Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 150
Filtrar
1.
Urology ; 177: 178-180, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36804444

RESUMO

Wilms tumor is the most common pediatric renal mass and occurs in up to 10% of predisposition syndromes. One such syndrome is CLOVES syndrome, an extremely rare disorder within the umbrella of PIK3CA-related overgrowth spectrum disorders. This case presents the management of a bilateral Wilms tumor in a patient with CLOVES syndrome and highlights the many intricacies in caring for complex oncology patients. Particularly highlighted in this case is the delicate line in balancing the risks of treatment-related morbidity against the risks of recurrence in predisposed patients, while still abiding by established treatment guidelines.


Assuntos
Neoplasias Renais , Lipoma , Anormalidades Musculoesqueléticas , Tumor de Wilms , Criança , Humanos , Tumor de Wilms/complicações , Tumor de Wilms/patologia , Anormalidades Musculoesqueléticas/patologia , Lipoma/patologia , Síndrome , Neoplasias Renais/complicações , Neoplasias Renais/patologia
2.
Rehabilitación (Madr., Ed. impr.) ; 56(1): 64-73, Ene - Mar 2022. ilus
Artigo em Espanhol | IBECS | ID: ibc-204890

RESUMO

La aplicación de ondas de choque focales y de ondas de presión radial en patología musculoesquelética ha tenido un gran desarrollo y difusión en la última década. Si bien la mayoría de las publicaciones han resaltado su seguridad y eficacia, no están exentas de malos resultados y complicaciones. Esta revisión analiza las principales causas de los malos resultados, efectos adversos y complicaciones, haciendo énfasis en su prevención.(AU)


The application of focused shockwaves and radial pressure waves in musculoskeletal pathology has had a great development in the last decade. Although most of the publications have highlighted their safety and efficacy, poor results and complications can occur. This review analyzes the main causes of its poor results, adverse effects, and complications, emphasizing their prevention.(AU)


Assuntos
Humanos , Ondas de Choque de Alta Energia , Doenças Musculoesqueléticas , Anormalidades Musculoesqueléticas , Fenômenos Fisiológicos Musculoesqueléticos , Doenças Musculoesqueléticas/patologia , Anormalidades Musculoesqueléticas/patologia , Morbidade , Erros de Diagnóstico , Erros Médicos
3.
Proc Natl Acad Sci U S A ; 118(39)2021 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-34548398

RESUMO

Skeletal ciliopathies (e.g., Jeune syndrome, short rib polydactyly syndrome, and Sensenbrenner syndrome) are frequently associated with nephronophthisis-like cystic kidney disease and other organ manifestations. Despite recent progress in genetic mapping of causative loci, a common molecular mechanism of cartilage defects and cystic kidneys has remained elusive. Targeting two ciliary chondrodysplasia loci (ift80 and ift172) by CRISPR/Cas9 mutagenesis, we established models for skeletal ciliopathies in Xenopus tropicalis Froglets exhibited severe limb deformities, polydactyly, and cystic kidneys, closely matching the phenotype of affected patients. A data mining-based in silico screen found ttc30a to be related to known skeletal ciliopathy genes. CRISPR/Cas9 targeting replicated limb malformations and renal cysts identical to the models of established disease genes. Loss of Ttc30a impaired embryonic renal excretion and ciliogenesis because of altered posttranslational tubulin acetylation, glycylation, and defective axoneme compartmentalization. Ttc30a/b transcripts are enriched in chondrocytes and osteocytes of single-cell RNA-sequenced embryonic mouse limbs. We identify TTC30A/B as an essential node in the network of ciliary chondrodysplasia and nephronophthisis-like disease proteins and suggest that tubulin modifications and cilia segmentation contribute to skeletal and renal ciliopathy manifestations of ciliopathies in a cell type-specific manner. These findings have implications for potential therapeutic strategies.


Assuntos
Osso e Ossos/anormalidades , Ciliopatias/patologia , Craniossinostoses/patologia , Proteínas do Citoesqueleto/metabolismo , Displasia Ectodérmica/patologia , Embrião não Mamífero/patologia , Anormalidades Musculoesqueléticas/patologia , Doenças Renais Policísticas/patologia , Tubulina (Proteína)/química , Animais , Osso e Ossos/metabolismo , Osso e Ossos/patologia , Ciliopatias/genética , Ciliopatias/metabolismo , Craniossinostoses/genética , Craniossinostoses/metabolismo , Proteínas do Citoesqueleto/genética , Modelos Animais de Doenças , Displasia Ectodérmica/genética , Displasia Ectodérmica/metabolismo , Embrião não Mamífero/metabolismo , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/metabolismo , Fenótipo , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Tubulina (Proteína)/metabolismo , Xenopus laevis
4.
Am J Hum Genet ; 108(9): 1669-1691, 2021 09 02.
Artigo em Inglês | MEDLINE | ID: mdl-34314705

RESUMO

Transportin-2 (TNPO2) mediates multiple pathways including non-classical nucleocytoplasmic shuttling of >60 cargoes, such as developmental and neuronal proteins. We identified 15 individuals carrying de novo coding variants in TNPO2 who presented with global developmental delay (GDD), dysmorphic features, ophthalmologic abnormalities, and neurological features. To assess the nature of these variants, functional studies were performed in Drosophila. We found that fly dTnpo (orthologous to TNPO2) is expressed in a subset of neurons. dTnpo is critical for neuronal maintenance and function as downregulating dTnpo in mature neurons using RNAi disrupts neuronal activity and survival. Altering the activity and expression of dTnpo using mutant alleles or RNAi causes developmental defects, including eye and wing deformities and lethality. These effects are dosage dependent as more severe phenotypes are associated with stronger dTnpo loss. Interestingly, similar phenotypes are observed with dTnpo upregulation and ectopic expression of TNPO2, showing that loss and gain of Transportin activity causes developmental defects. Further, proband-associated variants can cause more or less severe developmental abnormalities compared to wild-type TNPO2 when ectopically expressed. The impact of the variants tested seems to correlate with their position within the protein. Specifically, those that fall within the RAN binding domain cause more severe toxicity and those in the acidic loop are less toxic. Variants within the cargo binding domain show tissue-dependent effects. In summary, dTnpo is an essential gene in flies during development and in neurons. Further, proband-associated de novo variants within TNPO2 disrupt the function of the encoded protein. Hence, TNPO2 variants are causative for neurodevelopmental abnormalities.


Assuntos
Deficiências do Desenvolvimento/genética , Proteínas de Drosophila/genética , Oftalmopatias Hereditárias/genética , Deficiência Intelectual/genética , Carioferinas/genética , Anormalidades Musculoesqueléticas/genética , beta Carioferinas/genética , Proteína ran de Ligação ao GTP/genética , Alelos , Sequência de Aminoácidos , Animais , Deficiências do Desenvolvimento/metabolismo , Deficiências do Desenvolvimento/patologia , Proteínas de Drosophila/antagonistas & inibidores , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/genética , Drosophila melanogaster/crescimento & desenvolvimento , Drosophila melanogaster/metabolismo , Oftalmopatias Hereditárias/metabolismo , Oftalmopatias Hereditárias/patologia , Feminino , Dosagem de Genes , Regulação da Expressão Gênica no Desenvolvimento , Genoma Humano , Humanos , Lactente , Recém-Nascido , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Carioferinas/antagonistas & inibidores , Carioferinas/metabolismo , Masculino , Anormalidades Musculoesqueléticas/metabolismo , Anormalidades Musculoesqueléticas/patologia , Mutação , Neurônios/metabolismo , Neurônios/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos , Sequenciamento Completo do Genoma , beta Carioferinas/metabolismo , Proteína ran de Ligação ao GTP/metabolismo
5.
PLoS Genet ; 17(6): e1009605, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34081701

RESUMO

Homozygous mutation of the Csf1r locus (Csf1rko) in mice, rats and humans leads to multiple postnatal developmental abnormalities. To enable analysis of the mechanisms underlying the phenotypic impacts of Csf1r mutation, we bred a rat Csf1rko allele to the inbred dark agouti (DA) genetic background and to a Csf1r-mApple reporter transgene. The Csf1rko led to almost complete loss of embryonic macrophages and ablation of most adult tissue macrophage populations. We extended previous analysis of the Csf1rko phenotype to early postnatal development to reveal impacts on musculoskeletal development and proliferation and morphogenesis in multiple organs. Expression profiling of 3-week old wild-type (WT) and Csf1rko livers identified 2760 differentially expressed genes associated with the loss of macrophages, severe hypoplasia, delayed hepatocyte maturation, disrupted lipid metabolism and the IGF1/IGF binding protein system. Older Csf1rko rats developed severe hepatic steatosis. Consistent with the developmental delay in the liver Csf1rko rats had greatly-reduced circulating IGF1. Transfer of WT bone marrow (BM) cells at weaning without conditioning repopulated resident macrophages in all organs, including microglia in the brain, and reversed the mutant phenotypes enabling long term survival and fertility. WT BM transfer restored osteoclasts, eliminated osteopetrosis, restored bone marrow cellularity and architecture and reversed granulocytosis and B cell deficiency. Csf1rko rats had an elevated circulating CSF1 concentration which was rapidly reduced to WT levels following BM transfer. However, CD43hi non-classical monocytes, absent in the Csf1rko, were not rescued and bone marrow progenitors remained unresponsive to CSF1. The results demonstrate that the Csf1rko phenotype is autonomous to BM-derived cells and indicate that BM contains a progenitor of tissue macrophages distinct from hematopoietic stem cells. The model provides a unique system in which to define the pathways of development of resident tissue macrophages and their local and systemic roles in growth and organ maturation.


Assuntos
Fígado Gorduroso/genética , Macrófagos/metabolismo , Anormalidades Musculoesqueléticas/genética , Desenvolvimento Musculoesquelético/genética , Osteopetrose/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Animais , Medula Óssea/metabolismo , Medula Óssea/patologia , Transplante de Medula Óssea , Modelos Animais de Doenças , Embrião de Mamíferos , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Fígado Gorduroso/terapia , Feminino , Regulação da Expressão Gênica no Desenvolvimento , Técnicas de Inativação de Genes , Genes Reporter , Humanos , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/deficiência , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/genética , Fator de Crescimento Insulin-Like I/deficiência , Fator de Crescimento Insulin-Like I/genética , Metabolismo dos Lipídeos , Fígado/metabolismo , Fígado/patologia , Macrófagos/patologia , Masculino , Anormalidades Musculoesqueléticas/metabolismo , Anormalidades Musculoesqueléticas/patologia , Anormalidades Musculoesqueléticas/terapia , Osteopetrose/metabolismo , Osteopetrose/patologia , Osteopetrose/terapia , Ratos , Ratos Transgênicos , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/deficiência
6.
Am J Med Genet A ; 185(10): 3083-3091, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34061450

RESUMO

KCNT2 variants resulting in substitutions affecting the Arg190 residue have been shown to cause epileptic encephalopathy and a recognizable facial gestalt. We report two additional individuals with intellectual disability, dysmorphic features, hypertrichosis, macrocephaly and the same de novo KCNT2 missense variants affecting the Arg190 residue as previously described. Notably, neither patient has epilepsy. Homology modeling of these missense variants revealed that they are likely to disrupt the stabilization of a closed channel conformation of KCNT2 resulting in a constitutively open state. This is the first report of pathogenic variants in KCNT2 causing a developmental phenotype without epilepsy.


Assuntos
Epilepsia/genética , Hipertricose/genética , Deficiência Intelectual/genética , Megalencefalia/genética , Canais de Potássio Ativados por Sódio/genética , Adolescente , Arginina/genética , Criança , Pré-Escolar , Epilepsia/diagnóstico , Epilepsia/patologia , Feminino , Predisposição Genética para Doença , Humanos , Hipertricose/diagnóstico , Hipertricose/diagnóstico por imagem , Hipertricose/patologia , Deficiência Intelectual/diagnóstico por imagem , Deficiência Intelectual/patologia , Megalencefalia/diagnóstico por imagem , Megalencefalia/patologia , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Mutação de Sentido Incorreto/genética , Fenótipo
7.
Eur J Med Genet ; 64(7): 104226, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33872773

RESUMO

Different mutations in the Growth/Differentiation Factor 5 gene (GDF5) have been associated with varying types of skeletal dysplasia, including Grebe type chondrodysplasia (GTC), Hunter-Thompson syndrome, Du Pan Syndrome and Brachydactyly type C (BDC). Heterozygous pathogenic mutations exert milder effects, whereas homozygous mutations are known to manifest more severe phenotypes. In this study, we report a GDF5 frameshift mutation (c.404delC) segregating over six generations in an extended consanguineous Pakistani family. The family confirmed that both GTC and BDC are part of the GDF5 mutational spectrum, with severe GTC associated with homozygosity, and with a wide phenotypic variability among heterozygous carriers, ranging from unaffected non-penetrant carriers, to classical BDC and to novel unclassified types of brachydactylies.


Assuntos
Braquidactilia/genética , Fator 5 de Diferenciação de Crescimento/genética , Anormalidades Musculoesqueléticas/genética , Osteocondrodisplasias/genética , Braquidactilia/patologia , Feminino , Mutação da Fase de Leitura , Heterozigoto , Homozigoto , Humanos , Masculino , Anormalidades Musculoesqueléticas/patologia , Osteocondrodisplasias/patologia , Linhagem
8.
Am J Med Genet A ; 185(6): 1925-1931, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33704902

RESUMO

Variants of the diphthamide biosynthesis I (DPH1, OMIM*603527) are associated with developmental delay, short stature, and sparse hair syndrome (DEDSSH/DPH1 syndrome) (OMIM# 616901). Another name is Loucks-Innes syndrome. DPH1 syndrome is an ultrarare and severe neurodevelopmental disorder. Less than 20 patients were reported from different ethnicities. Here, we described the first Chinese adult with genetically confirmed DPH1 syndrome. We summarized previously reported patients in the literature and found that developmental delay, unusual skull shape, sparse hair, and facial dysmorphism were consistently present in all DPH1 syndrome patients. Dysplastic toenails and dental abnormalities are age-dependent characteristics of DPH1 syndrome. Our patient was the first reported patient with documented growth hormone deficiency. Dental and endocrine checkup should be considered in the routine follow-up of DPH1 syndrome patients.


Assuntos
Deficiências do Desenvolvimento/genética , Nanismo Hipofisário/genética , Antígenos de Histocompatibilidade Menor/genética , Transtornos do Neurodesenvolvimento/genética , Proteínas Supressoras de Tumor/genética , Adulto , Deficiências do Desenvolvimento/patologia , Nanismo Hipofisário/patologia , Humanos , Masculino , Anormalidades Musculoesqueléticas/diagnóstico , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Mutação/genética , Transtornos do Neurodesenvolvimento/patologia
9.
Am J Med Genet A ; 185(6): 1822-1835, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33765349

RESUMO

Monozygotic (MZ) twins ("identical twins") are essentially unique to human beings. Why and how they arise is not known. This article reviews the possible different types of MZ twinning recognized in the previous article on twins and arthrogryposis. There appear to be at least three subgroups of MZ twinning: spontaneous, familial, and those related to artificial reproductive technologies. Each is likely to have different etiologies and different secondary findings. Spontaneous MZ twinning may relate to "overripe ova." Amyoplasia, a specific nongenetic form of arthrogryposis, appears to occur in spontaneous MZ twinning and may be related to twin-twin transfusion.


Assuntos
Artrogripose/genética , Transfusão Feto-Fetal/genética , Anormalidades Musculoesqueléticas/genética , Gemelaridade Monozigótica/genética , Artrogripose/complicações , Artrogripose/epidemiologia , Artrogripose/patologia , Doenças em Gêmeos/epidemiologia , Doenças em Gêmeos/genética , Doenças em Gêmeos/patologia , Feminino , Transfusão Feto-Fetal/complicações , Transfusão Feto-Fetal/epidemiologia , Transfusão Feto-Fetal/patologia , Humanos , Recém-Nascido , Masculino , Anormalidades Musculoesqueléticas/complicações , Anormalidades Musculoesqueléticas/epidemiologia , Anormalidades Musculoesqueléticas/patologia , Gravidez , Gêmeos Dizigóticos/genética , Gêmeos Monozigóticos/genética
10.
Am J Med Genet A ; 185(6): 1816-1821, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33760374

RESUMO

Amyoplasia is a very specific, nongenetic clinically recognizable form of arthrogryposis, representing about one-third of individuals with arthrogryposis surviving the newborn period. There is a markedly increased number of individuals with Amyoplasia who are one of monozygotic (MZ) twins, with the other twin being normal. Thus, it would appear that Amyoplasia is definitely associated with and may be caused by an MZ twinning event. The twin-twin transfusion seen in MZ twins could play an etiologic role in producing Amyoplasia. In this article, Amyoplasia twinning is compared to twinning in other forms of arthrogryposis. The accompanying paper examines various types of MZ twinning (Hall, 2021). Amyoplasia is primarily associated with spontaneous MZ twinning.


Assuntos
Artrogripose/genética , Doenças em Gêmeos/genética , Transfusão Feto-Fetal/induzido quimicamente , Anormalidades Musculoesqueléticas/genética , Artrogripose/complicações , Artrogripose/patologia , Doenças em Gêmeos/epidemiologia , Doenças em Gêmeos/patologia , Feminino , Transfusão Feto-Fetal/complicações , Humanos , Recém-Nascido , Masculino , Anormalidades Musculoesqueléticas/complicações , Anormalidades Musculoesqueléticas/patologia , Gravidez , Gemelaridade Monozigótica , Gêmeos Dizigóticos/genética , Gêmeos Monozigóticos/genética
11.
Am J Med Genet A ; 185(5): 1486-1493, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33683002

RESUMO

The RASopathies are a group of similar genetic syndromes with cardiovascular abnormalities, characteristic facial features, short stature, abnormalities of the skin and musculoskeletal system, and variable neurodevelopmental challenges. The most common cardiovascular abnormalities include pulmonary valvular stenosis and hypertrophic cardiomyopathy. Congenital polyvalvular disease (CPVD) refers to congenital dysplasia of two or more cardiac valves. We diagnosed a RASopathy in two individuals with CPVD and noted that CPVD in RASopathies has rarely been reported in the literature. Thus, we performed a retrospective chart review and literature review to investigate the association and characterize the phenotype of CPVD in the RASopathies. CPVD was present in 2.5% (n = 6/243) of individuals in our RASopathy cohort. Involvement of two cardiac valves, commonly the aortic and pulmonic valves, was seen in the majority of individuals (6/8; 75%) in our cohort, but only 27% (3/11) of reported CPVD and RASopathy cases in the literature. CPVD should be considered an associated cardiovascular phenotype of the RASopathies, which has implications for diagnosis and management.


Assuntos
Cardiomiopatia Hipertrófica/genética , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Proto-Oncogênicas B-raf/genética , Estenose da Valva Pulmonar/genética , Adolescente , Valva Aórtica/patologia , Cardiomiopatia Hipertrófica/epidemiologia , Cardiomiopatia Hipertrófica/patologia , Anormalidades Cardiovasculares/epidemiologia , Anormalidades Cardiovasculares/genética , Anormalidades Cardiovasculares/patologia , Criança , Pré-Escolar , Nanismo/genética , Nanismo/patologia , Facies , Feminino , Cardiopatias Congênitas/genética , Cardiopatias Congênitas/patologia , Humanos , Lactente , Recém-Nascido , Masculino , Anormalidades Musculoesqueléticas/epidemiologia , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Síndrome de Noonan , Fenótipo , Estenose da Valva Pulmonar/epidemiologia , Estenose da Valva Pulmonar/patologia , Anormalidades da Pele/genética , Anormalidades da Pele/patologia , Proteínas ras/genética
12.
Eur J Med Genet ; 64(4): 104181, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33647455

RESUMO

In a consanguineous Pakistani kinship afflicted with mild to moderate intellectual disability (ID), mild lissencephaly, brain atrophy and skeletal anomalies, we detected homozygous CRADD c.2T > G (p.Met1?) and USP44 c.873_886delinsT (p.Leu291Phefs*8), two good candidates 1.85-Mb apart that segregated with the disorder. Biallelic damaging variants in CRADD cause recessive mental retardation-34 (MRT34; MIM 614499) with mild to moderate ID, "thin" lissencephaly, and variable megalencephaly and seizures. For USP44, only a single ID family has been reported with a homozygous deleterious variant, which is the same as the variant we detected. In affected individuals we present, at ages 29-32 years, clinical findings are similar yet not fully concordant with phenotypes for either gene considering the skeletal findings, and ID is not as severe as would be expected for defects in two genes with additive effect. Some variable CRADD-related features such as language impairment and seizures are not observed in the presented family. The presence of the two variants in the family is a very rare example of familial linked homozygous variants, and whether the damaging USP44 variant contributed to the disease in the family we present is not clear. As for the skeletal findings, facial dysmorphism and digestive problems, we did not find a candidate variant. This study is an example of both clinical variation and difficulty in variant detection and evaluation. Our findings highlight that even an extensive exome sequence analysis can fail to fully uncover the complex molecular basis of a syndrome even if potentially causative variants are identified.


Assuntos
Proteína Adaptadora de Sinalização CRADD/genética , Deficiências do Desenvolvimento/genética , Deficiência Intelectual/genética , Lisencefalia/genética , Anormalidades Musculoesqueléticas/genética , Estrabismo/genética , Ubiquitina Tiolesterase/genética , Adulto , Consanguinidade , Deficiências do Desenvolvimento/patologia , Feminino , Humanos , Deficiência Intelectual/patologia , Lisencefalia/patologia , Masculino , Anormalidades Musculoesqueléticas/patologia , Mutação , Linhagem , Estrabismo/patologia , Síndrome
13.
Am J Med Genet A ; 185(3): 866-870, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33300650

RESUMO

Infantile liver failure syndrome type 1 (ILFS1) is a recently recognized autosomal recessive disorder caused by deleterious mutations in the leucyl-tRNA synthetase 1 gene (LARS1). The LARS1 enzyme is responsible for incorporation of the amino acid leucine during protein polypeptide synthesis. Individuals with LARS1 mutations typically show liver failure from infancy to early childhood during periods of illness or other physiological stress. While 25 patients from 15 families with ILFS1 have been reported in the literature, histological reports from autopsy findings are limited. We report here a premature male neonate who presented with severe intrauterine growth retardation, microcytic anemia, and fulminant liver failure, and who was a compound heterozygote for two novel deleterious mutations in LARS1. An autopsy showed fulminant hepatitis-like hepatocellular injury and fibrogenesis in the liver and a lack of uniformity in skeletal muscle, accompanied by the disruption of striated muscle fibers. Striking dysgenesis in skeletal muscle detected in the present case indicates the effect of LARS1 functional deficiency on the musculature. Whole-exome sequencing may be useful for neonates with unexplained early liver failure if extensive genetic and metabolic testing is inconclusive.


Assuntos
Doenças do Prematuro/genética , Leucina-tRNA Ligase/genética , Falência Hepática/genética , Anormalidades Musculoesqueléticas/genética , Mutação de Sentido Incorreto , Mutação Puntual , Sítios de Splice de RNA/genética , Substituição de Aminoácidos , Anemia Neonatal/genética , Éxons/genética , Evolução Fatal , Retardo do Crescimento Fetal/genética , Genes Recessivos , Heterozigoto , Humanos , Hiperbilirrubinemia Neonatal/genética , Recém-Nascido , Recém-Nascido Prematuro , Doenças do Prematuro/patologia , Íntrons/genética , Leucina-tRNA Ligase/deficiência , Cirrose Hepática/etiologia , Falência Hepática/patologia , Falência Hepática Aguda/etiologia , Falência Hepática Aguda/patologia , Masculino , Insuficiência de Múltiplos Órgãos/etiologia , Músculo Esquelético/patologia , Anormalidades Musculoesqueléticas/patologia , Alinhamento de Sequência , Síndrome , Sequenciamento do Exoma
14.
Am J Med Genet A ; 185(2): 370-376, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33179433

RESUMO

Klippel-Feil syndrome 4 (KFS4; MIM# 616549) is an autosomal recessive disorder caused by biallelic pathogenic variants in MYO18B and comprises, in addition to Klippel-Feil anomaly (KFA), nemaline myopathy, facial dysmorphism, and short stature. We aim to outline the natural history of KFS4 and provide an updated description of its clinical, radiological, laboratory, and molecular findings. We comprehensively analyzed the medical records of 6 Saudi and 1 American patients (including 5 previously unpublished cases) with a molecularly confirmed diagnosis of KFS4. All patients had myopathy of varying severity that followed a slowly progressive or non-progressive course, affecting primarily the proximal musculature of the lower limb although hand involvement with distal arthrogryposis and abnormal interphalangeal creases was also observed. KFA and characteristic dysmorphic features, including ptosis and bulbous nose, were observed in all but two patients. The causal MYO18B variants were a founder NM_032608.5:c.6905C>A; p.(Ser2302*) variant in the Saudi patients (P1-P6) and a novel MYO18B homozygous variant (c.6660_6670del;p.[Arg2220Serfs*74]) in the American Caucasian patient (P7). We report the phenotypic and genetic findings in seven patients with KFS4. We describe the natural history of this disease, confirm myopathy as a universal feature and describe its pattern and progression, and note interesting differences between the phenotypes observed in patients with KFA and those without.


Assuntos
Cardiomiopatias/genética , Síndrome de Klippel-Feil/genética , Miopatias da Nemalina/genética , Miosinas/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Adulto , Artrogripose/complicações , Cardiomiopatias/complicações , Cardiomiopatias/patologia , Criança , Pré-Escolar , Face/anormalidades , Face/patologia , Feminino , Predisposição Genética para Doença , Homozigoto , Humanos , Lactente , Síndrome de Klippel-Feil/complicações , Síndrome de Klippel-Feil/patologia , Masculino , Anormalidades Musculoesqueléticas/complicações , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Miopatias da Nemalina/complicações , Miopatias da Nemalina/patologia , Linhagem , Fenótipo , Adulto Jovem
15.
Semin Pediatr Surg ; 29(5): 150974, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33069285

RESUMO

Overgrowth syndromes represent a diverse group of disorders with overlapping features. Interdisciplinary management by a team of experts in vascular anomalies is crucial for establishing the correct diagnosis and optimizing outcomes for these patients. Unique management considerations include increased risk for thrombosis and in some cases, cancer. In recent years, research has demonstrated that these disorders are primarily caused by somatic mutations in growth pathways, particularly the PI3K-mTOR pathway. This improved understanding had led to promising new therapies for this group of patients.


Assuntos
Síndrome do Hamartoma Múltiplo , Síndrome de Klippel-Trenaunay-Weber , Lipoma , Anormalidades Musculoesqueléticas , Nevo , Síndrome de Proteu , Síndrome de Sturge-Weber , Malformações Vasculares , Criança , Síndrome do Hamartoma Múltiplo/genética , Síndrome do Hamartoma Múltiplo/patologia , Síndrome do Hamartoma Múltiplo/terapia , Humanos , Síndrome de Klippel-Trenaunay-Weber/genética , Síndrome de Klippel-Trenaunay-Weber/patologia , Síndrome de Klippel-Trenaunay-Weber/terapia , Lipoma/genética , Lipoma/patologia , Lipoma/terapia , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Anormalidades Musculoesqueléticas/terapia , Nevo/genética , Nevo/patologia , Nevo/terapia , Síndrome de Proteu/genética , Síndrome de Proteu/patologia , Síndrome de Proteu/terapia , Síndrome de Sturge-Weber/genética , Síndrome de Sturge-Weber/patologia , Síndrome de Sturge-Weber/terapia , Malformações Vasculares/genética , Malformações Vasculares/patologia , Malformações Vasculares/terapia
16.
Semin Pediatr Surg ; 29(5): 150973, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-33069292

RESUMO

Vascular anomalies impact the musculoskeletal system dependent on the tissue involved (skin, subcutis, muscle, cartilage, or bone), the extent of involvement, and the type of anomalous vessels (arteries, capillaries, veins, or lymphatics). These malformations can cause a multitude of musculoskeletal problems for the patient. Leg-length discrepancy, intra-articular involvement, muscular lesions, and primary or secondary scoliosis are amongst the issues that patients face. All of these problems can cause pain, deformity, and a range of functional limitations. Surgical and nonsurgical treatment plans have a role in patient care. Patients with vascular anomalies may also suffer from life-threatening cardiovascular and hematologic abnormalities. For those patients who undergo surgery, the thromboembolic risk is elevated, wound breakdown and infection are much more common, and bleeding risk continues well into the postoperative course. Because of the complex nature of these disorders, the clinician must have a full understanding of the types of lesions, their natural history, appropriate diagnostic studies, associated medical problems, indications for treatment, and treatment options. For severe malformations, especially syndromes such as CLOVES and Klippel- Trenaunay syndrome, interdisciplinary team management is essential for the best outcomes.


Assuntos
Síndrome de Klippel-Trenaunay-Weber , Lipoma , Anormalidades Musculoesqueléticas , Nevo , Malformações Vasculares , Criança , Humanos , Síndrome de Klippel-Trenaunay-Weber/complicações , Síndrome de Klippel-Trenaunay-Weber/diagnóstico , Síndrome de Klippel-Trenaunay-Weber/patologia , Síndrome de Klippel-Trenaunay-Weber/terapia , Lipoma/complicações , Lipoma/diagnóstico , Lipoma/patologia , Lipoma/terapia , Anormalidades Musculoesqueléticas/complicações , Anormalidades Musculoesqueléticas/diagnóstico , Anormalidades Musculoesqueléticas/patologia , Anormalidades Musculoesqueléticas/terapia , Nevo/complicações , Nevo/diagnóstico , Nevo/patologia , Nevo/terapia , Malformações Vasculares/complicações , Malformações Vasculares/diagnóstico , Malformações Vasculares/patologia , Malformações Vasculares/terapia
17.
Am J Hum Genet ; 107(4): 763-777, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32937143

RESUMO

Distal hereditary motor neuropathies (HMNs) and axonal Charcot-Marie-Tooth neuropathy (CMT2) are clinically and genetically heterogeneous diseases characterized primarily by motor neuron degeneration and distal weakness. The genetic cause for about half of the individuals affected by HMN/CMT2 remains unknown. Here, we report the identification of pathogenic variants in GBF1 (Golgi brefeldin A-resistant guanine nucleotide exchange factor 1) in four unrelated families with individuals affected by sporadic or dominant HMN/CMT2. Genomic sequencing analyses in seven affected individuals uncovered four distinct heterozygous GBF1 variants, two of which occurred de novo. Other known HMN/CMT2-implicated genes were excluded. Affected individuals show HMN/CMT2 with slowly progressive distal muscle weakness and musculoskeletal deformities. Electrophysiological studies confirmed axonal damage with chronic neurogenic changes. Three individuals had additional distal sensory loss. GBF1 encodes a guanine-nucleotide exchange factor that facilitates the activation of members of the ARF (ADP-ribosylation factor) family of small GTPases. GBF1 is mainly involved in the formation of coatomer protein complex (COPI) vesicles, maintenance and function of the Golgi apparatus, and mitochondria migration and positioning. We demonstrate that GBF1 is present in mouse spinal cord and muscle tissues and is particularly abundant in neuropathologically relevant sites, such as the motor neuron and the growth cone. Consistent with the described role of GBF1 in Golgi function and maintenance, we observed marked increase in Golgi fragmentation in primary fibroblasts derived from all affected individuals in this study. Our results not only reinforce the existing link between Golgi fragmentation and neurodegeneration but also demonstrate that pathogenic variants in GBF1 are associated with HMN/CMT2.


Assuntos
Axônios/metabolismo , Doença de Charcot-Marie-Tooth/genética , Fatores de Troca do Nucleotídeo Guanina/genética , Debilidade Muscular/genética , Atrofia Muscular Espinal/genética , Anormalidades Musculoesqueléticas/genética , Adulto , Idoso , Idoso de 80 Anos ou mais , Sequência de Aminoácidos , Animais , Axônios/patologia , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/patologia , Doença de Charcot-Marie-Tooth/diagnóstico , Doença de Charcot-Marie-Tooth/metabolismo , Doença de Charcot-Marie-Tooth/patologia , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/patologia , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Heterozigoto , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Debilidade Muscular/diagnóstico , Debilidade Muscular/metabolismo , Debilidade Muscular/patologia , Atrofia Muscular Espinal/diagnóstico , Atrofia Muscular Espinal/metabolismo , Atrofia Muscular Espinal/patologia , Anormalidades Musculoesqueléticas/diagnóstico , Anormalidades Musculoesqueléticas/metabolismo , Anormalidades Musculoesqueléticas/patologia , Mutação , Linhagem , Cultura Primária de Células , Medula Espinal/anormalidades , Medula Espinal/metabolismo
18.
Clin Genet ; 98(6): 606-612, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32812661

RESUMO

RBM10, is an RNA binding protein that is important for development by regulating the expression of multiple genes. RBM10 is on the X chromosome, and nonsense and frameshift RBM10 variants cause TARP syndrome in males. In a 4-year-old male, we identified a novel maternally inherited missense RBM10 variant in the RRM2 RNA binding domain, c.965C>T, p.Pro322Leu. His clinical features included intellectual disability, developmental delay, growth restriction, hypotonia, and craniofacial malformations. These features were much milder than those described in previously reported cases of TARP syndrome. By in vitro assays, we found that the mutant p.Pro322Leu RBM10 protein retained its specific RNA binding capacity, while gaining a low-affinity nonspecific RNA binding. It was normally localized to the nucleus, but its expression level was significantly reduced with a significantly short half-life. These results indicated that the p.Pro322Leu missense variant causes a developmental disorder in humans through a unique loss-of-function mechanism.


Assuntos
Pé Torto Equinovaro/genética , Deficiências do Desenvolvimento/genética , Predisposição Genética para Doença , Cardiopatias Congênitas/genética , Síndrome de Pierre Robin/genética , Proteínas de Ligação a RNA/genética , Pré-Escolar , Pé Torto Equinovaro/complicações , Pé Torto Equinovaro/patologia , Anormalidades Craniofaciais/complicações , Anormalidades Craniofaciais/genética , Anormalidades Craniofaciais/patologia , Deficiências do Desenvolvimento/complicações , Deficiências do Desenvolvimento/patologia , Cardiopatias Congênitas/complicações , Cardiopatias Congênitas/patologia , Humanos , Deficiência Intelectual/complicações , Deficiência Intelectual/genética , Deficiência Intelectual/patologia , Masculino , Anormalidades Musculoesqueléticas/complicações , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Mutação de Sentido Incorreto/genética , Fenótipo , Síndrome de Pierre Robin/complicações , Síndrome de Pierre Robin/patologia , Sequenciamento do Exoma
19.
Biochem Biophys Res Commun ; 530(3): 520-526, 2020 09 24.
Artigo em Inglês | MEDLINE | ID: mdl-32620236

RESUMO

PIK3CA-related overgrowth spectrum is caused by mosaicism mutations in the PIK3CA gene. These mutations, which are also observed in various types of cancer, lead to a constitutive activation of the PI3K/AKT/mTOR pathway, increasing cell proliferation. Heat shock transcription factor 1 (HSF1) is the major stress-responsive transcription factor. Recent findings indicate that AKT phosphorylates and activates HSF1 independently of heat-shock in breast cancer cells. Here, we aimed to investigate the role of HSF1 in PIK3CA-related overgrowth spectrum. We observed a higher rate of proliferation and increased phosphorylation of AKT and p70S6K in mutant fibroblasts than in control cells. We also found elevated phosphorylation and activation of HSF1, which is directly correlated to AKT activation. Specific AKT inhibitors inhibit HSF1 phosphorylation as well as HSF1-dependent gene transcription. Finally, we demonstrated that targeting HSF1 with specific inhibitors reduced the proliferation of mutant cells. As there is currently no curative treatment for PIK3CA-related overgrowth spectrum, our results identify HSF1 as a new potential therapeutic target.


Assuntos
Proliferação de Células/efeitos dos fármacos , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Descoberta de Drogas , Fatores de Transcrição de Choque Térmico/antagonistas & inibidores , Lipoma/metabolismo , Anormalidades Musculoesqueléticas/metabolismo , Nevo/metabolismo , Malformações Vasculares/metabolismo , Células Cultivadas , Classe I de Fosfatidilinositol 3-Quinases/genética , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibroblastos/patologia , Fatores de Transcrição de Choque Térmico/metabolismo , Humanos , Lipoma/tratamento farmacológico , Lipoma/genética , Lipoma/patologia , Terapia de Alvo Molecular , Anormalidades Musculoesqueléticas/tratamento farmacológico , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Mutação , Nevo/tratamento farmacológico , Nevo/genética , Nevo/patologia , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Malformações Vasculares/tratamento farmacológico , Malformações Vasculares/genética , Malformações Vasculares/patologia
20.
Clin Genet ; 98(2): 191-197, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32530092

RESUMO

Structural eye disorders are increasingly recognised as having a genetic basis, although current genetic testing is limited in its success. De novo missense variants in WDR37 are a recently described cause of a multisystemic syndromic disorder featuring ocular coloboma. This study characterises the phenotypic spectrum of this disorder and reports 2 de novo heterozygous variants (p.Thr115Ile, p.Ser119Tyr) in three unrelated Caucasian individuals. All had a clinical phenotype consisting of bilateral iris and retinal coloboma, developmental delay and additional, variable multisystem features. The variants fall within a highly conserved region upstream of the WD-repeat domains, within an apparent mutation cluster. Consistent with the literature, intellectual disability, structural eye disorders, epilepsy, congenital heart disease, genitorenal anomalies and dysmorphic facial features were observed. In addition, a broader developmental profile is reported with a more specific musculoskeletal phenotype described in association with the novel variant (p.Thr115Ile). We further expand the phenotypic spectrum of WDR37-related disorders to include those with milder developmental delay and strengthen the association of ocular coloboma and musculoskeletal features. We promote the inclusion of WDR37 on gene panels for intellectual disability, epilepsy and structural eye disorders.


Assuntos
Coloboma/genética , Oftalmopatias/genética , Deficiência Intelectual/genética , Anormalidades Musculoesqueléticas/genética , Proteínas Nucleares/genética , Adolescente , Adulto , Criança , Pré-Escolar , Coloboma/complicações , Coloboma/patologia , Epilepsia/complicações , Epilepsia/genética , Epilepsia/patologia , Oftalmopatias/complicações , Oftalmopatias/patologia , Feminino , Humanos , Lactente , Recém-Nascido , Deficiência Intelectual/complicações , Deficiência Intelectual/patologia , Masculino , Anormalidades Musculoesqueléticas/complicações , Anormalidades Musculoesqueléticas/patologia , Mutação/genética , Mutação de Sentido Incorreto/genética , Fenótipo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...