Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 125
Filtrar
1.
J Allergy Clin Immunol ; 153(1): 287-296, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37793572

RESUMEN

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.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave , Niño , Humanos , Inmunodeficiencia Combinada Grave/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Incidencia , Canadá/epidemiología , Estudios Retrospectivos , Acondicionamiento Pretrasplante
2.
J Allergy Clin Immunol ; 151(6): 1634-1645, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36638922

RESUMEN

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.


Asunto(s)
Infecciones por Enterovirus , Trasplante de Células Madre Hematopoyéticas , Hepatitis , Inmunodeficiencia Combinada Grave , Virosis , Humanos , Inmunodeficiencia Combinada Grave/terapia , Inmunodeficiencia Combinada Grave/etiología , Linfocitos T CD8-positivos , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Virosis/etiología , Hepatitis/etiología
3.
Immunol Res ; 70(5): 561-565, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-35661972

RESUMEN

Exposure to immunosuppressive medication in utero is an important cause of secondary T cell lymphopenia in infancy, which can be detected via T cell receptor excision circle (TREC) quantification on severe combined immunodeficiency (SCID) newborn screening (NBS). At present, there is a paucity of literature surrounding management of these infants. A protocol including recommendations for vaccinations and follow-up is needed to augment care. Patients referred to immunology for abnormal TREC results on NBS were identified as having in utero exposure to immunosuppressive medications and were followed until lymphopenia improved. The natural history of these patients' lymphopenia was used to develop general management guidelines. Four infants with low TRECs secondary to in utero immunosuppressive exposure were evaluated. Medication exposures included azathioprine, infliximab, hydroxychloroquine, and fingolimod. All infants were born full term. TRECs ranged from 101-206 (normal value in IL ≥ 250 at time of testing, B-actin control). T cell lymphopenia (CD3 < 1500) was present in 50% of cases. Undetectably low effector CD4 naïve T cell population was present in 100% of cases. Mitogen proliferation was uniformly normal. Severity of TREC abnormality did not correlate with presence of T cell lymphopenia. Immune abnormalities normalized in 75% patients by age 4 months. All age-appropriate vaccinations, including live vaccines, were administered to all patients by age 4 months. It is critical to assess for in utero immunosuppressive exposure in infants with abnormal TREC results on NBS. In the infants evaluated, secondary T cell lymphopenia associated with maternal immunosuppressive use resolved or significantly improved by age 4 months. Once abnormal TREC count is deemed to be secondary to in utero immunosuppression and there are no other contraindications, infants may safely receive live vaccination, are able to drink breast milk, and do not require prophylactic anti-microbials.


Asunto(s)
Linfopenia , Inmunodeficiencia Combinada Grave , Vacunas , Actinas , Azatioprina , Femenino , Clorhidrato de Fingolimod , Humanos , Hidroxicloroquina , Lactante , Recién Nacido , Infliximab , Linfopenia/diagnóstico , Mitógenos , Tamizaje Neonatal/métodos , Receptores de Antígenos de Linfocitos T/genética , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/etiología , Inmunodeficiencia Combinada Grave/terapia
4.
Immunol Invest ; 51(4): 739-747, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-33472463

RESUMEN

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.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Inmunodeficiencia Combinada Grave , Estudios de Seguimiento , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Inmunoglobulinas Intravenosas , Estudios Retrospectivos , Inmunodeficiencia Combinada Grave/etiología , Inmunodeficiencia Combinada Grave/terapia
5.
Immunobiology ; 226(6): 152143, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34598034

RESUMEN

We report a rare case of agranulocytosis and lymphopenia complicated with hemophagocytic lymphohistiocytosis. Diagnosis of reticular dysgenesis was made by detection of a pathogenic stop gain variant in the AK2 gene on targeted next generation sequencing and confirmed by Sanger sequencing. Parents were found to be carriers for this variant. Bone marrow aspirate and biopsy was also performed with a clinical diagnosis of severe combined immunodeficiency with HLH. However, no hemophagocytosis was noted in the bone marrow aspirate or trephine biopsy. Instead, it showed aggregates of large histiocyte-like cells, scattered erythroid precursors and megakaryocytes. These cells were confused to be some form of storage cells, but did not resemble storage cells seen in Gaucher's disease or Niemann Pick disease. Myeloid precursors were very few in number. Reticular dysgenesis was not suspected during admission due to a lack of awareness of this entity. Testing for sensorineural deafness in neonates with severe agranulocytosis and lymphopenia would facilitate an early diagnosis of reticular dysgenesis. To the best of our knowledge, hemophagocytic lymphohistiocytosis has not been previously reported in association with reticular dysgenesis.


Asunto(s)
Médula Ósea/patología , Histiocitos/patología , Leucopenia/diagnóstico , Leucopenia/etiología , Linfohistiocitosis Hemofagocítica/complicaciones , Linfohistiocitosis Hemofagocítica/diagnóstico , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/etiología , Adenilato Quinasa/genética , Biomarcadores , Biopsia , Células de la Médula Ósea/patología , Análisis Mutacional de ADN , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Pruebas Hematológicas , Humanos , Lactante , Masculino , Mutación , Fenotipo , Índice de Severidad de la Enfermedad
6.
Iran J Allergy Asthma Immunol ; 20(4): 402-412, 2021 Aug 07.
Artículo en Inglés | MEDLINE | ID: mdl-34418894

RESUMEN

T-cell receptor excision circles (TREC)/Kappa-deleting recombination excision circles (KREC) assay has been recently recognized for detecting patients with primary (T- and/or B-cell) immunodeficiency (PID). We aimed to investigate the alterations of these biomarkers in some combined immunodeficiency patients compared to the healthy controls in different age groups. TREC and KREC were assessed in a total of 82 PID patients, most of them with exact genetic diagnosis (3 months to 42 years); using quantitative real-time-polymerase chain reaction (PCR). Patients had a final diagnosis of common variable immunodeficiency (n=23), ataxia-telangiectasia (AT) (n=17), hyper-IgE syndrome (HIES) (7 with DOCK8 deficiency, 4 with signal transducer and activator of transcription 3 (STAT3) deficiency, and 8 children with unknown genetic defects), Wiskott-Aldrich syndrome (WAS) (n=20), purine nucleoside phosphorylase (PNP)deficiency(n=1), dedicator of cytokinesis2 (DOCK2) deficiency (n=1), recombinase activating gene1 (RAG1) deficiency (n=1). Very low to zero amounts of TREC and/or KREC were detected in 14 out of 23 cases of common variable immunodeficiency (CVID), 14 out of 17 cases of AT, 8 out of 20 cases of WAS, 6 out of 7 cases of DOCK8-deficiency patients, 4 out of 8 cases of HIES with unknown genetic defects and all patients with defects in DOCK2, PNP, and RAG1. STAT3-deficient patients were normal for both biomarkers. All patients showed a significant difference in both markers compared to age-matched healthy controls. Our findings highlight that apart from severe types of T/B cell defects, this assay can also be used for early diagnosis the patients with late-onset of disease and even PIDs without a positive family history.


Asunto(s)
Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Variación Genética , Cadenas kappa de Inmunoglobulina/genética , Enfermedades de Inmunodeficiencia Primaria/etiología , Receptores de Antígenos de Linfocitos T/genética , Inmunodeficiencia Combinada Grave/etiología , Alelos , Estudios de Casos y Controles , Diagnóstico Diferencial , Humanos , Fenotipo , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Inmunodeficiencia Combinada Grave/diagnóstico
7.
Eur J Immunol ; 51(5): 1028-1038, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33682138

RESUMEN

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.


Asunto(s)
Proteínas de Unión al ADN/genética , Predisposición Genética a la Enfermedad , Proteínas de Homeodominio/genética , Proteínas Nucleares/genética , Inmunodeficiencia Combinada Grave/etiología , Inmunodeficiencia Combinada Grave/terapia , Recombinación V(D)J/genética , Animales , Diagnóstico Diferencial , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Terapia Genética , Genotipo , Humanos , Mutación , Fenotipo , Inmunodeficiencia Combinada Grave/diagnóstico
8.
J Clin Immunol ; 41(4): 756-768, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33464451

RESUMEN

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.


Asunto(s)
Factores de Transcripción Forkhead/genética , Heterocigoto , Homocigoto , Mutación , Fenotipo , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/etiología , Línea Celular , Preescolar , Análisis Mutacional de ADN , Manejo de la Enfermedad , Femenino , Factores de Transcripción Forkhead/química , Estudios de Asociación Genética , Sitios Genéticos , Predisposición Genética a la Enfermedad , Trasplante de Células Madre Hematopoyéticas , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Masculino , Modelos Moleculares , Conformación Molecular , Linaje , Inmunodeficiencia Combinada Grave/terapia , Relación Estructura-Actividad , Resultado del Tratamiento
9.
J Clin Immunol ; 41(3): 631-638, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33411152

RESUMEN

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.


Asunto(s)
Fenotipo , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/epidemiología , Alelos , Biomarcadores , Consanguinidad , Estudios Transversales , Diagnóstico Diferencial , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Predisposición Genética a la Enfermedad , Genotipo , Humanos , Patrón de Herencia , Marruecos/epidemiología , Vigilancia en Salud Pública , Inmunodeficiencia Combinada Grave/etiología
10.
J Clin Immunol ; 41(3): 621-628, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33415666

RESUMEN

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.


Asunto(s)
Tamizaje Neonatal/métodos , Enfermedades de Inmunodeficiencia Primaria/epidemiología , Enfermedades de Inmunodeficiencia Primaria/etiología , Receptores de Antígenos de Linfocitos T/genética , Verrugas/epidemiología , Verrugas/etiología , Biomarcadores , Análisis Mutacional de ADN , Diagnóstico Diferencial , Susceptibilidad a Enfermedades , Femenino , Humanos , Recién Nacido , Masculino , Mutación , Fenotipo , Enfermedades de Inmunodeficiencia Primaria/diagnóstico , Receptores CXCR4/genética , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/epidemiología , Inmunodeficiencia Combinada Grave/etiología , Verrugas/diagnóstico
11.
J Clin Immunol ; 41(3): 595-602, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33409868

RESUMEN

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.


Asunto(s)
Tamizaje Neonatal , Inmunodeficiencia Combinada Grave/epidemiología , Algoritmos , Toma de Decisiones Clínicas , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Historia del Siglo XXI , Humanos , Recién Nacido , Fenotipo , Vigilancia en Salud Pública , Derivación y Consulta , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/etiología , Inmunodeficiencia Combinada Grave/historia
13.
J Clin Immunol ; 41(1): 99-108, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33070266

RESUMEN

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.


Asunto(s)
Implementación de Plan de Salud , Tamizaje Neonatal , Padres/psicología , Aceptación de la Atención de Salud , Inmunodeficiencia Combinada Grave/epidemiología , Implementación de Plan de Salud/estadística & datos numéricos , Humanos , Recién Nacido , Tamizaje Neonatal/métodos , Tamizaje Neonatal/psicología , Países Bajos/epidemiología , Vigilancia en Salud Pública , Derivación y Consulta , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/etiología , Estrés Psicológico/diagnóstico , Estrés Psicológico/etiología , Encuestas y Cuestionarios
15.
J Clin Immunol ; 40(8): 1124-1131, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32880085

RESUMEN

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.


Asunto(s)
Adenosina Desaminasa/deficiencia , Inmunodeficiencia Combinada Grave/epidemiología , Inmunodeficiencia Combinada Grave/etiología , Niño , Preescolar , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Femenino , Terapia Genética , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Trasplante de Células Madre Hematopoyéticas/métodos , Humanos , Lactante , Recién Nacido , Infecciones/etiología , Masculino , Vigilancia en Salud Pública , Sistema de Registros , Inmunodeficiencia Combinada Grave/complicaciones , Estados Unidos/epidemiología
16.
Monaldi Arch Chest Dis ; 90(3)2020 Jul 29.
Artículo en Inglés | MEDLINE | ID: mdl-32724227

RESUMEN

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.


Asunto(s)
Linfocitos B/inmunología , Inmunoglobulinas/sangre , Sarcoidosis/complicaciones , Sarcoidosis/inmunología , Inmunodeficiencia Combinada Grave/etiología , Adulto , Agammaglobulinemia/diagnóstico , Agammaglobulinemia/epidemiología , Agammaglobulinemia/etiología , Anciano , Anciano de 80 o más Años , Biomarcadores/sangre , Calcio/sangre , Femenino , Humanos , Incidencia , Masculino , Persona de Mediana Edad , Peptidil-Dipeptidasa A/inmunología , Estudios Retrospectivos , Sarcoidosis/diagnóstico , Albúmina Sérica/inmunología , Inmunodeficiencia Combinada Grave/diagnóstico
17.
Immunobiology ; 225(3): 151961, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32517885

RESUMEN

BACKGROUND AND OBJECTIVE: Severe combined immunodeficiency disease (SCID) is a rare inherited severe immunodeficiency, in which functions of T cells and B cells are impaired. SCID is inherited either in X-linked recessive, or autosomal recessive forms, and is either radiosensitive or radioresistant. Artemis (DCLRE1C gene), DNA ligase IV, DNA-PKC, and Cernunnos/XLF proteins are regarded as NHEJ (Non-Homologous End-Joining) proteins that are involved in the repair process of double-strand DNA breaks and their mutations would lead to cellular radiosensitivity. Diagnostic radiosensitivity assays are important for the management of clinical BMT (Bone Marrow Transplantation) conditions, such as what conditioning agents and doses should be used. MATERIALS AND METHODS: In this study, five SCID patients and healthy controls were examined. Skin fibroblasts were cultured. After X-irradiation, cells either underwent clonogenic assay or incubated to allow DNA repair and examined by the alkaline comet assay. Finally, DCLRE1C, RAG-1, and RAG-2 genes sequenced. RESULTS: By clonogenic assay, three patients were detected as radiosensitive with possible mutations in NHEJ genes such as DCLRE1C gene. The percentage of DNA in the tail measured by comet assay, in all three patients, was significantly different from the two other patients and the control group (p-value < 0.05). By using Sanger sequencing, a mutation in DCLRE1C gene was detected in one of the radiosensitive patients and two mutations in RAG-1, and RAG-2 genes were detected in the two radioresistant patients. CONCLUSION: Our findings suggest that comet assay is a fast technique for the diagnosis of the radiosensitive form of SCID and is very suitable for the timely diagnosis of RS-SCID before BMT.


Asunto(s)
Trasplante de Médula Ósea , Ensayo Cometa , Inmunodeficiencia Combinada Grave/diagnóstico , Inmunodeficiencia Combinada Grave/terapia , Linfocitos B/inmunología , Linfocitos B/metabolismo , Biomarcadores , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Humanos , Mutación , Tolerancia a Radiación , Inmunodeficiencia Combinada Grave/etiología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Acondicionamiento Pretrasplante
18.
Immunol Allergy Clin North Am ; 39(4): 535-546, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31563187

RESUMEN

Newborn screening for severe combined immunodeficiency has been implemented in all 50 states. This screening identifies newborns with T-cell lymphopenia. After an abnormal screening, additional testing is needed to determine if the child has severe combined immunodeficiency. Because screening programs vary, it is imperative for the clinical immunologist to understand how screening is done in their state and to prepare an effective assessment protocol for the management of these patients. Part of this assessment should include training and helping to ensure the effective delivery of this news to the family, a skill neither intuitive nor classically taught to immunologists.


Asunto(s)
Inmunodeficiencia Combinada Grave/diagnóstico , Biomarcadores , Toma de Decisiones Clínicas , Manejo de la Enfermedad , Humanos , Recién Nacido , Tamizaje Neonatal/métodos , Tamizaje Neonatal/normas , Inmunodeficiencia Combinada Grave/epidemiología , Inmunodeficiencia Combinada Grave/etiología , Estados Unidos/epidemiología
19.
Curr Opin Immunol ; 60: 148-155, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31302571

RESUMEN

Environmental factors modify disease presentation and severity in allergic disorders. Primary atopic disorders (PADs) are a heterogenous group of single gene disorders that lead to significant atopic and allergic disease manifestations. However, a number of these monogenic diseases have variable penetrance suggesting that gene-gene and/or gene-environment interactions could modulate the clinical phenotype. Environmental factors such as diet, the microbiome at the epithelial-environment interface, the presence and/or extent of infection, and psychologic stress can alter disease phenotypic expression of allergic diseases, and PADs provide discrete contexts in which to understand these influences. We outline how gene-environment interactions likely contribute to a variable penetrance and expressivity in PADs. Dietary modifications of both macronutrients and/or micronutrients alter T-cell metabolism and may influence effector T-cell function. The mucosal microbiome may affect local inflammation and may remotely influence regulatory elements, while psychologic stress can affect mast cell and other allergic effector cell function. Understanding gene-environment interactions in PADs can hopefully provide a foundation for interrogating gene-environment interactions to common allergic disorders, and also present opportunities for personalized interventions based on the altered pathways and environmental influences in affected individuals.


Asunto(s)
Susceptibilidad a Enfermedades , Exposición a Riesgos Ambientales/efectos adversos , Interacción Gen-Ambiente , Hipersensibilidad Inmediata/etiología , Dermatitis Atópica/etiología , Dermatitis Atópica/metabolismo , Metabolismo Energético , Ambiente , Humanos , Hipersensibilidad Inmediata/metabolismo , Mastocitos/inmunología , Mastocitos/metabolismo , Microbiota , Estado Nutricional , Fenotipo , Inmunodeficiencia Combinada Grave/etiología , Inmunodeficiencia Combinada Grave/metabolismo , Estrés Fisiológico , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA