Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 1.553
Filtrer
1.
J Clin Immunol ; 44(7): 154, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38896123

RÉSUMÉ

Patients with chromosome 18q deletion syndrome generally experience hypogammaglobulinemia. Herein, we describe two patients with chromosome 18q deletion syndrome who presented with late-onset combined immune deficiency (LOCID), which has not been previously reported. Patient 1 was a 29-year-old male with 18q deletion syndrome, who was being managed for severe motor and intellectual disabilities at the Yamabiko Medical Welfare Center for 26 years. Although the patient had few infections, he developed Pneumocystis pneumonia at the age of 28. Patient 2, a 48-year-old female with intellectual disability and congenital malformations, was referred to Tokyo Medical and Dental University Hospital with abnormal bilateral lung shadows detected on her chest radiography. Computed tomography showed multiple lymphadenopathies and pneumonia. A lymph node biopsy of the inguinal region revealed granulomatous lymphadenitis, and a chromosomal examination revealed 18q deletion. Array-based genomic hybridization analysis revealed deletion at 18q21.32-q22.3 for patient 1 and at 18q21.33-qter for patient 2. Immune status work-up of the two patients revealed panhypogammaglobulinemia, decreased number of memory B cells and naïve CD4+ and/or CD8+ cells, reduced response on the carboxyfluorescein diacetate succinimidyl ester T-cell division test, and low levels of T-cell receptor recombination excision circles and Ig κ-deleting recombination excision circles. Consequently, both patients were diagnosed with LOCID. Although patients with 18q deletion syndrome generally experience humoral immunodeficiency, the disease can be further complicated by cell-mediated immunodeficiency, causing combined immunodeficiency. Therefore, patients with 18q deletion syndrome should be regularly tested for cellular/humoral immunocompetence.


Sujet(s)
Délétion de segment de chromosome , Maladies chromosomiques , Chromosomes humains de la paire 18 , Humains , Mâle , Femelle , Chromosomes humains de la paire 18/génétique , Maladies chromosomiques/diagnostic , Maladies chromosomiques/génétique , Adulte , Adulte d'âge moyen , Âge de début , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/complications , Déficience intellectuelle/génétique , Déficience intellectuelle/diagnostic , Déficience intellectuelle/étiologie , Déficits immunitaires/génétique , Déficits immunitaires/diagnostic , Déficits immunitaires/complications
2.
J Clin Immunol ; 44(7): 151, 2024 Jun 19.
Article de Anglais | MEDLINE | ID: mdl-38896336

RÉSUMÉ

A cell's ability to survive and to evade cancer is contingent on its ability to retain genomic integrity, which can be seriously compromised when nucleic acid phosphodiester bonds are disrupted. DNA Ligase 1 (LIG1) plays a key role in genome maintenance by sealing single-stranded nicks that are produced during DNA replication and repair. Autosomal recessive mutations in a limited number of individuals have been previously described for this gene. Here we report a homozygous LIG1 mutation (p.A624T), affecting a universally conserved residue, in a patient presenting with leukopenia, neutropenia, lymphopenia, pan-hypogammaglobulinemia, and diminished in vitro response to mitogen stimulation. Patient fibroblasts expressed normal levels of LIG1 protein but exhibited impaired growth, poor viability, high baseline levels of gamma-H2AX foci, and an enhanced susceptibility to DNA-damaging agents. The mutation reduced LIG1 activity by lowering its affinity for magnesium 2.5-fold. Remarkably, it also increased LIG1 fidelity > 50-fold against 3' end 8-Oxoguanine mismatches, exhibiting a marked reduction in its ability to process such nicks. This is expected to yield increased ss- and dsDNA breaks. Molecular dynamic simulations, and Residue Interaction Network studies, predicted an allosteric effect for this mutation on the protein loops associated with the LIG1 high-fidelity magnesium, as well as on DNA binding within the adenylation domain. These dual alterations of suppressed activity and enhanced fidelity, arising from a single mutation, underscore the mechanistic picture of how a LIG1 defect can lead to severe immunological disease.


Sujet(s)
DNA ligase ATP , Homozygote , Mutation , Immunodéficience combinée grave , Femelle , Humains , Mâle , DNA ligase ATP/génétique , DNA ligase ATP/métabolisme , Fibroblastes , Simulation de dynamique moléculaire , Mutation/génétique , Immunodéficience combinée grave/génétique , Nourrisson
3.
Pediatr Allergy Immunol ; 35(6): e14171, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38860449

RÉSUMÉ

BACKGROUND: Artemis deficiency is an autosomal recessive disorder characterized by a combined immunodeficiency with increased cellular radiosensitivity. In this review, the clinical and genetic characteristics of 15 patients with DCLRE1C variants are presented. METHODS: The demographic, clinical, immunologic, and genetic characteristics of patients with confirmed DCLRE1C variants diagnosed between 2013 and 2023 were collected retrospectively. Three patients were evaluated for radiosensitivity by the Comet assay, compared with age- and sex-matched healthy control. RESULTS: Seven patients who had severe infections in the first 6 months of life were diagnosed with T-B-NK+ SCID (severe combined immunodeficiency). Among them, four individuals underwent transplantation, and one of those died due to post-transplant complications in early life. Eight patients had hypomorphic variants. Half of them were awaiting a suitable donor, while the other half had already undergone transplantation. The majority of patients were born into a consanguineous family (93.3%). Most patients had recurrent sinopulmonary infections (73.3%), and one patient had no other infection than an acute respiratory infection before diagnosis. Two patients (13.3%) had autoimmunity in the form of autoimmune hemolytic anemia. Growth retardation was observed in only one patient (6.6%), and no malignancy was detected in the surviving 11 patients during the median (IQR) of 21.5 (12-45) months of follow-up. Three patients who had novel variants exhibited increased radiosensitivity and compromised DNA repair, providing a potential vulnerability to malignant transformation. CONCLUSION: Early diagnosis, radiation avoidance, and careful preparation for transplantation contribute to minimizing complications, enhancing life expectancy, and improving the patient's quality of life.


Sujet(s)
Protéines de liaison à l'ADN , Radiotolérance , Immunodéficience combinée grave , Humains , Radiotolérance/génétique , Mâle , Femelle , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/thérapie , Nourrisson , Protéines de liaison à l'ADN/génétique , Enfant d'âge préscolaire , Études rétrospectives , Endonucleases/génétique , Protéines nucléaires/génétique , Enfant , Études de cohortes
4.
PLoS One ; 19(6): e0306329, 2024.
Article de Anglais | MEDLINE | ID: mdl-38941330

RÉSUMÉ

BACKGROUND: Many newborn screening programs worldwide have introduced screening for diseases using DNA extracted from dried blood spots (DBS). In Germany, DNA-based assays are currently used to screen for severe combined immunodeficiency (SCID), spinal muscular atrophy (SMA), and sickle cell disease (SCD). METHODS: This study analysed the impact of pre-analytic DNA carry-over in sample preparation on the outcome of DNA-based newborn screening for SCID and SMA and compared the efficacy of rapid extraction versus automated protocols. Additionally, the distribution of T cell receptor excision circles (TREC) on DBS cards, commonly used for routine newborn screening, was determined. RESULTS: Contaminations from the punching procedure were detected in the SCID and SMA assays in all experimental setups tested. However, a careful evaluation of a cut-off allowed for a clear separation of true positive polymerase chain reaction (PCR) amplifications. Our rapid in-house extraction protocol produced similar amounts compared to automated commercial systems. Therefore, it can be used for reliable DNA-based screening. Additionally, the amount of extracted DNA significantly differs depending on the location of punching within a DBS. CONCLUSIONS: Newborn screening for SMA and SCID can be performed reliably. It is crucial to ensure that affected newborns are not overlooked. Therefore a carefully consideration of potential contaminating factors and the definition of appropriate cut-offs to minimise the risk of false results are of special concern. It is also important to note that the location of punching plays a pivotal role, and therefore an exact quantification of TREC numbers per µl may not be reliable and should therefore be avoided.


Sujet(s)
ADN , Amyotrophie spinale , Dépistage néonatal , Immunodéficience combinée grave , Humains , Dépistage néonatal/méthodes , Nouveau-né , Amyotrophie spinale/diagnostic , Amyotrophie spinale/génétique , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/génétique , ADN/génétique , ADN/sang , ADN/analyse , Dépistage sur goutte de sang séché/méthodes , Tests de criblage à haut débit/méthodes , Réaction de polymérisation en chaîne/méthodes
5.
J Clin Immunol ; 44(5): 117, 2024 May 17.
Article de Anglais | MEDLINE | ID: mdl-38758229

RÉSUMÉ

AIOLOS, a vital member of the IKAROS protein family, plays a significant role in lymphocyte development and function through DNA binding and protein-protein interactions. Mutations in the IKZF3 gene, which encodes AIOLOS, lead to a rare combined immunodeficiency often linked with infections and malignancy. In this study, we evaluated a 1-year-4-month-old female patient presenting with recurrent infections, diarrhea, and failure to thrive. Laboratory investigations revealed decreased T lymphocyte and immunoglobulin levels. Through whole-exome and Sanger sequencing, we discovered a de novo mutation in IKZF3 (NM_012481; exon 5 c.571G > C, p.Gly191Arg), corresponding to the third DNA-binding zinc finger region of the encoded protein AIOLOS. Notably, the patient with the AIOLOS G191R mutation showed reduced recent thymic emigrants in naïve CD4+T cells compared to healthy counterparts of the same age, while maintaining normal levels of Th1, Th2, Th17, Treg, and Tfh cells. This mutation also resulted in decreased switched memory B cells and lower CD23 and IgM expression. In vitro studies revealed that AIOLOS G191R does not impact the expression of AIOLOS but compromises its stability, DNA binding and pericentromeric targeting. Furthermore, AIOLOS G191R demonstrated a dominant-negative effect over the wild-type protein. This case represents the first reported instance of a mutation in the third DNA-binding zinc finger region of AIOLOS highlighting its pivotal role in immune cell functionality.


Sujet(s)
Facteur de transcription Ikaros , Mutation , Humains , Facteur de transcription Ikaros/génétique , Femelle , Mutation/génétique , Nourrisson , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/diagnostic , , Lymphocytes B/immunologie
6.
Sci Immunol ; 9(95): eade5705, 2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38787962

RÉSUMÉ

Inborn errors of T cell development present a pediatric emergency in which timely curative therapy is informed by molecular diagnosis. In 11 affected patients across four consanguineous kindreds, we detected homozygosity for a single deleterious missense variant in the gene NudC domain-containing 3 (NUDCD3). Two infants had severe combined immunodeficiency with the complete absence of T and B cells (T -B- SCID), whereas nine showed classical features of Omenn syndrome (OS). Restricted antigen receptor gene usage by residual T lymphocytes suggested impaired V(D)J recombination. Patient cells showed reduced expression of NUDCD3 protein and diminished ability to support RAG-mediated recombination in vitro, which was associated with pathologic sequestration of RAG1 in the nucleoli. Although impaired V(D)J recombination in a mouse model bearing the homologous variant led to milder immunologic abnormalities, NUDCD3 is absolutely required for healthy T and B cell development in humans.


Sujet(s)
Immunodéficience combinée grave , Recombinaison V(D)J , Humains , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/immunologie , Animaux , Souris , Recombinaison V(D)J/immunologie , Recombinaison V(D)J/génétique , Mâle , Femelle , Nourrisson , Lymphocytes B/immunologie , Protéines à homéodomaine/génétique , Protéines à homéodomaine/immunologie , Lymphocytes T/immunologie , Enfant d'âge préscolaire , Mutation faux-sens
7.
J Clin Immunol ; 44(4): 98, 2024 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-38598033

RÉSUMÉ

Biallelic null or hypomorphic variants in JAK3 cause SCID and less frequently Omenn syndrome. We investigated homozygous hypomorphic JAK3 mutations in two patients, and expression and function of a novel JAK3R431P variant in Omenn syndrome. Immunophenotyping of PBMC from the patient with the novel JAK3R431P variant was undertaken, by flow cytometry and Phosflow after stimulation with IL-2, IL-7, and IL-15. JAK3 expression was investigated by Western blotting. We report two patients with homozygous hypomorphic JAK3 variants and clinical features of Omenn syndrome. One patient had a previously described JAK3R775H variant, and the second had a novel JAK3R431P variant. One patient with a novel JAK3R431P variant had normal expression of JAK3 in immortalised EBV-LCL cells but reduced phosphorylation of STAT5 after stimulation with IL-2, IL-7, and IL-15 consistent with impaired kinase activity. These results suggest the JAK3R431P variant to be hypomorphic. Both patients are alive and well after allogeneic haematopoietic stem cell transplantation. They have full donor chimerism, restitution of thymopoiesis and development of appropriate antibody responses following vaccination. We expand the phenotype of hypomorphic JAK3 deficiency and demonstrate the importance of functional testing of novel variants in disease-causing genes.


Sujet(s)
Janus kinase 3 , Immunodéficience combinée grave , Humains , Nourrisson , Interleukine-15 , Interleukine-2 , Interleukine-7 , Janus kinase 3/génétique , Agranulocytes , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/thérapie
8.
J Clin Immunol ; 44(4): 93, 2024 Apr 05.
Article de Anglais | MEDLINE | ID: mdl-38578360

RÉSUMÉ

Newborn screening (NBS) for severe inborn errors of immunity (IEI), affecting T lymphocytes, and implementing measurements of T cell receptor excision circles (TREC) has been shown to be effective in early diagnosis and improved prognosis of patients with these genetic disorders. Few studies conducted on smaller groups of newborns report results of NBS that also include measurement of kappa-deleting recombination excision circles (KREC) for IEI affecting B lymphocytes. A pilot NBS study utilizing TREC/KREC detection was conducted on 202,908 infants born in 8 regions of Russia over a 14-month period. One hundred thirty-four newborns (0.66‰) were NBS positive after the first test and subsequent retest, 41% of whom were born preterm. After lymphocyte subsets were assessed via flow cytometry, samples of 18 infants (0.09‰) were sent for whole exome sequencing. Confirmed genetic defects were consistent with autosomal recessive agammaglobulinemia in 1/18, severe combined immunodeficiency - in 7/18, 22q11.2DS syndrome - in 4/18, combined immunodeficiency - in 1/18 and trisomy 21 syndrome - in 1/18. Two patients in whom no genetic defect was found met criteria of (severe) combined immunodeficiency with syndromic features. Three patients appeared to have transient lymphopenia. Our findings demonstrate the value of implementing combined TREC/KREC NBS screening and inform the development of policies and guidelines for its integration into routine newborn screening programs.


Sujet(s)
Lymphopénie , Immunodéficience combinée grave , Nourrisson , Nouveau-né , Humains , Dépistage néonatal/méthodes , Projets pilotes , Lymphopénie/diagnostic , Lymphocytes T , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/génétique , ADN , Récepteurs aux antigènes des cellules T/génétique
9.
Nat Commun ; 15(1): 3662, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38688902

RÉSUMÉ

Hematopoietic stem cell gene therapy (GT) using a γ-retroviral vector (γ-RV) is an effective treatment for Severe Combined Immunodeficiency due to Adenosine Deaminase deficiency. Here, we describe a case of GT-related T-cell acute lymphoblastic leukemia (T-ALL) that developed 4.7 years after treatment. The patient underwent chemotherapy and haploidentical transplantation and is currently in remission. Blast cells contain a single vector insertion activating the LIM-only protein 2 (LMO2) proto-oncogene, confirmed by physical interaction, and low Adenosine Deaminase (ADA) activity resulting from methylation of viral promoter. The insertion is detected years before T-ALL in multiple lineages, suggesting that further hits occurred in a thymic progenitor. Blast cells contain known and novel somatic mutations as well as germline mutations which may have contributed to transformation. Before T-ALL onset, the insertion profile is similar to those of other ADA-deficient patients. The limited incidence of vector-related adverse events in ADA-deficiency compared to other γ-RV GT trials could be explained by differences in transgenes, background disease and patient's specific factors.


Sujet(s)
Adenosine deaminase , Agammaglobulinémie , Thérapie génétique , Vecteurs génétiques , Transplantation de cellules souches hématopoïétiques , Leucémie-lymphome lymphoblastique à précurseurs T , Proto-oncogène Mas , Immunodéficience combinée grave , Humains , Adenosine deaminase/déficit , Adenosine deaminase/génétique , Thérapie génétique/méthodes , Leucémie-lymphome lymphoblastique à précurseurs T/thérapie , Leucémie-lymphome lymphoblastique à précurseurs T/génétique , Immunodéficience combinée grave/thérapie , Immunodéficience combinée grave/génétique , Vecteurs génétiques/génétique , Agammaglobulinémie/thérapie , Agammaglobulinémie/génétique , Mâle , Retroviridae/génétique
10.
J Allergy Clin Immunol Pract ; 12(6): 1622-1630.e4, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38636590

RÉSUMÉ

BACKGROUND: Inborn errors of immunity (IEIs) include 485 inherited disorders characterized by an increased susceptibility to life-threatening infectious diseases, autoimmunity, and malignant diseases with a high mortality rate in the first years of life. Severe combined immunodeficiency is the most severe of the IEIs, and its detection should be a primary goal in a newborn screening (NBS) program. The term "actionable" has recently been used for all IEIs with outcomes that can be demonstrably improved through early specialized intervention. OBJECTIVE: To evaluate the results of the expanded NBS strategy for IEIs in Tuscany Region (Italy), based on T-cell receptor excision circle, kappa recombining excision circle, and tandem mass-based assays. METHODS: This is a retrospective study collecting data from all infants born in Tuscany from October 10, 2018, to October 10, 2022. Tandem mass assay to identify adenosine deaminase and purine nucleoside phosphorylase deficiency, together with T-cell receptor excision circle and kappa recombining excision circle molecular analysis, was conducted on dried blood spot from the newborns' Guthrie Cards. A new dried blood spot and evaluation by an immunologist were carried out when the results of the first test were outside the diagnostic cutoffs. RESULTS: A total of 94,319 newborns were evaluated. Referral rates for T-cell recombining excision circles (0.031%) and kappa recombining excision circles (0.074%) in this study are in line with the data available in literature. The results from the expanded NBS strategy revealed an incidence rate of 1 per 9431 affected newborns. CONCLUSIONS: This work represents the first description of a sustainable and real-life-based expanded NBS program for IEIs with a high diagnostic incidence facilitating prompt management of identified patients.


Sujet(s)
Dépistage néonatal , Humains , Nouveau-né , Italie/épidémiologie , Études rétrospectives , Mâle , Femelle , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/épidémiologie , Immunodéficience combinée grave/immunologie , Immunodéficience combinée grave/génétique , Récepteurs aux antigènes des cellules T/génétique , Récepteurs aux antigènes des cellules T/immunologie
11.
Am J Hum Genet ; 111(4): 791-804, 2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38503300

RÉSUMÉ

Mutations in proteasome ß-subunits or their chaperone and regulatory proteins are associated with proteasome-associated autoinflammatory disorders (PRAAS). We studied six unrelated infants with three de novo heterozygous missense variants in PSMB10, encoding the proteasome ß2i-subunit. Individuals presented with T-B-NK± severe combined immunodeficiency (SCID) and clinical features suggestive of Omenn syndrome, including diarrhea, alopecia, and desquamating erythematous rash. Remaining T cells had limited T cell receptor repertoires, a skewed memory phenotype, and an elevated CD4/CD8 ratio. Bone marrow examination indicated severely impaired B cell maturation with limited V(D)J recombination. All infants received an allogeneic stem cell transplant and exhibited a variety of severe inflammatory complications thereafter, with 2 peri-transplant and 2 delayed deaths. The single long-term transplant survivor showed evidence for genetic rescue through revertant mosaicism overlapping the affected PSMB10 locus. The identified variants (c.166G>C [p.Asp56His] and c.601G>A/c.601G>C [p.Gly201Arg]) were predicted in silico to profoundly disrupt 20S immunoproteasome structure through impaired ß-ring/ß-ring interaction. Our identification of PSMB10 mutations as a cause of SCID-Omenn syndrome reinforces the connection between PRAAS-related diseases and SCID.


Sujet(s)
Immunodéficience combinée grave , Nourrisson , Humains , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/métabolisme , Proteasome endopeptidase complex/génétique , Proteasome endopeptidase complex/métabolisme , Mutation/génétique , Lymphocytes T/métabolisme , Mutation faux-sens/génétique
12.
J Clin Immunol ; 44(3): 73, 2024 Mar 01.
Article de Anglais | MEDLINE | ID: mdl-38424321

RÉSUMÉ

PURPOSE: Major histocompatibility complex (MHC) class II deficiency is one of the combined immune deficiency disorders caused by defects in the MHC class II regulatory genes leading to abnormal T cells development and function. Therefore, patients mainly present with increased susceptibility to infections, diarrhea, and failure to thrive. In this report, we present one MHC class II deficient patient with a novel presentation with Hemophagocytic Lymphohistiocytosis (HLH). METHODS: Immunophenotyping of lymphocyte subpopulations and HLA-DR expression was assess by flow cytometry. Gene mutational analysis was performed by whole exome and Sanger sequencing. RESULTS: We reported a 7-year-old girl, who was diagnosed at age of 2 years with MHC class II deficiency by genetic testing and flow cytometry. Two years later, she developed disseminated BCGitis which was treated with proper antimicrobial agents. At the age of 7 years, she presented with clinical features fulfilling 6 diagnostic criteria of HLH including evidence of hemophagocytic activity in bone marrow aspiration. Accordingly, the diagnosis of HLH was established and the patient was started on IV Dexamethasone, Anakinra and IVIG. Eventually, patient started to improve and was discharged in good condition. Few months later, the patient was readmitted with severe pneumonia and sepsis leading to death. CONCLUSION: Patients with MHC class II deficiency might present with disseminated BCGitis especially if the patient has severe T cell lymphopenia. Additionally, this immune defect might be added to the list of inborn errors of immunity that can be complicated with HLH.


Sujet(s)
Lymphohistiocytose hémophagocytaire , Immunodéficience combinée grave , Enfant , Femelle , Humains , Dépistage génétique , Antigènes d'histocompatibilité de classe II/génétique , Lymphohistiocytose hémophagocytaire/étiologie , Lymphohistiocytose hémophagocytaire/génétique , Complexe majeur d'histocompatibilité , Immunodéficience combinée grave/génétique
13.
Mol Biol Rep ; 51(1): 302, 2024 Feb 14.
Article de Anglais | MEDLINE | ID: mdl-38355773

RÉSUMÉ

BACKGROUND: Severe Combined Immunodeficiency (SCID) is an autosomal recessive inborn error of immunity (IEI) characterized by recurrent chest and gastrointestinal (GI) infections and in some cases associated with life-threatening disorders. METHODOLOGY AND RESULTS: This current study aims to unwind the molecular etiology of SCID and also extended the patients' phenotype associated with identified particular variants. Herein, we present 06 disease-causing variants identified in 07 SCID-patients in three different SCID related genes. Whole Exome Sequencing (WES) followed by Sanger Sequencing was employed to explore genetic variations. The results included identification of two previously reported heterozygous variants in homozygous form for the first time in RAG1gene [(p.Arg410Gln);(p.Arg737His)], followed by a recurrent variant (p.Trp959*) in RAG1, a novel variant in IL2RG (p.Asp48Lfs*24), a recurrent variant in IL2RG (p.Gly271Glu) and a recurrent variant in DCLRE1C (p.Arg191*) gene. CONCLUSION: To conclude, the immune-profiling and WES revealed two novel, two as homozygous state for the first time, and two recurrent disease causing variants contributing valuably to our existing knowledge of SCID.


Sujet(s)
Immunodéficience combinée grave , Humains , Immunodéficience combinée grave/génétique , Consanguinité , Pakistan , Homozygote , Phénotype , Mutation/génétique , Pedigree
14.
BMC Pediatr ; 24(1): 116, 2024 Feb 13.
Article de Anglais | MEDLINE | ID: mdl-38350907

RÉSUMÉ

BACKGROUND: Severe combined immunodeficiencies (SCIDs) are hereditary disorders characterized by impaired T and B cell function, resulting in significant immune system dysfunction. Recombination-activating gene (RAG) mutations account for a substantial proportion of SCID cases. Here, we present two sibling cases of SCID caused by a novel RAG2 gene mutation. CASE PRESENTATION: The index case was an 8-year-old boy who had a history of recurring infections. After a comprehensive immunological workup, the initial diagnosis of agammaglobulinemia was revised to combined immunodeficiency (CID). The patient underwent hematopoietic stem cell transplantation (HSCT) but succumbed to cytomegalovirus (CMV) infection. His brother, a 4-month-old boy, presented with CMV chorioretinitis. Leaky SCID was diagnosed based on genetic tests and immunological findings. The patient received appropriate treatment and was considered for HSCT. Both siblings had a homozygous RAG2 gene variant, with the first case classified as a variant of uncertain significance (VUS). The presence of the same mutation in the second brother, and the clinical phenotype, supports considering the mutation as likely pathogenic. CONCLUSIONS: This case report highlights a novel RAG2 gene mutation associated with CID. The classification of a VUS may evolve with accumulating evidence, and additional studies are warranted to establish its pathogenicity. Proper communication between genetic counselors and immunologists, accurate documentation of patient information, increased public awareness, and precise utilization of genetic techniques are essential for optimal patient management.


Sujet(s)
Infections à cytomégalovirus , Immunodéficience combinée grave , Mâle , Humains , Nourrisson , Enfant , Fratrie , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/thérapie , Mutation , Lymphocytes B , Infections à cytomégalovirus/diagnostic , Infections à cytomégalovirus/complications , Protéines de liaison à l'ADN/génétique , Protéines nucléaires/génétique
15.
Nat Med ; 30(2): 488-497, 2024 Feb.
Article de Anglais | MEDLINE | ID: mdl-38355973

RÉSUMÉ

Adenosine deaminase (ADA) deficiency leads to severe combined immunodeficiency (SCID). Previous clinical trials showed that autologous CD34+ cell gene therapy (GT) following busulfan reduced-intensity conditioning is a promising therapeutic approach for ADA-SCID, but long-term data are warranted. Here we report an analysis on long-term safety and efficacy data of 43 patients with ADA-SCID who received retroviral ex vivo bone marrow-derived hematopoietic stem cell GT. Twenty-two individuals (median follow-up 15.4 years) were treated in the context of clinical development or named patient program. Nineteen patients were treated post-marketing authorization (median follow-up 3.2 years), and two additional patients received mobilized peripheral blood CD34+ cell GT. At data cutoff, all 43 patients were alive, with a median follow-up of 5.0 years (interquartile range 2.4-15.4) and 2 years intervention-free survival (no need for long-term enzyme replacement therapy or allogeneic hematopoietic stem cell transplantation) of 88% (95% confidence interval 78.7-98.4%). Most adverse events/reactions were related to disease background, busulfan conditioning or immune reconstitution; the safety profile of the real world experience was in line with premarketing cohort. One patient from the named patient program developed a T cell leukemia related to treatment 4.7 years after GT and is currently in remission. Long-term persistence of multilineage gene-corrected cells, metabolic detoxification, immune reconstitution and decreased infection rates were observed. Estimated mixed-effects models showed that higher dose of CD34+ cells infused and younger age at GT affected positively the plateau of CD3+ transduced cells, lymphocytes and CD4+ CD45RA+ naive T cells, whereas the cell dose positively influenced the final plateau of CD15+ transduced cells. These long-term data suggest that the risk-benefit of GT in ADA remains favorable and warrant for continuing long-term safety monitoring. Clinical trial registration: NCT00598481 , NCT03478670 .


Sujet(s)
Agammaglobulinémie , Transplantation de cellules souches hématopoïétiques , Immunodéficience combinée grave , Humains , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/thérapie , Adenosine deaminase/génétique , Adenosine deaminase/usage thérapeutique , Busulfan/effets indésirables , Thérapie génétique , Retroviridae/génétique
16.
Immunol Rev ; 322(1): 138-147, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38287514

RÉSUMÉ

Severe combined immunodeficiency (SCID) is a rare and life-threatening genetic disorder that severely impairs the immune system's ability to defend the body against infections. Often referred to as the "bubble boy" disease, SCID gained widespread recognition due to the case of David Vetter, a young boy who lived in a sterile plastic bubble to protect him from germs. SCID is typically present at birth, and it results from genetic mutations that affect the development and function of immune cells, particularly T cells and B cells. These immune cells are essential for identifying and fighting off infections caused by viruses, bacteria, and fungi. In SCID patients, the immune system is virtually non-existent, leaving them highly susceptible to recurrent, severe infections. There are several forms of SCID, with varying degrees of severity, but all share common features. Newborns with SCID often exhibit symptoms such as chronic diarrhea, thrush, skin rashes, and persistent infections that do not respond to standard treatments. Without prompt diagnosis and intervention, SCID can lead to life-threatening complications and a high risk of mortality. There are over 20 possible affected genes. Treatment options for SCID primarily involve immune reconstitution, with the most well-known approach being hematopoietic stem cell transplantation (HSCT). Alternatively, gene therapy is also available for some forms of SCID. Once treated successfully, SCID patients can lead relatively normal lives, but they may still require vigilant infection control measures and lifelong medical follow-up to manage potential complications. In conclusion, severe combined immunodeficiency is a rare but life-threatening genetic disorder that severely compromises the immune system's function, rendering affected individuals highly vulnerable to infections. Early diagnosis and appropriate treatment are fundamental. With this respect, newborn screening is progressively and dramatically improving the prognosis of SCID.


Sujet(s)
Agammaglobulinémie , Transplantation de cellules souches hématopoïétiques , Immunodéficience combinée grave , Mâle , Nouveau-né , Humains , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/thérapie , Lymphocytes T , Diagnostic précoce , Mutation , Transplantation de cellules souches hématopoïétiques/méthodes
17.
Hum Gene Ther ; 35(7-8): 269-283, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38251667

RÉSUMÉ

Interleukin 7 Receptor alpha Severe Combined Immunodeficiency (IL7R-SCID) is a life-threatening disorder caused by homozygous mutations in the IL7RA gene. Defective IL7R expression in humans hampers T cell precursors' proliferation and differentiation during lymphopoiesis resulting in the absence of T cells in newborns, who succumb to severe infections and death early after birth. Previous attempts to tackle IL7R-SCID by viral gene therapy have shown that unregulated IL7R expression predisposes to leukemia, suggesting the application of targeted gene editing to insert a correct copy of the IL7RA gene in its genomic locus and mediate its physiological expression as a more feasible therapeutic approach. To this aim, we have first developed a CRISPR/Cas9-based IL7R-SCID disease modeling system that recapitulates the disease phenotype in primary human T cells and hematopoietic stem and progenitor cells (HSPCs). Then, we have designed a knockin strategy that targets IL7RA exon 1 and introduces through homology-directed repair a corrective, promoterless IL7RA cDNA followed by a reporter cassette through AAV6 transduction. Targeted integration of the corrective cassette in primary T cells restored IL7R expression and rescued functional downstream IL7R signaling. When applied to HSPCs further induced to differentiate into T cells in an Artificial Thymic Organoid system, our gene editing strategy overcame the T cell developmental block observed in IL7R-SCID patients, while promoting full maturation of T cells with physiological and developmentally regulated IL7R expression. Finally, genotoxicity assessment of the CRISPR/Cas9 platform in HSPCs using biased and unbiased technologies confirmed the safety of the strategy, paving the way for a new, efficient, and safe therapeutic option for IL7R-SCID patients.


Sujet(s)
Immunodéficience combinée grave , Nouveau-né , Humains , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/thérapie , Lymphocytes T/métabolisme , Systèmes CRISPR-Cas , Cellules souches hématopoïétiques/métabolisme , Édition de gène/méthodes , Récepteurs à l'interleukine-7/génétique , Récepteurs à l'interleukine-7/métabolisme
18.
Blood ; 143(15): 1476-1487, 2024 Apr 11.
Article de Anglais | MEDLINE | ID: mdl-38194689

RÉSUMÉ

ABSTRACT: Mutations in the small Rho-family guanosine triphosphate hydrolase RAC2, critical for actin cytoskeleton remodeling and intracellular signal transduction, are associated with neonatal severe combined immunodeficiency (SCID), infantile neutrophilic disorder resembling leukocyte adhesion deficiency (LAD), and later-onset combined immune deficiency (CID). We investigated 54 patients (23 previously reported) from 37 families yielding 15 novel RAC2 missense mutations, including one present only in homozygosity. Data were collected from referring physicians and literature reports with updated clinical information. Patients were grouped by presentation: neonatal SCID (n = 5), infantile LAD-like disease (n = 5), or CID (n = 44). Disease correlated to RAC2 activity: constitutively active RAS-like mutations caused neonatal SCID, dominant-negative mutations caused LAD-like disease, whereas dominant-activating mutations caused CID. Significant T- and B-lymphopenia with low immunoglobulins were seen in most patients; myeloid abnormalities included neutropenia, altered oxidative burst, impaired neutrophil migration, and visible neutrophil macropinosomes. Among 42 patients with CID with clinical data, upper and lower respiratory infections and viral infections were common. Twenty-three distinct RAC2 mutations, including 15 novel variants, were identified. Using heterologous expression systems, we assessed downstream effector functions including superoxide production, p21-activated kinase 1 binding, AKT activation, and protein stability. Confocal microscopy showed altered actin assembly evidenced by membrane ruffling and macropinosomes. Altered protein localization and aggregation were observed. All tested RAC2 mutant proteins exhibited aberrant function; no single assay was sufficient to determine functional consequence. Most mutants produced elevated superoxide; mutations unable to support superoxide formation were associated with bacterial infections. RAC2 mutations cause a spectrum of immune dysfunction, ranging from early onset SCID to later-onset combined immunodeficiencies depending on RAC2 activity. This trial was registered at www.clinicaltrials.gov as #NCT00001355 and #NCT00001467.


Sujet(s)
Déficits immunitaires , Déficit d'adhérence leucocytaire , Maladies d'immunodéficience primaire , Immunodéficience combinée grave , Humains , Nouveau-né , Déficits immunitaires/génétique , Déficits immunitaires/métabolisme , Granulocytes neutrophiles/métabolisme , Maladies d'immunodéficience primaire/génétique , Maladies d'immunodéficience primaire/métabolisme , Protéines G rac/génétique , Protéines G rac/métabolisme , Protéine G rac1/métabolisme , , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/métabolisme , Superoxydes/métabolisme
19.
J Allergy Clin Immunol Pract ; 12(5): 1139-1149, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38246560

RÉSUMÉ

During the past 20 years, gene editing has emerged as a novel form of gene therapy. Since the publication of the first potentially therapeutic gene editing platform for genetic disorders, increasingly sophisticated editing technologies have been developed. As with viral vector-mediated gene addition, inborn errors of immunity are excellent candidate diseases for a corrective autologous hematopoietic stem cell gene editing strategy. Research on gene editing for inborn errors of immunity is still entirely preclinical, with no trials yet underway. However, with editing techniques maturing, scientists are investigating this novel form of gene therapy in context of an increasing number of inborn errors of immunity. Here, we present an overview of these studies and the recent progress moving these technologies closer to clinical benefit.


Sujet(s)
Édition de gène , Thérapie génétique , Humains , Édition de gène/méthodes , Thérapie génétique/méthodes , Animaux , Systèmes CRISPR-Cas , Agammaglobulinémie/génétique , Agammaglobulinémie/thérapie , Immunodéficience combinée grave/génétique , Immunodéficience combinée grave/thérapie , Immunodéficience combinée grave/immunologie , Transplantation de cellules souches hématopoïétiques
20.
J Cosmet Dermatol ; 23(1): 68-75, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37466107

RÉSUMÉ

OBJECTIVE: Through a case of deficiency of adenosine deaminase 2 (DADA2) to improve domestic clinicians' understanding of the disease, and to review the literature, promote dermatologists for clinical secondary primary lesion diagnosis. METHOD: Analysis of a case diagnosed with DADA2 deficiency of clinical manifestations, laboratory, imaging examination and treatment methods, and discussion through literature analysis. RESULTS: The child with recurrent fever, limbs nodular erythema, gradually in the limbs. CT of lower limb skin showed mild edema of the spinous layer, intact basal layer, dilated vascular congestion in the superficial dermis, visible RBC extravasation, and changes of telangiectasia ring purpura were considered. Cranial magnetic resonance imaging (MRI) showed a left choroidal cleft cyst. Genetic test was the CECR1 mutation. The treatment with adalimumab was effective. CONCLUSION: In this case, DADA2 is the seventh case in China, and the CECR1 mutation site (c.254A> T p.N85I,c.851G>T p. G284V) was a compound heterozygous mutation. Mastering the clinical characteristics is helpful for clinicians to diagnose this disease.


Sujet(s)
Adenosine deaminase , Immunodéficience combinée grave , Enfant , Humains , Adenosine deaminase/génétique , Protéines et peptides de signalisation intercellulaire , Immunodéficience combinée grave/diagnostic , Immunodéficience combinée grave/génétique , Mutation
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...