Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 2.341
Filtrer
1.
ARP Rheumatol ; 3(2): 151-156, 2024.
Article de Anglais | MEDLINE | ID: mdl-38956997

RÉSUMÉ

Vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic (VEXAS) syndrome is an emerging adult-onset systemic autoinflammatory disorder affecting multiple organ systems. While lung involvement is common in this syndrome, literature regarding specific patterns is sparse. In this report, we present a case description of a patient with VEXAS syndrome who presented at the emergency department on two separate occasions with acute interstitial pneumonia (AIP) and diffuse alveolar hemorrhage (DAH). A literature review with a comparison of our observed findings to the general findings of VEXAS syndrome, AIP, and DAH is provided. This report underscores the rarity of specific pulmonary manifestations associated with VEXAS syndrome, contributing valuable insight to the limited literature available on this topic.


Sujet(s)
Hémorragie , Pneumopathies interstitielles , Alvéoles pulmonaires , Humains , Pneumopathies interstitielles/complications , Pneumopathies interstitielles/imagerie diagnostique , Pneumopathies interstitielles/diagnostic , Alvéoles pulmonaires/anatomopathologie , Mâle , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/complications , Maladies génétiques liées au chromosome X/diagnostic , Maladies pulmonaires/anatomopathologie , Vacuoles/anatomopathologie , Adulte d'âge moyen , Syndrome , Ubiquitin-activating enzymes
2.
Genes (Basel) ; 15(6)2024 May 24.
Article de Anglais | MEDLINE | ID: mdl-38927615

RÉSUMÉ

X-linked hypophosphatemia (XLH) is a rare inherited disorder of renal phosphate wasting with a highly variable phenotype caused by loss-of-function variants in the PHEX gene. The diagnosis of individuals with mild phenotypes can be challenging and often delayed. Here, we describe a three-generation family with a very mild clinical presentation of XLH. The diagnosis was unexpectedly found in a 39-year-old woman who was referred for genetic testing due to an unclear childhood diagnosis of a tubulopathy. Genetic testing performed by next-generation sequencing using a kidney disease gene panel identified a novel non-canonical splice site variant in the PHEX gene. Segregation analysis detected that the consultand's father, who presented with hypophosphatemia and decreased tubular phosphate reabsorption, and the consultand's son also carried this variant. RNA studies demonstrated that the non-canonical splice site variant partially altered the splicing of the PHEX gene, as both wild-type and aberrant splicing transcripts were detected in the two male members with only one copy of the PHEX gene. In conclusion, this case contributes to the understanding of the relationship between splicing variants and the variable expressivity of XLH disease. The mild phenotype of this family can be explained by the coexistence of PHEX transcripts with aberrant and wild-type splicing.


Sujet(s)
Rachitisme hypophosphatémique familial , PHEX Phosphate regulating neutral endopeptidase , Pedigree , Sites d'épissage d'ARN , Humains , PHEX Phosphate regulating neutral endopeptidase/génétique , Adulte , Femelle , Rachitisme hypophosphatémique familial/génétique , Mâle , Sites d'épissage d'ARN/génétique , Épissage des ARN/génétique , Phénotype , Maladies génétiques liées au chromosome X/génétique , Mutation
3.
Mol Autism ; 15(1): 28, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38877552

RÉSUMÉ

BACKGROUND: Mutations in the X-linked gene cyclin-dependent kinase-like 5 (CDKL5) cause a severe neurological disorder characterised by early-onset epileptic seizures, autism and intellectual disability (ID). Impaired hippocampal function has been implicated in other models of monogenic forms of autism spectrum disorders and ID and is often linked to epilepsy and behavioural abnormalities. Many individuals with CDKL5 deficiency disorder (CDD) have null mutations and complete loss of CDKL5 protein, therefore in the current study we used a Cdkl5-/y rat model to elucidate the impact of CDKL5 loss on cellular excitability and synaptic function of CA1 pyramidal cells (PCs). We hypothesised abnormal pre and/or post synaptic function and plasticity would be observed in the hippocampus of Cdkl5-/y rats. METHODS: To allow cross-species comparisons of phenotypes associated with the loss of CDKL5, we generated a loss of function mutation in exon 8 of the rat Cdkl5 gene and assessed the impact of the loss of CDLK5 using a combination of extracellular and whole-cell electrophysiological recordings, biochemistry, and histology. RESULTS: Our results indicate that CA1 hippocampal long-term potentiation (LTP) is enhanced in slices prepared from juvenile, but not adult, Cdkl5-/y rats. Enhanced LTP does not result from changes in NMDA receptor function or subunit expression as these remain unaltered throughout development. Furthermore, Ca2+ permeable AMPA receptor mediated currents are unchanged in Cdkl5-/y rats. We observe reduced mEPSC frequency accompanied by increased spine density in basal dendrites of CA1 PCs, however we find no evidence supporting an increase in silent synapses when assessed using a minimal stimulation protocol in slices. Additionally, we found no change in paired-pulse ratio, consistent with normal release probability at Schaffer collateral to CA1 PC synapses. CONCLUSIONS: Our data indicate a role for CDKL5 in hippocampal synaptic function and raise the possibility that altered intracellular signalling rather than synaptic deficits contribute to the altered plasticity. LIMITATIONS: This study has focussed on the electrophysiological and anatomical properties of hippocampal CA1 PCs across early postnatal development. Studies involving other brain regions, older animals and behavioural phenotypes associated with the loss of CDKL5 are needed to understand the pathophysiology of CDD.


Sujet(s)
Modèles animaux de maladie humaine , Potentialisation à long terme , Protein-Serine-Threonine Kinases , Récepteur de l'AMPA , Récepteurs du N-méthyl-D-aspartate , Spasmes infantiles , Animaux , Mâle , Rats , Région CA1 de l'hippocampe/métabolisme , Région CA1 de l'hippocampe/anatomopathologie , Région CA1 de l'hippocampe/physiopathologie , Syndromes épileptiques/génétique , Syndromes épileptiques/métabolisme , Potentiels post-synaptiques excitateurs , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/métabolisme , Maladies génétiques liées au chromosome X/physiopathologie , Hippocampe/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Protein-Serine-Threonine Kinases/génétique , Cellules pyramidales/métabolisme , Cellules pyramidales/anatomopathologie , Récepteur de l'AMPA/métabolisme , Récepteur de l'AMPA/génétique , Récepteurs du N-méthyl-D-aspartate/métabolisme , Récepteurs du N-méthyl-D-aspartate/génétique , Spasmes infantiles/génétique , Spasmes infantiles/métabolisme , Synapses/métabolisme
4.
Reumatismo ; 76(2)2024 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-38916169

RÉSUMÉ

Vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic (VEXAS) syndrome is a recently characterized disease associated with somatic mutations in the UBA1 gene, which cause dysregulation of ubiquitin-mediated processes. This case describes a 71-year-old male patient with VEXAS syndrome who presented with refractory lung inflammation with a pattern similar to computed tomography hypersensitivity pneumonitis, a novel finding in VEXAS syndrome. The presented clinical case highlights the protean involvement of the lung in VEXAS syndrome and emphasizes the importance of considering interstitial lung disease in the differential diagnosis.


Sujet(s)
Alvéolite allergique extrinsèque , Ubiquitin-activating enzymes , Humains , Mâle , Sujet âgé , Alvéolite allergique extrinsèque/génétique , Alvéolite allergique extrinsèque/diagnostic , Ubiquitin-activating enzymes/génétique , Syndrome , Vacuoles , Diagnostic différentiel , Tomodensitométrie , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/complications , Maladies génétiques liées au chromosome X/diagnostic , Maladies auto-inflammatoires héréditaires/génétique , Maladies auto-inflammatoires héréditaires/diagnostic , Maladies auto-inflammatoires héréditaires/complications , Mutation , Pneumopathies interstitielles/génétique , Pneumopathies interstitielles/étiologie , Pneumopathies interstitielles/imagerie diagnostique , Poumon/imagerie diagnostique , Poumon/anatomopathologie
5.
Biochemistry ; 63(13): 1636-1646, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38888931

RÉSUMÉ

The conserved enzyme aminolevulinic acid synthase (ALAS) initiates heme biosynthesis in certain bacteria and eukaryotes by catalyzing the condensation of glycine and succinyl-CoA to yield aminolevulinic acid. In humans, the ALAS isoform responsible for heme production during red blood cell development is the erythroid-specific ALAS2 isoform. Owing to its essential role in erythropoiesis, changes in human ALAS2 (hALAS2) function can lead to two different blood disorders. X-linked sideroblastic anemia results from loss of ALAS2 function, while X-linked protoporphyria results from gain of ALAS2 function. Interestingly, mutations in the ALAS2 C-terminal extension can be implicated in both diseases. Here, we investigate the molecular basis for enzyme dysfunction mediated by two previously reported C-terminal loss-of-function variants, hALAS2 V562A and M567I. We show that the mutations do not result in gross structural perturbations, but the enzyme stability for V562A is decreased. Additionally, we show that enzyme stability moderately increases with the addition of the pyridoxal 5'-phosphate (PLP) cofactor for both variants. The variants display differential binding to PLP and the individual substrates compared to wild-type hALAS2. Although hALAS2 V562A is a more active enzyme in vitro, it is less efficient concerning succinyl-CoA binding. In contrast, the M567I mutation significantly alters the cooperativity of substrate binding. In combination with previously reported cell-based studies, our work reveals the molecular basis by which hALAS2 C-terminal mutations negatively affect ALA production necessary for proper heme biosynthesis.


Sujet(s)
5-Aminolevulinate synthetase , Anémie sidéroblastique , Humains , 5-Aminolevulinate synthetase/génétique , 5-Aminolevulinate synthetase/métabolisme , 5-Aminolevulinate synthetase/composition chimique , 5-Aminolevulinate synthetase/déficit , Anémie sidéroblastique/génétique , Anémie sidéroblastique/métabolisme , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/métabolisme , Mutation perte de fonction , Stabilité enzymatique , Hème/métabolisme , Hème/composition chimique , Porphyries/génétique , Porphyries/métabolisme , Modèles moléculaires , Mutation , Protoporphyrie érythropoïétique
8.
Clin Neurol Neurosurg ; 242: 108351, 2024 07.
Article de Anglais | MEDLINE | ID: mdl-38801808

RÉSUMÉ

INTRODUCTION: VEXAS (Vacuoles, E1 Enzyme, X-linked, autoinflammatory, Somatic) syndrome is a recently described severe adult-onset autoinflammatory disorder mediated by X-linked gene UBA1 somatic mutations, responsible of recurrent fever, skin involvement, chondritis, macrocytic anemia and inflammatory syndrome. Neurological manifestations are rarely described, and predominantly involve peripheral nervous system (PNS) impairment. RESULTS: We report the first central nervous system (CNS) vasculitis in VEXAS syndrome, characterized by headache, cognitive dysfunction and focal signs (cerebellar ataxia). Magnetic resonance imaging (MRI) revealed multifocal white-matter lesions corresponding to recent ischemic strokes, combined with cortical hemorrhagic lesions and gadolinium enhancement of the distal wall vessels. Treatment with methylprednisone, ruxolitinib and tocilizumab led to clinical improvement and a decrease of the inflammatory syndrome. The patient died few months after due to infectious complications. CONCLUSION: CNS vasculitis, occurring as a manifestation of the systemic auto-inflammatory state of VEXAS syndrome, might be a rare but severe complication. We suggest that it be added to the list of inflammatory vasculopathies. More prospective studies are needed to optimize the treatment.


Sujet(s)
Vascularite du système nerveux central , Humains , Vascularite du système nerveux central/traitement médicamenteux , Mâle , Adulte d'âge moyen , Imagerie par résonance magnétique , Issue fatale , Adulte , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/complications , Ubiquitin-activating enzymes
9.
Am J Hum Genet ; 111(6): 1206-1221, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38772379

RÉSUMÉ

Utilizing trio whole-exome sequencing and a gene matching approach, we identified a cohort of 18 male individuals from 17 families with hemizygous variants in KCND1, including two de novo missense variants, three maternally inherited protein-truncating variants, and 12 maternally inherited missense variants. Affected subjects present with a neurodevelopmental disorder characterized by diverse neurological abnormalities, mostly delays in different developmental domains, but also distinct neuropsychiatric signs and epilepsy. Heterozygous carrier mothers are clinically unaffected. KCND1 encodes the α-subunit of Kv4.1 voltage-gated potassium channels. All variant-associated amino acid substitutions affect either the cytoplasmic N- or C-terminus of the channel protein except for two occurring in transmembrane segments 1 and 4. Kv4.1 channels were functionally characterized in the absence and presence of auxiliary ß subunits. Variant-specific alterations of biophysical channel properties were diverse and varied in magnitude. Genetic data analysis in combination with our functional assessment shows that Kv4.1 channel dysfunction is involved in the pathogenesis of an X-linked neurodevelopmental disorder frequently associated with a variable neuropsychiatric clinical phenotype.


Sujet(s)
Troubles du développement neurologique , Adolescent , Adulte , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Mâle , Épilepsie/génétique , , Maladies génétiques liées au chromosome X/génétique , Hétérozygote , Mutation faux-sens/génétique , Troubles du développement neurologique/génétique , Pedigree , Phénotype , Canaux potassiques Shal/génétique
10.
Ren Fail ; 46(1): 2349133, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38726999

RÉSUMÉ

OBJECTIVE:  The clinical characteristics, genetic mutation spectrum, treatment strategies and prognoses of 15 children with Dent disease were retrospectively analyzed to improve pediatricians' awareness of and attention to this disease. METHODS:  We analyzed the clinical and laboratory data of 15 Chinese children with Dent disease who were diagnosed and treated at our hospital between January 2017 and May 2023 and evaluated the expression of the CLCN5 and OCRL1 genes. RESULTS:  All 15 patients were male and complained of proteinuria, and the incidence of low-molecular-weight proteinuria (LMWP) was 100.0% in both Dent disease 1 (DD1) and Dent disease 2 (DD2) patients. The incidence of hypercalciuria was 58.3% (7/12) and 66.7% (2/3) in DD1 and DD2 patients, respectively. Nephrocalcinosis and nephrolithiasis were found in 16.7% (2/12) and 8.3% (1/12) of DD1 patients, respectively. Renal biopsy revealed focal segmental glomerulosclerosis (FSGS) in 1 patient, minimal change lesion in 5 patients, and small focal acute tubular injury in 1 patient. A total of 11 mutations in the CLCN5 gene were detected, including 3 missense mutations (25.0%, c.1756C > T, c.1166T > G, and c.1618G > A), 5 frameshift mutations (41.7%, c.407delT, c.1702_c.1703insC, c.137delC, c.665_666delGGinsC, and c.2200delG), and 3 nonsense mutations (25.0%, c.776G > A, c.1609C > T, and c.1152G > A). There was no significant difference in age or clinical phenotype among patients with different mutation types (p > 0.05). All three mutations in the OCRL1 gene were missense mutations (c.1477C > T, c.952C > T, and c.198A > G). CONCLUSION:  Pediatric Dent disease is often misdiagnosed. Protein electrophoresis and genetic testing can help to provide an early and correct diagnosis.


Sujet(s)
Canaux chlorure , Maladie de Dent , Phosphoric monoester hydrolases , Humains , Mâle , Enfant , Canaux chlorure/génétique , Études rétrospectives , Enfant d'âge préscolaire , Chine/épidémiologie , Maladie de Dent/génétique , Maladie de Dent/diagnostic , Phosphoric monoester hydrolases/génétique , Mutation , Protéinurie/génétique , Adolescent , Hypercalciurie/génétique , Néphrocalcinose/génétique , Néphrolithiase/génétique , Nourrisson , Dépistage génétique , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/diagnostic , Mutation faux-sens , Femelle , Glomérulonéphrite segmentaire et focale/génétique , Rein/anatomopathologie , Peuples d'Asie de l'Est
11.
Curr Opin Infect Dis ; 37(4): 227-231, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38747352

RÉSUMÉ

PURPOSE OF REVIEW: The purpose of this focused review is to discuss unusual presentations of viral infections in the context of specific inborn errors of immunity. We will discuss hyper immunoglobulin E (IgE) syndromes, epidermodysplasia verruciformis, and X-linked agammaglobulinemia as examples of inborn errors of immunity associated with specific presentations of viral infection and disease. RECENT FINDINGS: Advances in both genetic and viral diagnostics have broadened our understanding of viral pathogenesis in the setting of immune dysfunction and the variable phenotype of inborn errors of immunity. Dedicator of cytokinesis 8 (DOCK8) deficiency is now recognized as an inborn error of immunity within the hyper IgE syndrome phenotype and is associated with unusually aggressive cutaneous disease caused by herpes simplex and other viruses. Studies of patients with epidermodysplasia verruciformis have proven that rarely detected human papillomavirus subtypes may cause malignancy in the absence of adequate host defenses. Finally, patients with X-linked agammaglobulinemia may remain at risk for severe and chronic viral infections, even as immune globulin supplementation reduces the risk of bacterial infection. SUMMARY: Susceptibility to viral infections in patients with inborn errors of immunity is conferred by specific, molecular defects. Recurrent, severe, or otherwise unusual presentations of viral disease should prompt investigation for an underlying genetic defect.


Sujet(s)
Agammaglobulinémie , Maladies génétiques liées au chromosome X , Maladies virales , Humains , Agammaglobulinémie/immunologie , Maladies virales/immunologie , Maladies virales/complications , Maladies génétiques liées au chromosome X/immunologie , Maladies génétiques liées au chromosome X/génétique , Épidermodysplasie verruciforme/immunologie , Épidermodysplasie verruciforme/génétique , Syndrome de Job/immunologie , Syndrome de Job/génétique , Facteurs d'échange de nucléotides guanyliques
12.
Proc Natl Acad Sci U S A ; 121(21): e2404763121, 2024 May 21.
Article de Anglais | MEDLINE | ID: mdl-38743626

RÉSUMÉ

Congenital stationary night blindness (CSNB) is an inherited retinal disease that causes a profound loss of rod sensitivity without severe retinal degeneration. One well-studied rhodopsin point mutant, G90D-Rho, is thought to cause CSNB because of its constitutive activity in darkness causing rod desensitization. However, the nature of this constitutive activity and its precise molecular source have not been resolved for almost 30 y. In this study, we made a knock-in (KI) mouse line with a very low expression of G90D-Rho (equal in amount to ~0.1% of normal rhodopsin, WT-Rho, in WT rods), with the remaining WT-Rho replaced by REY-Rho, a mutant with a very low efficiency of activating transducin due to a charge reversal of the highly conserved ERY motif to REY. We observed two kinds of constitutive noise: one being spontaneous isomerization (R*) of G90D-Rho at a molecular rate (R* s-1) 175-fold higher than WT-Rho and the other being G90D-Rho-generated dark continuous noise comprising low-amplitude unitary events occurring at a very high molecular rate equivalent in effect to ~40,000-fold of R* s-1 from WT-Rho. Neither noise type originated from G90D-Opsin because exogenous 11-cis-retinal had no effect. Extrapolating the above observations at low (0.1%) expression of G90D-Rho to normal disease exhibited by a KI mouse model with RhoG90D/WTand RhoG90D/G90D genotypes predicts the disease condition very well quantitatively. Overall, the continuous noise from G90D-Rho therefore predominates, constituting the major equivalent background light causing rod desensitization in CSNB.


Sujet(s)
Maladies héréditaires de l'oeil , Maladies génétiques liées au chromosome X , Myopie , Héméralopie , Rhodopsine , Animaux , Héméralopie/génétique , Héméralopie/métabolisme , Maladies héréditaires de l'oeil/génétique , Maladies héréditaires de l'oeil/métabolisme , Souris , Rhodopsine/génétique , Rhodopsine/métabolisme , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/métabolisme , Myopie/génétique , Myopie/métabolisme , Cellules photoréceptrices en bâtonnet de la rétine/métabolisme , Cellules photoréceptrices en bâtonnet de la rétine/anatomopathologie , Obscurité , Transducine/génétique , Transducine/métabolisme , Techniques de knock-in de gènes , Modèles animaux de maladie humaine
13.
BMC Pediatr ; 24(1): 291, 2024 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-38689221

RÉSUMÉ

BACKGROUND: X-linked agammaglobulinemia (XLA) is a primary immunodeficiency disease caused by mutations in the Bruton tyrosine kinase (BTK) gene. Individuals diagnosed with XLA are at an increased risk of developing autoimmune diseases. However, renal involvement are rare in cases of XLA. CASE PRESENTATION: In this report, we discussed a specific case involving a 6-year-old boy with XLA who experienced recurrent upper respiratory tract infections since the age of one. He presented with symptoms of hematuria and proteinuria, and renal pathology confirmed the presence of immunoglobulin (Ig) A nephropathy. Treatment comprised glucocorticoids, mycophenolate mofetil, and intermittent intravenous immunoglobulin replacement therapy. Consequently, there was a remission of proteinuria and a partial improvement in hematuria. CONCLUSIONS: In this study, we describe the first case of IgA nephropathy associated with XLA. This is an interesting phenotype found in XLA, and it provides valuable insights into the process of autoimmunity and the regulation of immune function in individuals with XLA. Based on our findings, we recommend the evaluation of immunoglobulin levels in patients diagnosed with IgA nephropathy.


Sujet(s)
Agammaglobulinémie , Maladies génétiques liées au chromosome X , Glomérulonéphrite à dépôts d'IgA , Humains , Agammaglobulinémie/complications , Agammaglobulinémie/diagnostic , Agammaglobulinémie/génétique , Mâle , Glomérulonéphrite à dépôts d'IgA/complications , Glomérulonéphrite à dépôts d'IgA/diagnostic , Maladies génétiques liées au chromosome X/complications , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/diagnostic , Enfant , Immunoglobulines par voie veineuse/usage thérapeutique , Glucocorticoïdes/usage thérapeutique , Acide mycophénolique/usage thérapeutique , Immunosuppresseurs/usage thérapeutique
16.
Cell Rep ; 43(4): 114068, 2024 Apr 23.
Article de Anglais | MEDLINE | ID: mdl-38614085

RÉSUMÉ

The precise anatomical degree of brain X chromosome inactivation (XCI) that is sufficient to alter X-linked disorders in females is unclear. Here, we quantify whole-brain XCI at single-cell resolution to discover a prevalent activation ratio of maternal to paternal X at 60:40 across all divisions of the adult brain. This modest, non-random XCI influences X-linked disease penetrance: maternal transmission of the fragile X mental retardation 1 (Fmr1)-knockout (KO) allele confers 55% of total brain cells with mutant X-active, which is sufficient for behavioral penetrance, while 40% produced from paternal transmission is tolerated. Local XCI mosaicism within affected maternal Fmr1-KO mice further specifies sensorimotor versus social anxiety phenotypes depending on which distinct brain circuitry is most affected, with only a 50%-55% mutant X-active threshold determining penetrance. Thus, our results define a model of X-linked disease penetrance in females whereby distributed XCI among single cells populating brain circuitries can regulate the behavioral penetrance of an X-linked mutation.


Sujet(s)
Encéphale , Souris knockout , Pénétrance , Inactivation du chromosome X , Inactivation du chromosome X/génétique , Animaux , Femelle , Souris , Encéphale/métabolisme , Mâle , Protéine du syndrome X fragile/génétique , Protéine du syndrome X fragile/métabolisme , Comportement animal , Souris de lignée C57BL , Mosaïcisme , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/anatomopathologie
17.
Mol Ther ; 32(6): 1628-1642, 2024 Jun 05.
Article de Anglais | MEDLINE | ID: mdl-38556793

RÉSUMÉ

Severe congenital neutropenia (CN) is an inherited pre-leukemia bone marrow failure syndrome commonly caused by autosomal-dominant ELANE mutations (ELANE-CN). ELANE-CN patients are treated with daily injections of recombinant human granulocyte colony-stimulating factor (rhG-CSF). However, some patients do not respond to rhG-CSF, and approximately 15% of ELANE-CN patients develop myelodysplasia or acute myeloid leukemia. Here, we report the development of a curative therapy for ELANE-CN through inhibition of ELANE mRNA expression by introducing two single-strand DNA breaks at the opposing DNA strands of the ELANE promoter TATA box using CRISPR-Cas9D10A nickases-termed MILESTONE. This editing effectively restored defective neutrophil differentiation of ELANE-CN CD34+ hematopoietic stem and progenitor cells (HSPCs) in vitro and in vivo, without affecting the functions of the edited neutrophils. CRISPResso analysis of the edited ELANE-CN CD34+ HSPCs revealed on-target efficiencies of over 90%. Simultaneously, GUIDE-seq, CAST-Seq, and rhAmpSeq indicated a safe off-target profile with no off-target sites or chromosomal translocations. Taken together, ex vivo gene editing of ELANE-CN HSPCs using MILESTONE in the setting of autologous stem cell transplantation could be a universal, safe, and efficient gene therapy approach for ELANE-CN patients.


Sujet(s)
Systèmes CRISPR-Cas , Insuffisances médullaires congénitales , Édition de gène , Thérapie génétique , Leukocyte elastase , Neutropénie , Régions promotrices (génétique) , Édition de gène/méthodes , Humains , Neutropénie/congénital , Neutropénie/thérapie , Neutropénie/génétique , Thérapie génétique/méthodes , Insuffisances médullaires congénitales/thérapie , Insuffisances médullaires congénitales/génétique , Leukocyte elastase/génétique , Leukocyte elastase/métabolisme , Animaux , Souris , Granulocytes neutrophiles/métabolisme , Cellules souches hématopoïétiques/métabolisme , Mutation , Modèles animaux de maladie humaine , Facteur de stimulation des colonies de granulocytes/génétique , Maladies génétiques liées au chromosome X/thérapie , Maladies génétiques liées au chromosome X/génétique
18.
J Genet ; 1032024.
Article de Anglais | MEDLINE | ID: mdl-38562040

RÉSUMÉ

Intellectual developmental disorder, X-linked 104 (XLID104), caused by the FRMPD4 gene variant, is a rare X-linked genetic disease that primarily manifests as intellectual disability (ID) and language delay, and may be accompanied by behavioural abnormalities. Currently, only 11 patients from four families have been reported to carry FRMPD4 gene variants. Here, we report a rare case of a Chinese patient with XLID104 who was presented with severe ID and language impairment. Genetic testing results showed that the patient had a novel hemizygous variant on FRMPD4 inherited from the heterozygous variant NM_001368397: c.1772A>C (p.Glu591Ala) carried by his mother. To our knowledge, this variant has not been reported previously. Western blot results for the recombinant plasmid constructed in vitro indicated that the expression of the mutant protein may be reduced. Using molecular dynamics simulations, we predicted that the mutant protein may affect the interaction of the FRMPD4 protein with DLG4. In this study, we expand the spectrum of FRMPD4 variants and suggest that the clinical awareness of the genetic diagnosis of nonsyndromic ID should be strengthened.


Sujet(s)
Maladies génétiques liées au chromosome X , Déficience intellectuelle , Enfant , Humains , Déficience intellectuelle/génétique , Domaines FERM , Gènes liés au chromosome X , Maladies génétiques liées au chromosome X/génétique , Protéines mutantes/génétique
20.
J Allergy Clin Immunol ; 154(1): 195-208.e8, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38479630

RÉSUMÉ

BACKGROUND: X-linked agammaglobulinemia (XLA) is an inborn error of immunity that renders boys susceptible to life-threatening infections due to loss of mature B cells and circulating immunoglobulins. It is caused by defects in the gene encoding the Bruton tyrosine kinase (BTK) that mediates the maturation of B cells in the bone marrow and their activation in the periphery. This paper reports on a gene editing protocol to achieve "knock-in" of a therapeutic BTK cassette in hematopoietic stem and progenitor cells (HSPCs) as a treatment for XLA. METHODS: To rescue BTK expression, this study employed a clustered regularly interspaced short palindromic repeats/CRISPR-associated protein 9 system that creates a DNA double-strand break in an early exon of the BTK locus and an adeno-associated virus 6 virus that carries the donor template for homology-directed repair. The investigators evaluated the efficacy of the gene editing approach in HSPCs from patients with XLA that were cultured in vitro under B-cell differentiation conditions or that were transplanted in immunodeficient mice to study B-cell output in vivo. RESULTS: A (feeder-free) B-cell differentiation protocol was successfully applied to blood-mobilized HSPCs to reproduce in vitro the defects in B-cell maturation observed in patients with XLA. Using this system, the investigators could show the rescue of B-cell maturation by gene editing. Transplantation of edited XLA HSPCs into immunodeficient mice led to restoration of the human B-cell lineage compartment in the bone marrow and immunoglobulin production in the periphery. CONCLUSIONS: Gene editing efficiencies above 30% could be consistently achieved in human HSPCs. Given the potential selective advantage of corrected cells, as suggested by skewed X-linked inactivation in carrier females and by competitive repopulating experiments in mouse models, this work demonstrates the potential of this strategy as a future definitive therapy for XLA.


Sujet(s)
Agammaglobulinaemia tyrosine kinase , Agammaglobulinémie , Lymphocytes B , Édition de gène , Maladies génétiques liées au chromosome X , Cellules souches hématopoïétiques , Agammaglobulinémie/génétique , Agammaglobulinémie/thérapie , Agammaglobulinémie/immunologie , Animaux , Agammaglobulinaemia tyrosine kinase/génétique , Maladies génétiques liées au chromosome X/génétique , Maladies génétiques liées au chromosome X/thérapie , Maladies génétiques liées au chromosome X/immunologie , Humains , Lymphocytes B/immunologie , Souris , Mâle , Transplantation de cellules souches hématopoïétiques , Différenciation cellulaire/génétique , Systèmes CRISPR-Cas
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...