Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 125
Filter
Add more filters

Publication year range
1.
J Allergy Clin Immunol ; 153(1): 287-296, 2024 01.
Article in English | MEDLINE | ID: mdl-37793572

ABSTRACT

BACKGROUND: The Primary Immune Deficiency Treatment Consortium (PIDTC) enrolled children in the United States and Canada onto a retrospective multicenter natural history study of hematopoietic cell transplantation (HCT). OBJECTIVE: We investigated outcomes of HCT for severe combined immunodeficiency (SCID). METHODS: We evaluated the chronic and late effects (CLE) after HCT for SCID in 399 patients transplanted from 1982 to 2012 at 32 PIDTC centers. Eligibility criteria included survival to at least 2 years after HCT without need for subsequent cellular therapy. CLE were defined as either conditions present at any time before 2 years from HCT that remained unresolved (chronic), or new conditions that developed beyond 2 years after HCT (late). RESULTS: The cumulative incidence of CLE was 25% in those alive at 2 years, increasing to 41% at 15 years after HCT. CLE were most prevalent in the neurologic (9%), neurodevelopmental (8%), and dental (8%) categories. Chemotherapy-based conditioning was associated with decreased-height z score at 2 to 5 years after HCT (P < .001), and with endocrine (P < .001) and dental (P = .05) CLE. CD4 count of ≤500 cells/µL and/or continued need for immunoglobulin replacement therapy >2 years after transplantation were associated with lower-height z scores. Continued survival from 2 to 15 years after HCT was 90%. The presence of any CLE was associated with increased risk of late death (hazard ratio, 7.21; 95% confidence interval, 2.71-19.18; P < .001). CONCLUSION: Late morbidity after HCT for SCID was substantial, with an adverse impact on overall survival. This study provides evidence for development of survivorship guidelines based on disease characteristics and treatment exposure for patients after HCT for SCID.


Subject(s)
Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Child , Humans , Severe Combined Immunodeficiency/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Incidence , Canada/epidemiology , Retrospective Studies , Transplantation Conditioning
2.
J Allergy Clin Immunol ; 151(6): 1634-1645, 2023 06.
Article in English | MEDLINE | ID: mdl-36638922

ABSTRACT

BACKGROUND: Allogenic hematopoietic stem cell transplantation (HSCT) and gene therapy (GT) are potentially curative treatments for severe combined immunodeficiency (SCID). Late-onset posttreatment manifestations (such as persistent hepatitis) are not uncommon. OBJECTIVE: We sought to characterize the prevalence and pathophysiology of persistent hepatitis in transplanted SCID patients (SCIDH+) and to evaluate risk factors and treatments. METHODS: We used various techniques (including pathology assessments, metagenomics, single-cell transcriptomics, and cytometry by time of flight) to perform an in-depth study of different tissues from patients in the SCIDH+ group and corresponding asymptomatic similarly transplanted SCID patients without hepatitis (SCIDH-). RESULTS: Eleven patients developed persistent hepatitis (median of 6 years after HSCT or GT). This condition was associated with the chronic detection of enteric viruses (human Aichi virus, norovirus, and sapovirus) in liver and/or stools, which were not found in stools from the SCIDH- group (n = 12). Multiomics analysis identified an expansion of effector memory CD8+ T cells with high type I and II interferon signatures. Hepatitis was associated with absence of myeloablation during conditioning, split chimerism, and defective B-cell function, representing 25% of the 44 patients with SCID having these characteristics. Partially myeloablative retransplantation or GT of patients with this condition (which we have named as "enteric virus infection associated with hepatitis") led to the reconstitution of T- and B-cell immunity and remission of hepatitis in 5 patients, concomitantly with viral clearance. CONCLUSIONS: Enteric virus infection associated with hepatitis is related to chronic enteric viral infection and immune dysregulation and is an important risk for transplanted SCID patients with defective B-cell function.


Subject(s)
Enterovirus Infections , Hematopoietic Stem Cell Transplantation , Hepatitis , Severe Combined Immunodeficiency , Virus Diseases , Humans , Severe Combined Immunodeficiency/therapy , Severe Combined Immunodeficiency/etiology , CD8-Positive T-Lymphocytes , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Virus Diseases/etiology , Hepatitis/etiology
3.
Eur J Immunol ; 51(5): 1028-1038, 2021 05.
Article in English | MEDLINE | ID: mdl-33682138

ABSTRACT

The RAG1 and RAG2 proteins initiate the process of V(D)J recombination and therefore play an essential role in adaptive immunity. While null mutations in the RAG genes cause severe combined immune deficiency with lack of T and B cells (T- B- SCID) and susceptibility to life-threatening, early-onset infections, studies in humans and mice have demonstrated that hypomorphic RAG mutations are associated with defects of central and peripheral tolerance resulting in immune dysregulation. In this review, we provide an overview of the extended spectrum of RAG deficiencies and their associated clinical and immunological phenotypes in humans. We discuss recent advances in the mechanisms that control RAG expression and function, the effects of perturbed RAG activity on lymphoid development and immune homeostasis, and propose novel approaches to correct this group of disorders.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Nuclear Proteins/genetics , Severe Combined Immunodeficiency/etiology , Severe Combined Immunodeficiency/therapy , V(D)J Recombination/genetics , Animals , Diagnosis, Differential , Disease Management , Disease Models, Animal , Genetic Association Studies , Genetic Therapy , Genotype , Humans , Mutation , Phenotype , Severe Combined Immunodeficiency/diagnosis
4.
Immunol Invest ; 51(4): 739-747, 2022 May.
Article in English | MEDLINE | ID: mdl-33472463

ABSTRACT

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.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Severe Combined Immunodeficiency , Follow-Up Studies , Graft vs Host Disease/etiology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Immunoglobulins, Intravenous , Retrospective Studies , Severe Combined Immunodeficiency/etiology , Severe Combined Immunodeficiency/therapy
5.
J Clin Immunol ; 41(3): 631-638, 2021 04.
Article in English | MEDLINE | ID: mdl-33411152

ABSTRACT

Severe combined immunodeficiency (SCID) is a heterogeneous group of primary immunodeficiency diseases (PIDs) characterized by a lack of autologous T lymphocytes. This severe PID is rare, but has a higher prevalence in populations with high rates of consanguinity. The epidemiological, clinical, and immunological features of SCIDs in Moroccan patients have never been reported. The aim of this study was to provide a clinical and immunological description of SCID in Morocco and to assess changes in the care of SCID patients over time. This cross-sectional retrospective study included 96 Moroccan patients referred to the national PID reference center at Casablanca Children's Hospital for SCID over two decades, from 1998 to 2019. The case definition for this study was age < 2 years, with a clinical phenotype suggestive of SCID, and lymphopenia, with very low numbers of autologous T cells, according to the IUIS Inborn Errors of Immunity classification. Our sample included 50 male patients, and 66% of the patients were born to consanguineous parents. The median age at onset and diagnosis were 3.3 and 6.5 months, respectively. The clinical manifestations commonly observed in these patients were recurrent respiratory tract infection (82%), chronic diarrhea (69%), oral candidiasis (61%), and failure to thrive (65%). The distribution of SCID phenotypes was as follows: T-B-NK+ in 44.5%, T-B-NK- in 32%, T-B+NK- in 18.5%, and T-B+NK+ in 5%. An Omenn syndrome phenotype was observed in 15 patients. SCID was fatal in 84% in the patients in our cohort, due to the difficulties involved in obtaining urgent access to hematopoietic stem cell transplantation, which, nevertheless, saved 16% of the patients. The autosomal recessive forms of the clinical and immunological phenotypes of SCID, including the T-B-NK+ phenotype in particular, were more frequent than those in Western countries. A marked improvement in the early detection of SCID cases over the last decade was noted. Despite recent progress in SCID diagnosis, additional efforts are required, for genetic confirmation and particularly for HSCT.


Subject(s)
Phenotype , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/epidemiology , Alleles , Biomarkers , Consanguinity , Cross-Sectional Studies , Diagnosis, Differential , Disease Management , Disease Susceptibility , Genetic Predisposition to Disease , Genotype , Humans , Inheritance Patterns , Morocco/epidemiology , Public Health Surveillance , Severe Combined Immunodeficiency/etiology
6.
J Clin Immunol ; 41(3): 595-602, 2021 04.
Article in English | MEDLINE | ID: mdl-33409868

ABSTRACT

In 2008, newborn screening (NBS) for severe combined immunodeficiency (SCID) began as a pilot study in Wisconsin and has recently been added to every state's newborn screen panel. The incidence of SCID is estimated at 1 per 58,000 births which may suggest infrequent NBS SCID screen positive results in states with low annual birth rates. In this study, we report our center's experience with NBS positive SCID screen referrals over a 10-year period. A total of 68 full-term newborns were referred to our center for confirmatory testing. Of these referrals, 50% were false positives, 12% were SCID diagnoses, 20% syndromic T cell lymphopenia (TCL) disorders, and 18% non-SCID, non-syndromic TCL. Through collaboration with our newborn screening lab, second-tier targeted gene sequencing was performed for newborns with SCID screen positive results from communities with known founder pathogenic variants and provided rapid genetic confirmation of SCID and non-SCID TCL disorders. Despite extensive genetic testing, two of the eight (25%) identified newborns with SCID diagnoses lacked a definable genetic defect. Additionally, our referrals included ten newborns who were otherwise healthy newborns with idiopathic TCL and varied CD3+ T cell number longitudinal trajectories. Collectively, referrals to our single site over a 10-year period describe a broad spectrum of medically actionable and idiopathic TCL disorders which highlight the importance of clinical immunology expertise in all states, demonstrate efficiencies and challenges for second-tier genetic testing, and further emphasize the need to development standardized evaluation algorithms for non-SCID TCL.


Subject(s)
Neonatal Screening , Severe Combined Immunodeficiency/epidemiology , Algorithms , Clinical Decision-Making , Disease Management , Disease Susceptibility , Genetic Association Studies , Genetic Predisposition to Disease , History, 21st Century , Humans , Infant, Newborn , Phenotype , Public Health Surveillance , Referral and Consultation , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/etiology , Severe Combined Immunodeficiency/history
7.
J Clin Immunol ; 41(4): 756-768, 2021 05.
Article in English | MEDLINE | ID: mdl-33464451

ABSTRACT

Human nude SCID is a rare autosomal recessive inborn error of immunity (IEI) characterized by congenital athymia, alopecia, and nail dystrophy. Few cases have been reported to date. However, the recent introduction of newborn screening for IEIs and high-throughput sequencing has led to the identification of novel and atypical cases. Moreover, immunological alterations have been recently described in patients carrying heterozygous mutations. The aim of this paper is to describe the extended phenotype associated with FOXN1 homozygous, compound heterozygous, or heterozygous mutations. We collected clinical and laboratory information of a cohort of 11 homozygous, 2 compound heterozygous, and 5 heterozygous patients with recurrent severe infections. All, except one heterozygous patient, had signs of CID or SCID. Nail dystrophy and alopecia, that represent the hallmarks of the syndrome, were not always present, while almost 50% of the patients developed Omenn syndrome. One patient with hypomorphic compound heterozygous mutations had a late-onset atypical phenotype. A SCID-like phenotype was observed in 4 heterozygous patients coming from the same family. A spectrum of clinical manifestations may be associated with different mutations. The severity of the clinical phenotype likely depends on the amount of residual activity of the gene product, as previously observed for other SCID-related genes. The severity of the manifestations in this heterozygous family may suggest a mechanism of negative dominance of the specific mutation or the presence of additional mutations in noncoding regions.


Subject(s)
Forkhead Transcription Factors/genetics , Heterozygote , Homozygote , Mutation , Phenotype , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/etiology , Cell Line , Child, Preschool , DNA Mutational Analysis , Disease Management , Female , Forkhead Transcription Factors/chemistry , Genetic Association Studies , Genetic Loci , Genetic Predisposition to Disease , Hematopoietic Stem Cell Transplantation , High-Throughput Nucleotide Sequencing , Humans , Male , Models, Molecular , Molecular Conformation , Pedigree , Severe Combined Immunodeficiency/therapy , Structure-Activity Relationship , Treatment Outcome
8.
J Clin Immunol ; 41(3): 621-628, 2021 04.
Article in English | MEDLINE | ID: mdl-33415666

ABSTRACT

PURPOSE: T cell receptor excision circle (TREC) quantification is a recent addition to newborn screening (NBS) programs and is intended to identify infants with severe combined immunodeficiencies (SCID). However, other primary immunodeficiency diseases (PID) have also been identified as the result of TREC screening. We recently reported a newborn with a low TREC level on day 1 of life who was diagnosed with WHIM (warts, hypogammaglobulinemia, infections, myelokathexis) syndrome, a non-SCID primary immunodeficiency caused by mutations in the chemokine receptor CXCR4. METHODS: We have now retrospectively reviewed the birth and clinical histories of all known WHIM infants born after the implementation of NBS for SCID. RESULTS: We identified six infants with confirmed WHIM syndrome who also had TREC quantification on NBS. Three of the six WHIM infants had low TREC levels on NBS. All six patients were lymphopenic but only one infant had a T cell count below 1,500 cells/µL. The most common clinical manifestation was viral bronchiolitis requiring hospitalization. One infant died of complications related to Tetralogy of Fallot, a known WHIM phenotype. CONCLUSION: The results suggest that WHIM syndrome should be considered in the differential diagnosis of newborns with low NBS TREC levels. TRIAL REGISTRATION: Not applicable.


Subject(s)
Neonatal Screening/methods , Primary Immunodeficiency Diseases/epidemiology , Primary Immunodeficiency Diseases/etiology , Receptors, Antigen, T-Cell/genetics , Warts/epidemiology , Warts/etiology , Biomarkers , DNA Mutational Analysis , Diagnosis, Differential , Disease Susceptibility , Female , Humans , Infant, Newborn , Male , Mutation , Phenotype , Primary Immunodeficiency Diseases/diagnosis , Receptors, CXCR4/genetics , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/epidemiology , Severe Combined Immunodeficiency/etiology , Warts/diagnosis
9.
J Clin Immunol ; 41(1): 99-108, 2021 01.
Article in English | MEDLINE | ID: mdl-33070266

ABSTRACT

PURPOSE: While neonatal bloodspot screening (NBS) for severe combined immunodeficiency (SCID) has been introduced more than a decade ago, implementation in NBS programs remains challenging in many countries. Even if high-quality test methods and follow-up care are available, public uptake and parental acceptance are not guaranteed. The aim of this study was to describe the parental perspective on NBS for SCID in the context of an implementation pilot. Psychosocial aspects have never been studied before for NBS for SCID and are important for societal acceptance, a major criterion when introducing new disorders in NBS programs. METHODS: To evaluate the perspective of parents, interviews were conducted with parents of newborns with abnormal SCID screening results (N = 17). In addition, questionnaires about NBS for SCID were sent to 2000 parents of healthy newborns who either participated or declined participation in the SONNET-study that screened 140,593 newborns for SCID. RESULTS: Support for NBS for SCID was expressed by the majority of parents in questionnaires from both a public health perspective and a personal perspective. Parents emphasized the emotional impact of an abnormal screening result in interviews. (Long-term) stress and anxiety can be experienced during and after referral indicating the importance of uniform follow-up protocols and adequate information provision. CONCLUSION: The perspective of parents has led to several recommendations for NBS programs that are considering screening for SCID or other disorders. A close partnership of NBS programs' stakeholders, immunologists, geneticists, and pediatricians-immunologists in different countries is required for moving towards universal SCID screening for all infants.


Subject(s)
Health Plan Implementation , Neonatal Screening , Parents/psychology , Patient Acceptance of Health Care , Severe Combined Immunodeficiency/epidemiology , Health Plan Implementation/statistics & numerical data , Humans , Infant, Newborn , Neonatal Screening/methods , Neonatal Screening/psychology , Netherlands/epidemiology , Public Health Surveillance , Referral and Consultation , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/etiology , Stress, Psychological/diagnosis , Stress, Psychological/etiology , Surveys and Questionnaires
10.
Blood ; 134(4): 395-406, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31015188

ABSTRACT

Reduction of adenosine deaminase 2 (ADA2) activity due to autosomal-recessive loss-of-function mutations in the ADA2 gene (previously known as CECR1) results in a systemic vasculitis known as deficiency of ADA2 (DADA2). Neutrophils and a subset of neutrophils known as low-density granulocytes (LDGs) have been implicated in the pathogenesis of vasculitis, at least in part, through the formation of neutrophil extracellular traps (NETs). The study objective was to determine whether neutrophils and NETs play a pathogenic role in DADA2. In vivo evidence demonstrated NETs and macrophages in affected gastrointestinal tissue from patients with DADA2. An abundance of circulating LDGs prone to spontaneous NET formation was observed during active disease in DADA2 and were significantly reduced after remission induction by anti-tumor necrosis factor (TNF) therapy. Increased circulating LDGs were identified in unaffected family members with monoallelic ADA2 mutations. Adenosine triggered NET formation, particularly in neutrophils from female patients, by engaging A1 and A3 adenosine receptors (ARs) and through reactive oxygen species- and peptidylarginine deiminase-dependent pathways. Adenosine-induced NET formation was inhibited by recombinant ADA2, A1/A3 AR antagonists, or by an A2A agonist. M1 macrophages incubated with NETs derived from patients with DADA2 released significantly greater amounts of TNF-α. Treatment with an A2AAR agonist decreased nuclear translocation of NF-κB and subsequent production of inflammatory cytokines in DADA2 monocyte-derived macrophages. These results suggest that neutrophils may play a pathogenic role in DADA2. Modulation of adenosine-mediated NET formation may contribute a novel and directed therapeutic approach in the treatment of DADA2 and potentially other inflammatory diseases.


Subject(s)
Adenosine Deaminase/deficiency , Adenosine/metabolism , Agammaglobulinemia/etiology , Agammaglobulinemia/metabolism , Extracellular Traps/immunology , Extracellular Traps/metabolism , Intercellular Signaling Peptides and Proteins/deficiency , Severe Combined Immunodeficiency/etiology , Severe Combined Immunodeficiency/metabolism , Tumor Necrosis Factor-alpha/biosynthesis , Adenosine Deaminase/metabolism , Biomarkers/metabolism , Cytokines/metabolism , Enzyme Activation , Female , Humans , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Male , NF-kappa B/metabolism , Neutrophils/immunology , Neutrophils/metabolism , Receptors, Purinergic P1/metabolism , Sex Factors
11.
J Clin Immunol ; 40(8): 1124-1131, 2020 11.
Article in English | MEDLINE | ID: mdl-32880085

ABSTRACT

Clinical data from ADA-SCID patients registered in the U.S. Immunodeficiency Network (USIDNet) Repository were analyzed. Sixty-four ADA-SCID patients born between 1981 and 2017 had clinical data entered by their local (or home) enrolling institution. Median age at diagnosis was 1 month for those with a positive family history and 3 months for those without a prior family history, with some diagnosed at birth and one as late as 9 years of age. Overall survival was 79.7%, which increased to 94.1% since 2010. These patients had multiple infections and pulmonary, gastrointestinal, and neurological complications. The majority received enzyme replacement therapy (ERT) at some time, including 88% of those born since 2010. Twenty-six patients underwent allogeneic hematopoietic stem cell transplant (HSCT). HSCT successfully supported survival (17/26, 65%) using a variety of cell sources (bone marrow, mobilized peripheral blood, and cord blood) from sibling, family and unrelated donors. Nineteen patients underwent autologous HSCT with gene therapy (GT) using retroviral and lentiviral vectors and all are surviving. The prognosis for patients with ADA-SCID has continued to improve but these patients do have multiple early and potentially long-term conditions that require medical monitoring and management.


Subject(s)
Adenosine Deaminase/deficiency , Severe Combined Immunodeficiency/epidemiology , Severe Combined Immunodeficiency/etiology , Child , Child, Preschool , Disease Management , Disease Susceptibility , Female , Genetic Therapy , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/methods , Humans , Infant , Infant, Newborn , Infections/etiology , Male , Public Health Surveillance , Registries , Severe Combined Immunodeficiency/complications , United States/epidemiology
12.
Monaldi Arch Chest Dis ; 90(3)2020 Jul 29.
Article in English | MEDLINE | ID: mdl-32724227

ABSTRACT

B-cell immunity and immunoglobulins are less commonly affected in sarcoidosis. We aimed to evaluate immune status in sarcoidosis patients. Retrospective chart review of sarcoidosis patients attending a out-patient clinic over 3 months period. Immunoglobulins levels were recorded (A, M, G, E) along with clinical and serological data. They were divided in group A (normal IgG), group B (increased IgG), group C (decreased IgG) and group D (decreased IgG and IgM and/or IgA). Of 50 subjects, 68% were females and 62% of Caucasian origin. 22 (44%) had normal IgG levels, 16 (32%) had increased IgG levels, 10 (20%) had hypogammaglobulinemia and 2 (4%) had combined hypogammaglobulinemia, diagnosed with combined sarcoidosis and common variable immunodeficiency. Decreased IgA values was found in groups C and D. IgE was high in group B. Globulin was increased in group B and decreased in groups C and D. Decreased neutrophils were found in group D (all statistically significant). Correlation analysis showed significant association of angiotensin converting enzyme with IgA and IgM, inverse correlation of IgG with white blood cells and neutrophils, of IgA with globulin and inverse with albumin and of calcium with albumin. Most sarcoidosis patients have normal or increased immunoglobulin levels, that correlate with serum biomarkers of disease activity. Hypogammaglobulinemia may reflect treatment side effects and accompanied by blood leukocytosis. Combined severe immunodeficiency occurs in sarcoidosis.


Subject(s)
B-Lymphocytes/immunology , Immunoglobulins/blood , Sarcoidosis/complications , Sarcoidosis/immunology , Severe Combined Immunodeficiency/etiology , Adult , Agammaglobulinemia/diagnosis , Agammaglobulinemia/epidemiology , Agammaglobulinemia/etiology , Aged , Aged, 80 and over , Biomarkers/blood , Calcium/blood , Female , Humans , Incidence , Male , Middle Aged , Peptidyl-Dipeptidase A/immunology , Retrospective Studies , Sarcoidosis/diagnosis , Serum Albumin/immunology , Severe Combined Immunodeficiency/diagnosis
13.
J Clin Immunol ; 38(6): 727-732, 2018 08.
Article in English | MEDLINE | ID: mdl-30105620

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) is curative for severe combined immunodeficiency (SCID), but data on long-term impact of pre-HSCT chemotherapy, immune reconstitution and quality of life (QoL) of specific SCID genotypes are limited. We evaluated the long-term immune-reconstitution, health outcome and QoL in IL7Rα SCID, Artemis and RAG1 and 2 SCID survivors > 2 years post-HSCT in our center. Clinical data and immune reconstitution parameters were collated, and patients/families answered PedsQL generic core scale v4.0 questionnaires. Thirty-nine patients with a diagnosis of IL7Rα SCID (17 patients), Artemis SCID (8 patients) and RAG1/2 SCID (13 patients) had undergone HSCT with median age at last follow up for IL7Rα SCID, 14 years (range 4-27) and Artemis and RAG1/2 SCID, 10 years (range 2-18). Many patients have ongoing medical issues at latest follow-up [IL7Rα (73%), Artemis (85%), RAG1/2 (55%)]. Artemis SCID patients experienced more sequela than RAG1/2 SCID. Conditioned recipients with Artemis and RAG SCID had more CD4+ naïve lymphocytes compared to unconditioned recipients. All patients except those of IL7Rα SCID reported lower QoL; further subset group analysis showed parents and Artemis and RAG1/2 survivors without ongoing medical issues reported normal QoL. Conditioned recipients have superior long-term thymopoiesis, chimerism and immunoglobulin-independence. QoL was normal in those who did not have medical issues at long-term follow-up.


Subject(s)
DNA-Binding Proteins/deficiency , Endonucleases/deficiency , Homeodomain Proteins/genetics , Nuclear Proteins/deficiency , Quality of Life , Receptors, Interleukin-7/deficiency , Severe Combined Immunodeficiency/epidemiology , Severe Combined Immunodeficiency/etiology , Adolescent , Adult , Child , Child, Preschool , Disease Susceptibility , Female , Hematopoietic Stem Cell Transplantation , Humans , Longitudinal Studies , Male , Outcome Assessment, Health Care , Retrospective Studies , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/therapy , Time Factors , Young Adult
14.
J Clin Immunol ; 38(4): 527-536, 2018 05.
Article in English | MEDLINE | ID: mdl-29948574

ABSTRACT

PURPOSE: All reported patients with hypomorphic X-linked severe combined immunodeficiency (X-SCID) due to c.664C>T (p.R222C) mutations in the gene (IL2RG) encoding the common γ chain (γc) have presented with opportunistic infections within the first year of life, despite the presence of nearly normal NK and T cell numbers. Reporting five children of one extended family with hemizygous mutations in IL2RG, we explore potential diagnostic clues and extend our comprehension of the functional impact of this mutation. METHODS: Whole exome sequencing (WES); detailed immune phenotyping; cytokine-induced STAT phosphorylation; B, T, and NK cell activation; and quantification of sjTRECs in five Arab children with c.664C>T (p.R222C) IL2RG mutation. RESULTS: The mean age at clinical presentation with respiratory tract infection or diarrhea was 6.8 (range: 2-12) months. None of the children presented with opportunistic infections. Diagnostic clues were early onset in the first year of life, and a suggestive family history associated with reduced naïve CD4 T cells and absent switched memory B cells. Number and phenotype of NK cells and innate-like lymphocytes were normal. The diagnosis was made by WES and corroborated by absent STAT phosphorylation and reduced functional response after IL-2 and IL-21 stimulation. Four patients underwent successful hematopoietic stem cell transplantation. CONCLUSIONS: As early diagnosis and treatment are important, a high index of suspicion in the diagnosis of c.664C>T (p.R222C) X-SCID is needed. This requires prompt genetic testing by next generation sequencing in order to avoid unnecessary delays in the definite diagnosis since immunological work up may not be discriminating. Assays directly testing cytokine signaling or cytokine-dependent functions are helpful in confirming the functional impact of the identified hypomorphic variants.


Subject(s)
Interleukin Receptor Common gamma Subunit/genetics , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mutation , Severe Combined Immunodeficiency/diagnosis , Severe Combined Immunodeficiency/etiology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Adolescent , Adult , Biomarkers , Cell Differentiation , Child , Child, Preschool , Cytokines/metabolism , DNA Mutational Analysis , Female , Genetic Testing , Humans , Immunity, Humoral , Immunophenotyping , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Male , Pedigree , Signal Transduction , Young Adult
15.
Curr Opin Hematol ; 24(1): 16-22, 2017 01.
Article in English | MEDLINE | ID: mdl-27749373

ABSTRACT

PURPOSE OF REVIEW: Primary immunodeficiencies (PIDs) are inherited conditions where components of the immune system are missing or dysfunctional. Over 300 genes have been causally linked to monogenic forms of PID, including a number that regulate the actin cytoskeleton. The majority of cytoskeletal defects disrupt assembly and disassembly of filamentous actin in multiple immune cell lineages impacting functions such as cell migration and adhesion, pathogen uptake, intercellular communication, intracellular signalling, and cell division. RECENT FINDINGS: In the past 24 months, new actin defects have been identified through next generation sequencing technologies. Substantial progress has also been made in understanding the pathogenic mechanisms that contribute to immunological dysfunction, and also how the cytoskeleton participates in normal physiological immune processes. SUMMARY: This review summarises recent advances in the field, raising awareness of these conditions and our current understanding of their presentation. Description of further cases and new conditions will extend the clinical phenotype of actin-related disorders, and will promote the development of more effective and targeted therapies.


Subject(s)
Actin Cytoskeleton/metabolism , Immunologic Deficiency Syndromes/etiology , Immunologic Deficiency Syndromes/metabolism , Animals , Cell Adhesion , Cell Movement , GTPase-Activating Proteins , Guanine Nucleotide Exchange Factors/deficiency , Humans , Leukocytes/immunology , Leukocytes/metabolism , Microfilament Proteins/deficiency , Protein Binding , Protein Multimerization , Severe Combined Immunodeficiency/etiology , Severe Combined Immunodeficiency/metabolism , Signal Transduction , Trans-Activators/deficiency
16.
J Investig Allergol Clin Immunol ; 27(5): 299-304, 2017.
Article in English | MEDLINE | ID: mdl-28266921

ABSTRACT

BACKGROUND: Severe combined immunodeficiency (SCID) is a life-threatening pediatric disease. We report on the clinical evaluation, immunological assessment, molecular analysis, and outcomes of SCID patients in a tertiary referral center in Iran. METHODS: From January 2006 to December 2015, we performed a prospective cohort study in which initial screening and advanced immunological tests were carried out on patients suspected of having SCID. Genetic analysis was also performed to confirm the diagnosis. RESULTS: A total of 63 patients were diagnosed with SCID (43 male [68.3%]). The median age at onset and diagnosis and diagnostic delay were 40 and 110 and 60 days respectively. A total of 49 patients (77.8%) had a history of BCG vaccination, and of these, one-third experienced BCG-associated complications. The most common clinical manifestations were pneumonia, recurrent oral candidiasis, chronic diarrhea, and failure to thrive. Of the thirteen patients who underwent hematopoietic stem cell transplantation, 8 survived and 5 died before they could receive the transplant. Most patients (34.9%) were classified as having T-B-NK+ SCID and had a mutation in the RAG2 or RAG1 gene. CONCLUSIONS: Autosomal recessive SCID is the most common type in Iranian patients. Providing high-quality training to physicians and patients' families to reduce the diagnostic delay should be prioritized. It is also important to raise awareness of live vaccination and to expand stem cell donor registries to speed up the transplantation process.


Subject(s)
Severe Combined Immunodeficiency/diagnosis , Biomarkers , Disease Susceptibility , Female , Humans , Infant , Infant, Newborn , Male , Molecular Diagnostic Techniques , Mutation , Phenotype , Severe Combined Immunodeficiency/complications , Severe Combined Immunodeficiency/etiology , Severe Combined Immunodeficiency/therapy , Symptom Assessment
17.
Pediatr Dermatol ; 34(1): e24-e25, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27699886

ABSTRACT

Verrucae vulgaris in patients with severe combined immunodeficiency (SCID) after hematopoietic stem cell transplantation (HCST) can be challenging to manage. We describe two brothers with X-linked SCID who had severe, persistent verrucae that did not respond to traditional topical therapies, including liquid nitrogen, imiquimod, salicylic acid, sinecatechins, 40% urea, and 5-fluorourcil. Both brothers had full response to topical 3% cidofovir, which should be considered in recalcitrant warts in individuals with SCID after HSCT.


Subject(s)
Antiviral Agents/therapeutic use , Cytosine/analogs & derivatives , Hematopoietic Stem Cell Transplantation/adverse effects , Organophosphonates/therapeutic use , Severe Combined Immunodeficiency/etiology , Warts/drug therapy , Child , Cidofovir , Cytosine/therapeutic use , Humans , Male , Treatment Outcome , Warts/etiology
18.
J Allergy Clin Immunol ; 137(2): 517-526.e3, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26441229

ABSTRACT

BACKGROUND: Severe combined immunodeficiency (SCID) represents congenital disorders characterized by a deficiency of T cells caused by arrested development in the thymus. Yet the nature of these developmental blocks has remained elusive because of the difficulty of taking thymic biopsy specimens from affected children. OBJECTIVE: We sought to identify the stages of arrest in human T-cell development caused by various major types of SCID. METHODS: We performed transplantation of SCID CD34(+) bone marrow stem/progenitor cells into an optimized NSG xenograft mouse model, followed by detailed phenotypic and molecular characterization using flow cytometry, immunoglobulin and T-cell receptor spectratyping, and deep sequencing of immunoglobulin heavy chain (IGH) and T-cell receptor δ (TRD) loci. RESULTS: Arrests in T-cell development caused by mutations in IL-7 receptor α (IL7RA) and IL-2 receptor γ (IL2RG) were observed at the most immature thymocytes much earlier than expected based on gene expression profiling of human thymocyte subsets and studies with corresponding mouse mutants. T-cell receptor rearrangements were functionally required at the CD4(-)CD8(-)CD7(+)CD5(+) stage given the developmental block and extent of rearrangements in mice transplanted with Artemis-SCID cells. The xenograft model used is not informative for adenosine deaminase-SCID, whereas hypomorphic mutations lead to less severe arrests in development. CONCLUSION: Transplanting CD34(+) stem cells from patients with SCID into a xenograft mouse model provides previously unattainable insight into human T-cell development and functionally identifies the arrest in thymic development caused by several SCID mutations.


Subject(s)
Cell Differentiation , Severe Combined Immunodeficiency/etiology , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/metabolism , Animals , Antigens, Surface/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Differentiation/genetics , Cell Differentiation/immunology , Female , Gene Rearrangement , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Heterografts , Humans , Immunoglobulin Heavy Chains/genetics , Immunophenotyping , Lymphoid Progenitor Cells/cytology , Lymphoid Progenitor Cells/metabolism , Mice , Mice, Inbred NOD , Mice, Transgenic , Mutation , Phenotype , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Thymus Gland/embryology
SELECTION OF CITATIONS
SEARCH DETAIL