Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 15.864
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 184(14): 3812-3828.e30, 2021 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-34214472

RESUMO

We study a patient with the human papilloma virus (HPV)-2-driven "tree-man" phenotype and two relatives with unusually severe HPV4-driven warts. The giant horns form an HPV-2-driven multifocal benign epithelial tumor overexpressing viral oncogenes in the epidermis basal layer. The patients are unexpectedly homozygous for a private CD28 variant. They have no detectable CD28 on their T cells, with the exception of a small contingent of revertant memory CD4+ T cells. T cell development is barely affected, and T cells respond to CD3 and CD2, but not CD28, costimulation. Although the patients do not display HPV-2- and HPV-4-reactive CD4+ T cells in vitro, they make antibodies specific for both viruses in vivo. CD28-deficient mice are susceptible to cutaneous infections with the mouse papillomavirus MmuPV1. The control of HPV-2 and HPV-4 in keratinocytes is dependent on the T cell CD28 co-activation pathway. Surprisingly, human CD28-dependent T cell responses are largely redundant for protective immunity.


Assuntos
Antígenos CD28/deficiência , Padrões de Herança/genética , Papillomaviridae/fisiologia , Pele/virologia , Linfócitos T/imunologia , Adulto , Sequência de Aminoácidos , Animais , Sequência de Bases , Antígenos CD28/genética , Antígenos CD28/metabolismo , Linfócitos T CD4-Positivos/imunologia , Criança , Endopeptidases/metabolismo , Feminino , Genes Recessivos , Células HEK293 , Homozigoto , Humanos , Imunidade Humoral , Memória Imunológica , Células Jurkat , Queratinócitos/patologia , Masculino , Camundongos Endogâmicos C57BL , Oncogenes , Papiloma/patologia , Papiloma/virologia , Linhagem , Sinais Direcionadores de Proteínas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
2.
Nature ; 613(7944): 519-525, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36653560

RESUMO

Identifying causal factors for Mendelian and common diseases is an ongoing challenge in medical genetics1. Population bottleneck events, such as those that occurred in the history of the Finnish population, enrich some homozygous variants to higher frequencies, which facilitates the identification of variants that cause diseases with recessive inheritance2,3. Here we examine the homozygous and heterozygous effects of 44,370 coding variants on 2,444 disease phenotypes using data from the nationwide electronic health records of 176,899 Finnish individuals. We find associations for homozygous genotypes across a broad spectrum of phenotypes, including known associations with retinal dystrophy and novel associations with adult-onset cataract and female infertility. Of the recessive disease associations that we identify, 13 out of 20 would have been missed by the additive model that is typically used in genome-wide association studies. We use these results to find many known Mendelian variants whose inheritance cannot be adequately described by a conventional definition of dominant or recessive. In particular, we find variants that are known to cause diseases with recessive inheritance with significant heterozygous phenotypic effects. Similarly, we find presumed benign variants with disease effects. Our results show how biobanks, particularly in founder populations, can broaden our understanding of complex dosage effects of Mendelian variants on disease.


Assuntos
Alelos , Bancos de Espécimes Biológicos , Doença , Animais , Feminino , Estudo de Associação Genômica Ampla , Fenótipo , Doença/genética , Finlândia , Distrofias Retinianas , Catarata , Infertilidade Feminina , Genes Recessivos , Heterozigoto , Efeito Fundador , Dosagem de Genes , Registros Eletrônicos de Saúde
3.
Annu Rev Cell Dev Biol ; 30: 705-22, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25288120

RESUMO

Most animal genomes are diploid, and mammalian development depends on specific adaptations that have evolved secondary to diploidy. Genomic imprinting and dosage compensation restrict haploid development to early embryos. Recently, haploid mammalian development has been reinvestigated since the establishment of haploid embryonic stem cells (ESCs) from mouse embryos. Haploid cells possess one copy of each gene, facilitating the generation of loss-of-function mutations in a single step. Recessive mutations can then be assessed in forward genetic screens. Applications of haploid mammalian cell systems in screens have been illustrated in several recent publications. Haploid ESCs are characterized by a wide developmental potential and can contribute to chimeric embryos and mice. Different strategies for introducing genetic modifications from haploid ESCs into the mouse germline have been further developed. Haploid ESCs therefore introduce new possibilities in mammalian genetics and could offer an unprecedented tool for genome exploration in the future.


Assuntos
Células-Tronco Embrionárias/citologia , Haploidia , Animais , Blastocisto/citologia , Quimera , Transferência Embrionária , Desenvolvimento Embrionário , Genes Recessivos , Genes Reporter , Testes Genéticos/métodos , Impressão Genômica , Mutação em Linhagem Germinativa , Humanos , Camundongos , Camundongos Transgênicos , Neoplasias/genética , Partenogênese , Especificidade da Espécie , Transgenes
4.
Cell ; 150(3): 533-48, 2012 Aug 03.
Artigo em Inglês | MEDLINE | ID: mdl-22863007

RESUMO

Nephronophthisis-related ciliopathies (NPHP-RC) are degenerative recessive diseases that affect kidney, retina, and brain. Genetic defects in NPHP gene products that localize to cilia and centrosomes defined them as "ciliopathies." However, disease mechanisms remain poorly understood. Here, we identify by whole-exome resequencing, mutations of MRE11, ZNF423, and CEP164 as causing NPHP-RC. All three genes function within the DNA damage response (DDR) pathway. We demonstrate that, upon induced DNA damage, the NPHP-RC proteins ZNF423, CEP164, and NPHP10 colocalize to nuclear foci positive for TIP60, known to activate ATM at sites of DNA damage. We show that knockdown of CEP164 or ZNF423 causes sensitivity to DNA damaging agents and that cep164 knockdown in zebrafish results in dysregulated DDR and an NPHP-RC phenotype. Our findings link degenerative diseases of the kidney and retina, disorders of increasing prevalence, to mechanisms of DDR.


Assuntos
Dano ao DNA , Proteínas de Ligação a DNA/metabolismo , Exoma , Doenças Renais Císticas/genética , Proteínas dos Microtúbulos/metabolismo , Animais , Cílios/metabolismo , Técnicas de Silenciamento de Genes , Genes Recessivos , Humanos , Proteína Homóloga a MRE11 , Camundongos , Proteínas , Transdução de Sinais , Peixe-Zebra/embriologia , Peixe-Zebra/metabolismo
5.
Hum Mol Genet ; 32(5): 825-834, 2023 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-36173926

RESUMO

In human autosomal recessive woolly hair/hypotrichosis (ARWH/HT), many mutations have been identified in a gene encoding LPA6, a G protein-coupled receptor (GPCR) for lysophosphatidic acid (LPA). However, information regarding the effects of such mutations on receptor function is limited. In this study, we examined functional impacts of selected amino acid changes in LPA6 identified in ARWH/HT patients. In our exogenous expression experiments, all mutants except S3T failed to respond to LPA, indicating that they are loss-of-function mutants. Among the nine mutants, five (D63V, G146R, N246D, L277P and C278Y) displayed impaired expression at the cell surface because of endoplasmic reticulum (ER) retention, indicating that these mutants are trafficking-defective, as reported in other disease-associated GPCRs. Notably, alkyl-OMPT, a potent synthetic agonist for LPA6 restored the defective cell surface expression of two of the ER-retained mutants, D63V and N246D, possibly by its chaperoning function that allows them to escape intracellular retention as well as proteasomal degradation. Furthermore, the alkyl-OMPT-rescued N246D mutant was shown be functional. Our findings encourage future application of pharmacoperone therapy for ARWH/HT patients with specific LPA6 mutations.


Assuntos
Doenças do Cabelo , Hipotricose , Humanos , Hipotricose/genética , Cabelo , Doenças do Cabelo/genética , Mutação , Genes Recessivos
6.
Am J Hum Genet ; 109(1): 33-49, 2022 01 06.
Artigo em Inglês | MEDLINE | ID: mdl-34951958

RESUMO

The identification of genes that evolve under recessive natural selection is a long-standing goal of population genetics research that has important applications to the discovery of genes associated with disease. We found that commonly used methods to evaluate selective constraint at the gene level are highly sensitive to genes under heterozygous selection but ubiquitously fail to detect recessively evolving genes. Additionally, more sophisticated likelihood-based methods designed to detect recessivity similarly lack power for a human gene of realistic length from current population sample sizes. However, extensive simulations suggested that recessive genes may be detectable in aggregate. Here, we offer a method informed by population genetics simulations designed to detect recessive purifying selection in gene sets. Applying this to empirical gene sets produced significant enrichments for strong recessive selection in genes previously inferred to be under recessive selection in a consanguineous cohort and in genes involved in autosomal recessive monogenic disorders.


Assuntos
Frequência do Gene , Genes Recessivos , Genética Populacional , Seleção Genética , Algoritmos , Alelos , Genes Dominantes , Predisposição Genética para Doença , Variação Genética , Genética Populacional/métodos , Genômica/métodos , Genótipo , Humanos , Padrões de Herança , Funções Verossimilhança , Modelos Genéticos , Mutação , Reino Unido
7.
Hum Genomics ; 18(1): 35, 2024 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-38570878

RESUMO

BACKGROUND: To investigate the genetics of early-onset progressive cerebellar ataxia in Iran, we conducted a study at the Children's Medical Center (CMC), the primary referral center for pediatric disorders in the country, over a three-year period from 2019 to 2022. In this report, we provide the initial findings from the national registry. METHODS: We selected all early-onset patients with an autosomal recessive mode of inheritance to assess their phenotype, paraclinical tests, and genotypes. The clinical data encompassed clinical features, the Scale for the Assessment and Rating of Ataxia (SARA) scores, Magnetic Resonance Imaging (MRI) results, Electrodiagnostic exams (EDX), and biomarker features. Our genetic investigations included single-gene testing, Whole Exome Sequencing (WES), and Whole Genome Sequencing (WGS). RESULTS: Our study enrolled 162 patients from various geographic regions of our country. Among our subpopulations, we identified known and novel pathogenic variants in 42 genes in 97 families. The overall genetic diagnostic rate was 59.9%. Notably, we observed PLA2G6, ATM, SACS, and SCA variants in 19, 14, 12, and 10 families, respectively. Remarkably, more than 59% of the cases were attributed to pathogenic variants in these genes. CONCLUSIONS: Iran, being at the crossroad of the Middle East, exhibits a highly diverse genetic etiology for autosomal recessive hereditary ataxia. In light of this heterogeneity, the development of preventive strategies and targeted molecular therapeutics becomes crucial. A national guideline for the diagnosis and management of patients with these conditions could significantly aid in advancing healthcare approaches and improving patient outcomes.


Assuntos
Degenerações Espinocerebelares , Criança , Humanos , Irã (Geográfico)/epidemiologia , Degenerações Espinocerebelares/genética , Testes Genéticos , Fenótipo , Genes Recessivos
8.
Nature ; 568(7753): 541-545, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30971820

RESUMO

Osteoclasts are multinucleated giant cells that resorb bone, ensuring development and continuous remodelling of the skeleton and the bone marrow haematopoietic niche. Defective osteoclast activity leads to osteopetrosis and bone marrow failure1-9, whereas excess activity can contribute to bone loss and osteoporosis10. Osteopetrosis can be partially treated by bone marrow transplantation in humans and mice11-18, consistent with a haematopoietic origin of osteoclasts13,16,19 and studies that suggest that they develop by fusion of monocytic precursors derived from haematopoietic stem cells in the presence of CSF1 and RANK ligand1,20. However, the developmental origin and lifespan of osteoclasts, and the mechanisms that ensure maintenance of osteoclast function throughout life in vivo remain largely unexplored. Here we report that osteoclasts that colonize fetal ossification centres originate from embryonic erythro-myeloid progenitors21,22. These erythro-myeloid progenitor-derived osteoclasts are required for normal bone development and tooth eruption. Yet, timely transfusion of haematopoietic-stem-cell-derived monocytic cells in newborn mice is sufficient to rescue bone development in early-onset autosomal recessive osteopetrosis. We also found that the postnatal maintenance of osteoclasts, bone mass and the bone marrow cavity involve iterative fusion of circulating blood monocytic cells with long-lived osteoclast syncytia. As a consequence, parabiosis or transfusion of monocytic cells results in long-term gene transfer in osteoclasts in the absence of haematopoietic-stem-cell chimerism, and can rescue an adult-onset osteopetrotic phenotype caused by cathepsin K deficiency23,24. In sum, our results identify the developmental origin of osteoclasts and a mechanism that controls their maintenance in bones after birth. These data suggest strategies to rescue osteoclast deficiency in osteopetrosis and to modulate osteoclast activity in vivo.


Assuntos
Células-Tronco Hematopoéticas/citologia , Osteoclastos/citologia , Osteoclastos/metabolismo , Osteopetrose/genética , Animais , Animais Recém-Nascidos , Desenvolvimento Ósseo , Feminino , Genes Recessivos , Masculino , Camundongos , Osteopetrose/patologia , Erupção Dentária
9.
J Med Genet ; 61(8): 783-787, 2024 Jul 19.
Artigo em Inglês | MEDLINE | ID: mdl-38719349

RESUMO

BACKGROUND: We aimed to analyse the efficacy and added value of a targeted Israeli expanded carrier screening panel (IL-ECSP), beyond the first-tier test covered by the Israeli Ministry of Health (IMOH) and the second-tier covered by the Health Maintenance Organisations (HMOs). METHODS: A curated variant-based IL-ECSP, tailored to the uniquely diverse Israeli population, was offered at two tertiary hospitals and a major genetics laboratory. The panel includes 1487 variants in 357 autosomal recessive and X-linked genes. RESULTS: We analysed 10 115 Israeli samples during an 18-month period. Of these, 6036 (59.7%) were tested as couples and 4079 (40.3%) were singles. Carriers were most frequently identified with mutations in the following genes: GJB2/GJB6 (1:22 allele frequency), CFTR (1:28), GBA (1:34), TYR (1:39), PAH (1:50), SMN1 (1:52) and HEXA (1:56). Of 3018 couples tested, 753 (25%) had no findings, in 1464 (48.5%) only one partner was a carrier, and in 733 (24.3%) both were carriers of different diseases. We identified 79 (2.6%) at-risk couples, where both partners are carriers of the same autosomal recessive condition, or the female carries an X-linked disease. Importantly, 48.1% of these would not have been detected by ethnically-based screening tests currently provided by the IMOH and HMOs, for example, variants in GBA, TYR, PAH and GJB2/GJB6. CONCLUSION: This is the largest cohort of targeted ECSP testing, tailored to the diverse Israeli population. The IL-ECSP expands the identification of couples at risk and empowers their reproductive choices. We recommend endorsing an expanded targeted panel to the National Genetic Carrier Screening programme.


Assuntos
Conexina 26 , Testes Genéticos , Humanos , Israel/epidemiologia , Feminino , Testes Genéticos/métodos , Masculino , Conexina 26/genética , Conexinas/genética , Triagem de Portadores Genéticos/métodos , Mutação , Cuidado Pré-Concepcional/métodos , Frequência do Gene , Aconselhamento Genético , Heterozigoto , Genes Recessivos , Adulto
10.
Hum Hered ; 89(1): 1-7, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38342085

RESUMO

INTRODUCTION: Previous studies have demonstrated effects of rare coding variants on common, clinically relevant phenotypes although the additive burden of these variants makes only a small contribution to overall trait variance. Although recessive effects of individual homozygous variants have been studied, little work has been done to elucidate the impact of rare coding variants occurring together as compound heterozygotes. METHODS: In this study, attempts were made to identify pairs of variants likely to be occurring as compound heterozygotes using 200,000 exome-sequenced subjects from the UK Biobank. Pairs of variants, which were seen together in the same subject more often than would be expected by chance, were excluded as it was assumed that these might be present in the same haplotype. Attention was restricted to variants with minor allele frequency ≤0.05 and to those predicted to alter amino acid sequence or prevent normal gene expression. For each gene, compound heterozygotes were assigned scores based on the rarity and predicted functional consequences of the constituent variants and the scores were used in a logistic regression analysis to test for association with hypertension, hyperlipidaemia, and type 2 diabetes. RESULTS: No statistically significant associations were observed and the results conformed to the distribution, which would be expected under the null hypothesis. The average number of apparently compound heterozygous subjects for each gene was only 282.2. CONCLUSION: It seems difficult to detect an effect of compound heterozygotes on the risk of these phenotypes. Even if recessive effects from compound heterozygotes do occur, they would only affect a small number of people and overall would not make a substantial contribution to phenotypic variance. This research has been conducted using the UK Biobank Resource.


Assuntos
Exoma , Fenótipo , Feminino , Humanos , Masculino , Diabetes Mellitus Tipo 2/genética , Exoma/genética , Sequenciamento do Exoma/métodos , Frequência do Gene/genética , Genes Recessivos , Predisposição Genética para Doença , Variação Genética , Heterozigoto , Biobanco do Reino Unido , Reino Unido
11.
J Cell Mol Med ; 28(8): e18119, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38534090

RESUMO

Hearing loss is a clinically and genetically heterogeneous disorder, with over 148 genes and 170 loci associated with its pathogenesis. The spectrum and frequency of causal variants vary across different genetic ancestries and are more prevalent in populations that practice consanguineous marriages. Pakistan has a rich history of autosomal recessive gene discovery related to non-syndromic hearing loss. Since the first linkage analysis with a Pakistani family that led to the mapping of the DFNB1 locus on chromosome 13, 51 genes associated with this disorder have been identified in this population. Among these, 13 of the most prevalent genes, namely CDH23, CIB2, CLDN14, GJB2, HGF, MARVELD2, MYO7A, MYO15A, MSRB3, OTOF, SLC26A4, TMC1 and TMPRSS3, account for more than half of all cases of profound hearing loss, while the prevalence of other genes is less than 2% individually. In this review, we discuss the most common autosomal recessive non-syndromic hearing loss genes in Pakistani individuals as well as the genetic mapping and sequencing approaches used to discover them. Furthermore, we identified enriched gene ontology terms and common pathways involved in these 51 autosomal recessive non-syndromic hearing loss genes to gain a better understanding of the underlying mechanisms. Establishing a molecular understanding of the disorder may aid in reducing its future prevalence by enabling timely diagnostics and genetic counselling, leading to more effective clinical management and treatments of hearing loss.


Assuntos
Surdez , Perda Auditiva , Humanos , Genes Recessivos , Paquistão , Mutação , Perda Auditiva/genética , Linhagem , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Serina Endopeptidases/genética , Proteína 2 com Domínio MARVEL/genética
12.
BMC Genomics ; 25(1): 417, 2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38678201

RESUMO

BACKGROUND: Between 2020 and 2022, eight calves in a Nebraska herd (composite Simmental, Red Angus, Gelbvieh) displayed exercise intolerance during forced activity. In some cases, the calves collapsed and did not recover. Available sire pedigrees contained a paternal ancestor within 2-4 generations in all affected calves. Pedigrees of the calves' dams were unavailable, however, the cows were ranch-raised and retained from prior breeding seasons, where bulls used for breeding occasionally had a common ancestor. Therefore, it was hypothesized that a de novo autosomal recessive variant was causative of exercise intolerance in these calves. RESULTS: A genome-wide association analysis utilizing SNP data from 6 affected calves and 715 herd mates, followed by whole-genome sequencing of 2 affected calves led to the identification of a variant in the gene PYGM (BTA29:g.42989581G > A). The variant, confirmed to be present in the skeletal muscle transcriptome, was predicted to produce a premature stop codon (p.Arg650*). The protein product of PYGM, myophosphorylase, breaks down glycogen in skeletal muscle. Glycogen concentrations were fluorometrically assayed as glucose residues demonstrating significantly elevated glycogen concentrations in affected calves compared to cattle carrying the variant and to wild-type controls. The absence of the PYGM protein product in skeletal muscle was confirmed by immunohistochemistry and label-free quantitative proteomics analysis; muscle degeneration was confirmed in biopsy and necropsy samples. Elevated skeletal muscle glycogen persisted after harvest, resulting in a high pH and dark-cutting beef, which is negatively perceived by consumers and results in an economic loss to the industry. Carriers of the variant did not exhibit differences in meat quality or any measures of animal well-being. CONCLUSIONS: Myophosphorylase deficiency poses welfare concerns for affected animals and negatively impacts the final product. The association of the recessive genotype with dark-cutting beef further demonstrates the importance of genetics to not only animal health but to the quality of their product. Although cattle heterozygous for the variant may not immediately affect the beef industry, identifying carriers will enable selection and breeding strategies to prevent the production of affected calves.


Assuntos
Estudo de Associação Genômica Ampla , Glicogênio Fosforilase Muscular , Animais , Bovinos , Feminino , Masculino , Doenças dos Bovinos/genética , Genes Recessivos , Glicogênio Fosforilase Muscular/genética , Glicogênio Fosforilase Muscular/deficiência , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Linhagem , Polimorfismo de Nucleotídeo Único , Sequenciamento Completo do Genoma
13.
Neurogenetics ; 25(2): 79-83, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38240911

RESUMO

Narcolepsy with cataplexy is a complex disease with both genetic and environmental risk factors. To gain further insight into the homozygous HCRT-related narcolepsy, we present a case series of five patients from two consanguineous families, each harboring a novel homozygous variant of HCRT c.17_18del. All affected individuals exhibited severe cataplexy accompanied by narcolepsy symptoms during infancy. Additionally, cataplexy symptoms improved or disappeared in the majority of patients over time. Pathogenic variants in HCRT cause autosomal recessive narcolepsy with cataplexy. Genetic testing of the HCRT gene should be conducted in specific subgroups of narcolepsy, particularly those with early onset, familial cases, and a predominantly cataplexy phenotype.


Assuntos
Narcolepsia , Linhagem , Adolescente , Adulto , Criança , Feminino , Humanos , Masculino , Alelos , Cataplexia/genética , Consanguinidade , Genes Recessivos , Homozigoto , Mutação/genética , Narcolepsia/genética , Orexinas/genética , Fenótipo
14.
Hum Genet ; 143(3): 311-329, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38459354

RESUMO

Identification of genes associated with nonsyndromic hearing loss is a crucial endeavor given the substantial number of individuals who remain without a diagnosis after even the most advanced genetic testing. PKHD1L1 was established as necessary for the formation of the cochlear hair-cell stereociliary coat and causes hearing loss in mice and zebrafish when mutated. We sought to determine if biallelic variants in PKHD1L1 also cause hearing loss in humans. Exome sequencing was performed on DNA of four families segregating autosomal recessive nonsyndromic sensorineural hearing loss. Compound heterozygous p.[(Gly129Ser)];p.[(Gly1314Val)] and p.[(Gly605Arg)];p[(Leu2818TyrfsTer5)], homozygous missense p.(His2479Gln) and nonsense p.(Arg3381Ter) variants were identified in PKHD1L1 that were predicted to be damaging using in silico pathogenicity prediction methods. In vitro functional analysis of two missense variants was performed using purified recombinant PKHD1L1 protein fragments. We then evaluated protein thermodynamic stability with and without the missense variants found in one of the families and performed a minigene splicing assay for another variant. In silico molecular modeling using AlphaFold2 and protein sequence alignment analysis were carried out to further explore potential variant effects on structure. In vitro functional assessment indicated that both engineered PKHD1L1 p.(Gly129Ser) and p.(Gly1314Val) mutant constructs significantly reduced the folding and structural stabilities of the expressed protein fragments, providing further evidence to support pathogenicity of these variants. Minigene assay of the c.1813G>A p.(Gly605Arg) variant, located at the boundary of exon 17, revealed exon skipping leading to an in-frame deletion of 48 amino acids. In silico molecular modeling exposed key structural features that might suggest PKHD1L1 protein destabilization. Multiple lines of evidence collectively associate PKHD1L1 with nonsyndromic mild-moderate to severe sensorineural hearing loss. PKHD1L1 testing in individuals with mild-moderate hearing loss may identify further affected families.


Assuntos
Surdez , Mutação de Sentido Incorreto , Linhagem , Receptores de Superfície Celular , Estereocílios , Animais , Feminino , Humanos , Masculino , Surdez/genética , Sequenciamento do Exoma , Genes Recessivos , Perda Auditiva Neurossensorial/genética , Perda Auditiva Neurossensorial/patologia , Modelos Moleculares , Receptores de Superfície Celular/genética , Estereocílios/metabolismo , Estereocílios/patologia , Estereocílios/genética
15.
Am J Hum Genet ; 108(5): 764-785, 2021 05 06.
Artigo em Inglês | MEDLINE | ID: mdl-33811808

RESUMO

Tandem repeats represent one of the most abundant class of variations in human genomes, which are polymorphic by nature and become highly unstable in a length-dependent manner. The expansion of repeat length across generations is a well-established process that results in human disorders mainly affecting the central nervous system. At least 50 disorders associated with expansion loci have been described to date, with half recognized only in the last ten years, as prior methodological difficulties limited their identification. These limitations still apply to the current widely used molecular diagnostic methods (exome or gene panels) and thus result in missed diagnosis detrimental to affected individuals and their families, especially for disorders that are very rare and/or clinically not recognizable. Most of these disorders have been identified through family-driven approaches and many others likely remain to be identified. The recent development of long-read technologies provides a unique opportunity to systematically investigate the contribution of tandem repeats and repeat expansions to the genetic architecture of human disorders. In this review, we summarize the current and most recent knowledge about the genetics of repeat expansion disorders and the diversity of their pathophysiological mechanisms and outline the perspectives of developing personalized treatments in the future.


Assuntos
Pesquisa Biomédica/tendências , Expansão das Repetições de Trinucleotídeos , Antecipação Genética , Efeito Fundador , Genes Dominantes , Genes Recessivos , Genoma Humano/genética , Humanos , Fatores de Tempo , Expansão das Repetições de Trinucleotídeos/genética
16.
Am J Hum Genet ; 108(7): 1318-1329, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34077761

RESUMO

TP73 belongs to the TP53 family of transcription factors and has therefore been well studied in cancer research. Studies in mice, however, have revealed non-oncogenic activities related to multiciliogenesis. Utilizing whole-exome sequencing analysis in a cohort of individuals with a mucociliary clearance disorder and cortical malformation, we identified homozygous loss-of-function variants in TP73 in seven individuals from five unrelated families. All affected individuals exhibit a chronic airway disease as well as a brain malformation consistent with lissencephaly. We performed high-speed video microscopy, immunofluorescence analyses, and transmission electron microscopy in respiratory epithelial cells after spheroid or air liquid interface culture to analyze ciliary function, ciliary length, and number of multiciliated cells (MCCs). The respiratory epithelial cells studied display reduced ciliary length and basal bodies mislocalized within the cytoplasm. The number of MCCs is severely reduced, consistent with a reduced number of cells expressing the transcription factors crucial for multiciliogenesis (FOXJ1, RFX2). Our data demonstrate that autosomal-recessive deleterious variants in the TP53 family member TP73 cause a mucociliary clearance disorder due to a defect in MCC differentiation.


Assuntos
Lisencefalia/genética , Depuração Mucociliar/genética , Mucosa Respiratória/metabolismo , Proteína Tumoral p73/genética , Diferenciação Celular/genética , Células Cultivadas , Ciliopatias/genética , Genes Recessivos , Homozigoto , Humanos , Mutação com Perda de Função , Microscopia de Vídeo , Mucosa Respiratória/citologia , Mucosa Respiratória/ultraestrutura , Sequenciamento do Exoma
17.
Am J Hum Genet ; 108(4): 608-619, 2021 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-33740458

RESUMO

The number and distribution of recessive alleles in the population for various diseases are not known at genome-wide-scale. Based on 6,447 exome sequences of healthy, genetically unrelated Europeans of two distinct ancestries, we estimate that every individual is a carrier of at least 2 pathogenic variants in currently known autosomal-recessive (AR) genes and that 0.8%-1% of European couples are at risk of having a child affected with a severe AR genetic disorder. This risk is 16.5-fold higher for first cousins but is significantly more increased for skeletal disorders and intellectual disabilities due to their distinct genetic architecture.


Assuntos
Consanguinidade , Características da Família , Genes Recessivos/genética , Variação Genética/genética , Fenótipo , População Branca/genética , Estudos de Coortes , Europa (Continente)/etnologia , Exoma/genética , Feminino , Testes Genéticos , Saúde , Heterozigoto , Humanos , Deficiência Intelectual/genética , Masculino
18.
Am J Hum Genet ; 108(1): 186-193, 2021 01 07.
Artigo em Inglês | MEDLINE | ID: mdl-33417887

RESUMO

POLR3B encodes the second-largest catalytic subunit of RNA polymerase III, an enzyme involved in transcription. Bi-allelic pathogenic variants in POLR3B are a well-established cause of hypomyelinating leukodystrophy. We describe six unrelated individuals with de novo missense variants in POLR3B and a clinical presentation substantially different from POLR3-related leukodystrophy. These individuals had afferent ataxia, spasticity, variable intellectual disability and epilepsy, and predominantly demyelinating sensory motor peripheral neuropathy. Protein modeling and proteomic analysis revealed a distinct mechanism of pathogenicity; the de novo POLR3B variants caused aberrant association of individual enzyme subunits rather than affecting overall enzyme assembly or stability. We expand the spectrum of disorders associated with pathogenic variants in POLR3B to include a de novo heterozygous POLR3B-related disorder.


Assuntos
Ataxia/genética , Doenças Desmielinizantes Hereditárias do Sistema Nervoso Central/genética , RNA Polimerase III/genética , Adolescente , Adulto , Ataxia Cerebelar/genética , Criança , Pré-Escolar , Feminino , Genes Recessivos/genética , Heterozigoto , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Proteômica/métodos , Adulto Jovem
19.
Am J Hum Genet ; 108(7): 1301-1317, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34038740

RESUMO

Human C2orf69 is an evolutionarily conserved gene whose function is unknown. Here, we report eight unrelated families from which 20 children presented with a fatal syndrome consisting of severe autoinflammation and progredient leukoencephalopathy with recurrent seizures; 12 of these subjects, whose DNA was available, segregated homozygous loss-of-function C2orf69 variants. C2ORF69 bears homology to esterase enzymes, and orthologs can be found in most eukaryotic genomes, including that of unicellular phytoplankton. We found that endogenous C2ORF69 (1) is loosely bound to mitochondria, (2) affects mitochondrial membrane potential and oxidative respiration in cultured neurons, and (3) controls the levels of the glycogen branching enzyme 1 (GBE1) consistent with a glycogen-storage-associated mitochondriopathy. We show that CRISPR-Cas9-mediated inactivation of zebrafish C2orf69 results in lethality by 8 months of age due to spontaneous epileptic seizures, which is preceded by persistent brain inflammation. Collectively, our results delineate an autoinflammatory Mendelian disorder of C2orf69 deficiency that disrupts the development/homeostasis of the immune and central nervous systems.


Assuntos
Encefalite/genética , Doenças Mitocondriais/genética , Animais , Evolução Biológica , Sistemas CRISPR-Cas , Linhagem Celular , Encefalite/mortalidade , Feminino , Genes Recessivos , Glicogênio/metabolismo , Humanos , Inflamação/genética , Masculino , Proteínas de Membrana/genética , Doenças Mitocondriais/mortalidade , Linhagem , Convulsões/genética , Convulsões/mortalidade , Peixe-Zebra/genética
20.
N Engl J Med ; 385(14): 1292-1301, 2021 09 30.
Artigo em Inglês | MEDLINE | ID: mdl-34587386

RESUMO

BACKGROUND: Structural birth defects occur in approximately 3% of live births; most such defects lack defined genetic or environmental causes. Despite advances in surgical approaches, pharmacologic prevention remains largely out of reach. METHODS: We queried worldwide databases of 20,248 families that included children with neurodevelopmental disorders and that were enriched for parental consanguinity. Approximately one third of affected children in these families presented with structural birth defects or microcephaly. We performed exome or genome sequencing of samples obtained from the children, their parents, or both to identify genes with biallelic pathogenic or likely pathogenic mutations present in more than one family. After identifying disease-causing variants, we generated two mouse models, each with a pathogenic variant "knocked in," to study mechanisms and test candidate treatments. We administered a small-molecule Wnt agonist to pregnant animals and assessed their offspring. RESULTS: We identified homozygous mutations in WLS, which encodes the Wnt ligand secretion mediator (also known as Wntless or WLS) in 10 affected persons from 5 unrelated families. (The Wnt ligand secretion mediator is essential for the secretion of all Wnt proteins.) Patients had multiorgan defects, including microcephaly and facial dysmorphism as well as foot syndactyly, renal agenesis, alopecia, iris coloboma, and heart defects. The mutations affected WLS protein stability and Wnt signaling. Knock-in mice showed tissue and cell vulnerability consistent with Wnt-signaling intensity and individual and collective functions of Wnts in embryogenesis. Administration of a pharmacologic Wnt agonist partially restored embryonic development. CONCLUSIONS: Genetic variations affecting a central Wnt regulator caused syndromic structural birth defects. Results from mouse models suggest that what we have named Zaki syndrome is a potentially preventable disorder. (Funded by the National Institutes of Health and others.).


Assuntos
Anormalidades Múltiplas/genética , Anormalidades Congênitas/genética , Pleiotropia Genética , Peptídeos e Proteínas de Sinalização Intracelular/genética , Mutação , Receptores Acoplados a Proteínas G/genética , Proteínas Wnt/metabolismo , Animais , Modelos Animais de Doenças , Fibroblastos/metabolismo , Técnicas de Introdução de Genes , Genes Recessivos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Camundongos Transgênicos , Linhagem , Fenótipo , Receptores Acoplados a Proteínas G/metabolismo , Síndrome , Via de Sinalização Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA