

RÉSUMÉ

PURPOSE: Transient hypogammaglobulinemia of infancy (THI) is a common immunodeficiency, but definitive diagnosis can only be made retrospectively. While the pathogenesis is still unknown, abnormalities have been reported in the B cell compartment. In this study, we analysed the B cell subsets of patients with an initial THI diagnosis (n = 20) and compared them with those of healthy age-matched Turkish children (n = 72). METHODS: Flow cytometric analyses of the B subsets were performed by staining with anti-CD27-PE, anti-CD19-PerCP, anti-IgD-FITC and anti-IgM-APC antibodies. RESULTS: During a median follow-up of 6.6 years, 13 patients whose IgG levels had normalized before they reached four years of age were diagnosed with definitive THI. The memory subsets of these patients were lower but not statistically different from the healthy controls (HC). The remaining seven patients had prolonged hypogammaglobulinemia after the age of four and had significantly lower memory B cell subsets compared to the HC. On follow-up, these patients had not experienced recurrent infections or autoimmunity. Re-evaluation of patients' B cell subsets six years later showed that the memory B cell ratios had increased to levels comparable to HC, despite the patients still having mildly low IgG levels. CONCLUSION: Patients with prolonged hypogammaglobulinemia had lower levels of memory B cells despite having a similar clinical course to patients who had been diagnosed with definitive THI. This subgroup of putative THI patients poses a diagnostic and classification dilemma. Our results suggested that these patients' memory B cells and IgG levels may recover over time.


Sujet(s)
Agammaglobulinémie/immunologie , Sous-populations de lymphocytes B/immunologie , Lymphocytes B/immunologie , Maladies néonatales/immunologie , Infections/immunologie , Agammaglobulinémie/diagnostic , Auto-immunité , Séparation cellulaire , Enfant , Enfant d'âge préscolaire , Femelle , Cytométrie en flux , Études de suivi , Humains , Immunoglobuline G/sang , Mémoire immunologique , Nourrisson , Nouveau-né , Maladies néonatales/diagnostic , Infections/diagnostic , Mâle , Évaluation des résultats des patients , Turquie
11.
J Clin Immunol ; 38(4): 484-493, 2018 05.
Article de Anglais | MEDLINE | ID: mdl-29744787

RÉSUMÉ

INTRODUCTION: Adenosine deaminase (ADA) deficiency is an autosomal recessive primary immunodeficiency. It results in the intracellular accumulation of toxic metabolites which have effects particularly on lymphocytes and the brain. The aim of this study was to evaluate the outcome of 13 ADA-deficient patients. We planned to evaluate their clinical and laboratory findings before and after enzyme replacement therapy (ERT), allogeneic hematopoietic stem cell transplantation (aHSCT), and hematopoietic stem cell gene therapy (HSCGT). METHODS: Measurement of ADA enzyme activity and metabolites and sequencing of the ADA gene were performed in most of the patients with ADA deficiency. One of the patients with late-onset ADA deficiency was diagnosed by the help of primary immunodeficiency panel screening. RESULTS: Ten out of 13 patients were diagnosed as SCID, while 3 out of 13 were diagnosed as delayed-/late-onset ADA deficiency. Late-onset ADA deficiency patients had clinical and laboratory findings of combined immunodeficiency (CID). Eight patients with ADA-SCID were found to have higher levels of ADA metabolite (dAXP%) (62.1% (34.6-71.9)) than 3 patients with delayed-/late-onset ADA deficiency (6.9% (2.1-8.9). All but one patient with SCID had T-B-NK- phenotype, one had T-B-NK+ phenotype. Genetic defect was documented in 11 patients. Four out of 11 patients had compound heterozygous defects. Three out of 4 patients with compound heterozygous defects had delayed-onset/late-onset ADA deficiency. Seven out of 11 patients with SCID had homozygous defects. Five out of 7 had the same homozygous indel frameshift mutation (c.955-959delGAAGA) showing a founder effect. There were two novel splice site defects: one (IVS10+2T>C) was heterozygous in a patient with late-onset ADA deficiency, and the other was homozygous (IVS2delT+2) in a SCID patient. Other defects were missense defects. Nine out of 13 patients were put on pegylated ADA ERT. Four out of six patients were transplanted without using a conditioning regimen. HSCGT was performed to one of the patients. CONCLUSION: The genetic diagnosis of SCID is utmost important. There is a chance to give ERT before the definitive therapy if the patient with SCID/CID has ADA deficiency. Although ERT was insufficient to restore a normal immune function in ADA-SCID patients, it was useful to improve and stabilize the clinical status before curative therapy (aHSCT/HSCGT). Enzyme replacement therapy was successful in patients with late-/delayed-onset ADA deficiency who presented with the features of combined immunodeficiency. Gastrointestinal polyposis in a patient with late-onset ADA deficiency may be an association or a coincidental finding. Intermittent neurodevelopmental evaluation especially for hearing impairment should be performed in most of the ADA-deficient patients. This may alleviate the speech delay and cognitive abnormalities which may be observed in the follow-up.


Sujet(s)
Adenosine deaminase/déficit , Agammaglobulinémie/diagnostic , Études d'associations génétiques , Immunodéficience combinée grave/diagnostic , Adenosine deaminase/sang , Adenosine deaminase/génétique , Agammaglobulinémie/mortalité , Agammaglobulinémie/thérapie , Âge de début , Marqueurs biologiques , Biopsie , Prise en charge de la maladie , Activation enzymatique , Thérapie enzymatique substitutive , Femelle , Dépistage génétique , Thérapie génétique , Génotype , Transplantation de cellules souches hématopoïétiques , Homozygote , Humains , Nourrisson , Nouveau-né , Mâle , Mutation , Phénotype , Analyse de séquence d'ADN , Immunodéficience combinée grave/mortalité , Immunodéficience combinée grave/thérapie , Résultat thérapeutique
13.
Front Immunol ; 8: 685, 2017.
Article de Anglais | MEDLINE | ID: mdl-28952612

RÉSUMÉ

Immunodeficiency-associated vaccine-derived polioviruses (iVDPVs) have been isolated from primary immunodeficiency (PID) patients exposed to oral poliovirus vaccine (OPV). Patients may excrete poliovirus strains for months or years; the excreted viruses are frequently highly divergent from the parental OPV and have been shown to be as neurovirulent as wild virus. Thus, these patients represent a potential reservoir for transmission of neurovirulent polioviruses in the post-eradication era. In support of WHO recommendations to better estimate the prevalence of poliovirus excreters among PIDs and characterize genetic evolution of these strains, 635 patients including 570 with primary antibody deficiencies and 65 combined immunodeficiencies were studied from 13 OPV-using countries. Two stool samples were collected over 4 days, tested for enterovirus, and the poliovirus positive samples were sequenced. Thirteen patients (2%) excreted polioviruses, most for less than 2 months following identification of infection. Five (0.8%) were classified as iVDPVs (only in combined immunodeficiencies and mostly poliovirus serotype 2). Non-polio enteroviruses were detected in 30 patients (4.7%). Patients with combined immunodeficiencies had increased risk of delayed poliovirus clearance compared to primary antibody deficiencies. Usually, iVDPV was detected in subjects with combined immunodeficiencies in a short period of time after OPV exposure, most for less than 6 months. Surveillance for poliovirus excretion among PID patients should be reinforced until polio eradication is certified and the use of OPV is stopped. Survival rates among PID patients are improving in lower and middle income countries, and iVDPV excreters are identified more frequently. Antivirals or enhanced immunotherapies presently in development represent the only potential means to manage the treatment of prolonged excreters and the risk they present to the polio endgame.

14.
Clin Immunol ; 178: 74-78, 2017 05.
Article de Anglais | MEDLINE | ID: mdl-28161407

RÉSUMÉ

Major histocompatibility complex (MHC) class I deficiency syndrome is a rare primary immunodeficiency caused by mutations in the peptide transporter complex associated with antigen presentation (TAP) gene which plays a crucial role in intracellular peptide antigen presentation. A few cases have been reported to date. Recurrent sinopulmonary infections and skin ulcers are the main characteristics of the syndrome. Here we report two siblings diagnosed with TAP1 deficiency syndrome associated only with recurrent sinopulmonary infections with the description of a novel mutation leading to a premature stop codon in TAP1 gene and review of the relevant literature. Both of the siblings had recurrent sinopulmonary infections since childhood, responded to antibiotherapy well, neither of them had hospitalization history because of infections. One had chronic hepatitis B infection which may possibly be related to TAP1 gene defect.


Sujet(s)
Membre-2 de la sous-famille B à cassette de liaison à l'ATP/génétique , Mutation avec décalage du cadre de lecture , Immunodéficience combinée grave/génétique , Adolescent , Adulte , Dilatation des bronches/étiologie , Dilatation des bronches/immunologie , Femelle , Hépatite B chronique/étiologie , Hépatite B chronique/immunologie , Antigènes d'histocompatibilité de classe I/immunologie , Humains , Mâle , Récidive , Infections de l'appareil respiratoire/étiologie , Infections de l'appareil respiratoire/immunologie , Immunodéficience combinée grave/complications , Immunodéficience combinée grave/immunologie , Fratrie , Jeune adulte
15.
Turk J Pediatr ; 59(5): 555-560, 2017.
Article de Anglais | MEDLINE | ID: mdl-29745117

RÉSUMÉ

Streptococcus pneumoniae is a common pathogen responsible for pulmonary infections and the leading cause of mortality and morbidity in patients with particularly B cell immunodeficiencies. Antibody production is the principal protective immune response against S. pneumoniae and measurement of the production of antipolysaccharide antibodies is important in the evaluation of B cell deficiencies. We quantified serotype-specific immunoglobulin G antibodies against seven common pneumococcal serotypes before and three weeks after unconjugated vaccine in 416 patients with recurrent respiratory tract infections; fifty-five (13%) of whom showed impaired antibody response. We could evaluate 41 of these 55 patients for their particular clinical features. Specific antibody deficiency, was diagnosed in 10 of these patients, common variable immunodeficiency in 18, ataxia telangiectasia in 10 and other antibody deficiencies in 7 (transient hypogammaglobulinemia in 4, IgG subclass deficiency in 1, partial and selective IgA deficiency in 1) patients. Evaluation of the antibody response to polysaccharide antigens should be considered early on in patients with recurrent respiratory infections and required particularly for the diagnosis of specific antibody deficiency and the decision of the appropriate treatment approaches.


Sujet(s)
Anticorps antibactériens/sang , Vaccins antipneumococciques/immunologie , Infections de l'appareil respiratoire/immunologie , Streptococcus pneumoniae/immunologie , Adolescent , Adulte , Production d'anticorps , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Immunoglobuline G/sang , Déficits immunitaires/diagnostic , Mâle , Adulte d'âge moyen , Infections à pneumocoques/prévention et contrôle , Récidive , Jeune adulte
17.
Nat Immunol ; 17(12): 1352-1360, 2016 Dec.
Article de Anglais | MEDLINE | ID: mdl-27776107

RÉSUMÉ

RASGRP1 is an important guanine nucleotide exchange factor and activator of the RAS-MAPK pathway following T cell antigen receptor (TCR) signaling. The consequences of RASGRP1 mutations in humans are unknown. In a patient with recurrent bacterial and viral infections, born to healthy consanguineous parents, we used homozygosity mapping and exome sequencing to identify a biallelic stop-gain variant in RASGRP1. This variant segregated perfectly with the disease and has not been reported in genetic databases. RASGRP1 deficiency was associated in T cells and B cells with decreased phosphorylation of the extracellular-signal-regulated serine kinase ERK, which was restored following expression of wild-type RASGRP1. RASGRP1 deficiency also resulted in defective proliferation, activation and motility of T cells and B cells. RASGRP1-deficient natural killer (NK) cells exhibited impaired cytotoxicity with defective granule convergence and actin accumulation. Interaction proteomics identified the dynein light chain DYNLL1 as interacting with RASGRP1, which links RASGRP1 to cytoskeletal dynamics. RASGRP1-deficient cells showed decreased activation of the GTPase RhoA. Treatment with lenalidomide increased RhoA activity and reversed the migration and activation defects of RASGRP1-deficient lymphocytes.


Sujet(s)
Actines/métabolisme , Lymphocytes B/immunologie , Cytosquelette/métabolisme , Protéines de liaison à l'ADN/génétique , Facteurs d'échange de nucléotides guanyliques/génétique , Déficits immunitaires/génétique , Cellules tueuses naturelles/immunologie , Lymphocytes T/immunologie , Adolescent , Inhibiteurs de l'angiogenèse/pharmacologie , Lymphocytes B/effets des médicaments et des substances chimiques , Mouvement cellulaire/effets des médicaments et des substances chimiques , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Enfant , Cytotoxicité immunologique/génétique , Analyse de mutations d'ADN , Dynéines/métabolisme , Femelle , Cellules HEK293 , Humains , Commutation de classe des immunoglobulines/génétique , Déficits immunitaires/traitement médicamenteux , Cellules Jurkat , Cellules tueuses naturelles/effets des médicaments et des substances chimiques , Lénalidomide , Mâle , Mutation/génétique , Pedigree , Petit ARN interférent/génétique , Lymphocytes T/effets des médicaments et des substances chimiques , Thalidomide/analogues et dérivés , Thalidomide/pharmacologie
18.
Nat Commun ; 7: 11496, 2016 05 19.
Article de Anglais | MEDLINE | ID: mdl-27193971

RÉSUMÉ

ISG15 is an interferon (IFN)-α/ß-induced ubiquitin-like protein. It exists as a free molecule, intracellularly and extracellularly, and conjugated to target proteins. Studies in mice have demonstrated a role for Isg15 in antiviral immunity. By contrast, human ISG15 was shown to have critical immune functions, but not in antiviral immunity. Namely, free extracellular ISG15 is crucial in IFN-γ-dependent antimycobacterial immunity, while free intracellular ISG15 is crucial for USP18-mediated downregulation of IFN-α/ß signalling. Here we describe ISG15-deficient patients who display no enhanced susceptibility to viruses in vivo, in stark contrast to Isg15-deficient mice. Furthermore, fibroblasts derived from ISG15-deficient patients display enhanced antiviral protection, and expression of ISG15 attenuates viral resistance to WT control levels. The species-specific gain-of-function in antiviral immunity observed in ISG15 deficiency is explained by the requirement of ISG15 to sustain USP18 levels in humans, a mechanism not operating in mice.


Sujet(s)
Cytokines/métabolisme , Ubiquitines/métabolisme , Maladies virales/immunologie , Animaux , Lignée cellulaire , Cytokines/génétique , Cytokines/immunologie , Femelle , Régulation de l'expression des gènes , Humains , Interférons/métabolisme , Souris , Culture de cellules primaires , Ubiquitin thiolesterase/métabolisme , Ubiquitines/génétique , Ubiquitines/immunologie
19.
J Allergy Clin Immunol ; 137(3): 879-88.e2, 2016 Mar.
Article de Anglais | MEDLINE | ID: mdl-26476480

RÉSUMÉ

BACKGROUND: Coronin-1A (CORO1A) is a regulator of actin dynamics important for T-cell homeostasis. CORO1A deficiency causes T(-)B(+) natural killer-positive severe combined immunodeficiency or T-cell lymphopenia with severe viral infections. However, because all known human mutations in CORO1A abrogate protein expression, the role of the protein's functional domains in host immunity is unknown. OBJECTIVE: We sought to identify the cause of the primary immunodeficiency in 2 young adult siblings with a history of disseminated varicella, cutaneous warts, and CD4(+) T-cell lymphopenia. METHODS: We performed immunologic, genetic, and biochemical studies in the patients, family members, and healthy control subjects. RESULTS: Both patients had CD4(+) T-cell lymphopenia and decreased lymphocyte proliferation to mitogens. IgG, IgM, IgA, and specific antibody responses were normal. Whole-genome sequencing identified a homozygous frameshift mutation in CORO1A disrupting the last 2 C-terminal domains by replacing 61 amino acids with a novel 91-amino-acid sequence. The CORO1A(S401fs) mutant was expressed in the patients' lymphocytes at a level comparable with that of wild-type CORO1A in normal lymphocytes but did not oligomerize and had impaired cytoskeletal association. CORO1A(S401fs) was associated with increased filamentous actin accumulation in T cells, severely defective thymic output, and impaired T-cell survival but normal calcium flux and cytotoxicity, demonstrating the importance of CORO1A oligomerization and subcellular localization in T-cell homeostasis. CONCLUSIONS: We describe a truncating mutation in CORO1A that permits protein expression and survival into young adulthood. Our studies demonstrate the importance of intact CORO1A C-terminal domains in thymic egress and T-cell survival, as well as in defense against viral pathogens.


Sujet(s)
Cytosquelette/métabolisme , Homozygote , Protéines des microfilaments/génétique , Mutation , Multimérisation de protéines , Maladies virales/étiologie , Maladies virales/métabolisme , Actines/composition chimique , Actines/métabolisme , Adolescent , Animaux , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Dégranulation cellulaire/génétique , Dégranulation cellulaire/immunologie , Survie cellulaire/génétique , Analyse de mutations d'ADN , Femelle , Mutation avec décalage du cadre de lecture , Humains , Immunoglobulines/sang , Immunoglobulines/immunologie , Numération des lymphocytes , Lymphopénie , Mâle , Souris , Protéines des microfilaments/composition chimique , Protéines des microfilaments/métabolisme , Pedigree , Phénotype , Multimérisation de protéines/génétique , Transport des protéines , Fratrie , Transduction du signal , Maladies de la peau/anatomopathologie , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Maladies virales/diagnostic , Verrues/anatomopathologie
20.
Turk J Pediatr ; 58(4): 356-361, 2016.
Article de Anglais | MEDLINE | ID: mdl-28276206

RÉSUMÉ

Mendelian susceptibility to mycobacterial disease (MSMD) is characterized by infections with weakly virulent mycobacteria (BCG and environmental mycobacteria), M. tuberculosis, Salmonella, candida and some other intracellular microorganisms. Nine different genetic defects have been defined to cause MSMD and IL-12Rß1 deficiency is the most common form. We present here the clinical and genetic features of 18 patients with IL12Rß1 deficiency diagnosed by surface expression of IL-12Rß1 and Sanger's sequencing. Seventeen patients showed classical presentation (infections with BCG, salmonella and candida) while one patient experienced recurrent leishmaniasis. In all patients the percentage of activated lymphocytes with surface expression of IL12Rß1 was < 1% indicating that it is an effective method for the screening of these patients. Three recurrent mutations were responsible for 85% of our families. Prognosis was good in patients, in whom specific antimicrobial therapy was given before dissemination occurs, as well as prophylactic antimicrobial treatment when needed and IFN-γ therapy for severe infectious episodes.


Sujet(s)
Mycobacterium tuberculosis/immunologie , Récepteurs à l'interleukine-12/génétique , Adolescent , Enfant , Enfant d'âge préscolaire , Femelle , Prédisposition génétique à une maladie , Humains , Mâle , Mutation , Récepteurs à l'interleukine-12/déficit , Jeune adulte
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...