Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 86
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.
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
3.
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
4.
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
5.
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 , Fácies , 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
6.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
BMC Musculoskelet Disord ; 21(1): 392, 2020 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-32571287

RESUMO

BACKGROUND: Butterfly vertebrae are a rare congenital vertebral anomaly. An overlap of this spinal anomaly with other diseases has been reported. However, to the authors' knowledge, the coexistence of butterfly vertebrae and spinal cord injury has not been reported in the literature. CASE PRESENTATION: A 42-year-old male was admitted to our emergency department after a motor vehicle accident. His complaint was back pain, and he was unable to move both lower limbs. Upon physical examination, the patient was not ambulatory. Sensory examination revealed the absence of sensation below the T12 level. The strength of the bilateral lower limbs was grade 0. The patient received a radiographic evaluation. The initial diagnosis was T11 fracture with complete paraplegia of the lower limbs. Magnetic resonance imaging (MRI) was then performed. Sagittal MRI demonstrated an isointense lesion on T1-weighted imaging and a high-signal spindle-like lesion on T2-weighted imaging of the spinal cord adjacent to the T11 vertebra. The fat-suppressed sequence also revealed hyperintensities of the cord. There was no evidence of acute injury of the T11 vertebral body except for cuneiform anterior wedging. The patient was ultimately diagnosed with complete paraplegia with a T11 butterfly vertebra. He underwent urgent posterior decompressive and fixation surgery from T10 to T12. His postoperative recovery was uneventful. CONCLUSIONS: The coexistence of a butterfly vertebra with spinal cord injury was reported for the first time. Although butterfly vertebrae may be incidentally detected, it is important to be familiar with their radiographic features to distinguish them from fractures.


Assuntos
Acidentes de Trânsito , Anormalidades Musculoesqueléticas/patologia , Paraplegia/etiologia , Traumatismos da Medula Espinal/diagnóstico , Vértebras Torácicas/anormalidades , Adulto , Dor nas Costas/etiologia , Dor nas Costas/patologia , Descompressão Cirúrgica , Fixação Interna de Fraturas , Humanos , Imageamento por Ressonância Magnética , Masculino , Anormalidades Musculoesqueléticas/diagnóstico por imagem , Traumatismos da Medula Espinal/patologia , Traumatismos da Medula Espinal/cirurgia , Vértebras Torácicas/diagnóstico por imagem , Tomografia Computadorizada por Raios X , Resultado do Tratamento
13.
Nat Commun ; 11(1): 3168, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32576830

RESUMO

In humans, mutations in the PIEZO2 gene, which encodes for a mechanosensitive ion channel, were found to result in skeletal abnormalities including scoliosis and hip dysplasia. Here, we show in mice that loss of Piezo2 expression in the proprioceptive system recapitulates several human skeletal abnormalities. While loss of Piezo2 in chondrogenic or osteogenic lineages does not lead to human-like skeletal abnormalities, its loss in proprioceptive neurons leads to spine malalignment and hip dysplasia. To validate the non-autonomous role of proprioception in hip joint morphogenesis, we studied this process in mice mutant for proprioceptive system regulators Runx3 or Egr3. Loss of Runx3 in the peripheral nervous system, but not in skeletal lineages, leads to similar joint abnormalities, as does Egr3 loss of function. These findings expand the range of known regulatory roles of the proprioception system on the skeleton and provide a central component of the underlying molecular mechanism, namely Piezo2.


Assuntos
Canais Iônicos/metabolismo , Anormalidades Musculoesqueléticas/metabolismo , Sistema Musculoesquelético/metabolismo , Neurônios/metabolismo , Propriocepção/fisiologia , Anormalidades Múltiplas , Animais , Remodelação Óssea , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Modelos Animais de Doenças , Proteína 3 de Resposta de Crescimento Precoce/metabolismo , Predisposição Genética para Doença/genética , Luxação do Quadril/genética , Luxação do Quadril/metabolismo , Luxação do Quadril/patologia , Articulação do Quadril/anatomia & histologia , Articulação do Quadril/metabolismo , Articulação do Quadril/patologia , Canais Iônicos/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Sistema Musculoesquelético/patologia , Escoliose
14.
Am J Med Genet A ; 182(8): 1944-1946, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32432408

RESUMO

Dental anomalies and short stature (DASS) has been recently identified as a distinct entity, associated with bi-allelic hypomorphic variants in LTBP3 gene. Only 20 individuals from nine families have been previously reported, with a consistent phenotype of short stature, brachyolmia, and amelogenesis imperfecta. We report the first case from India, with novel radiographic and molecular findings in LTBP3 gene, thereby expanding the phenotypic spectrum of DASS.


Assuntos
Amelogênese Imperfeita/genética , Nanismo/genética , Proteínas de Ligação a TGF-beta Latente/genética , Anormalidades Musculoesqueléticas/genética , Alelos , Amelogênese Imperfeita/patologia , Criança , Nanismo/patologia , Feminino , Humanos , Mutação com Perda de Função/genética , Masculino , Anormalidades Musculoesqueléticas/patologia , Mutação/genética , Osteocondrodisplasias/genética , Osteocondrodisplasias/patologia , Linhagem , Fenótipo
16.
Mol Genet Genomic Med ; 8(3): e1064, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31943948

RESUMO

BACKGROUND: In the last few years trio-whole exome sequencing (WES) analysis has demonstrated its potential in obtaining genetic diagnoses even in nonspecific clinical pictures and in atypical presentations of known diseases. Moreover WES allows the detection of variants in multiple genes causing different genetic conditions in a single patient, in about 5% of cases. The resulting phenotype may be clinically discerned as variability in the expression of a known phenotype, or as a new unreported syndromic condition. METHODS: Trio-WES was performed on a 4-month-old baby with a complex clinical presentation characterized by skeletal anomalies, congenital heart malformation, congenital hypothyroidism, generalized venous and arterial hypoplasia, and recurrent infections. RESULTS: WES detected two different homozygous variants, one in CEP57, the gene responsible for mosaic variegated aneuploidy syndrome 2, the other in DYNC2H1, the main gene associated with short-rib thoracic dysplasia. CONCLUSION: The contribution of these two different genetic causes in determining the phenotype of our patient is discussed, including some clinical signs not explained by the detected variants. The report then highlights the role of WES in providing complete and fast diagnosis in patients with complex presentations of rare genetic syndromes, with important implications in the assessment of recurrence risk.


Assuntos
Dineínas do Citoplasma/genética , Cardiopatias Congênitas/genética , Hipotireoidismo/genética , Proteínas Associadas aos Microtúbulos/genética , Anormalidades Musculoesqueléticas/genética , Proteínas Nucleares/genética , Fenótipo , Cardiopatias Congênitas/patologia , Homozigoto , Humanos , Hipotireoidismo/patologia , Lactente , Masculino , Mosaicismo , Anormalidades Musculoesqueléticas/patologia , Mutação , Síndrome , Sequenciamento do Exoma
17.
Clin Genet ; 97(2): 362-369, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31600839

RESUMO

Aymé-Gripp syndrome (AYGRPS) is a recognizable condition caused by a restricted spectrum of dominantly acting missense mutations affecting the transcription factor MAF. Major clinical features of AYGRPS include congenital cataracts, sensorineural hearing loss, intellectual disability, and a distinctive flat facial appearance. Skeletal abnormalities have also been observed in affected individuals, even though these features have not been assessed systematically. Expanding the series with four additional patients, here we provide a more accurate delineation of the molecular aspects and clinical phenotype, particularly focusing on the skeletal features characterizing this disorder. Apart from previously reported malar flattening and joint limitations, we document that carpal/tarsal and long bone defects, and hip dysplasia occur in affected subjects more frequently than formerly appreciated.


Assuntos
Catarata/genética , Predisposição Genética para Doença , Transtornos do Crescimento/genética , Perda Auditiva Neurossensorial/genética , Deficiência Intelectual/genética , Anormalidades Musculoesqueléticas/genética , Proteínas Proto-Oncogênicas c-maf/genética , Adolescente , Adulto , Catarata/patologia , Criança , Pré-Escolar , Fácies , Feminino , Transtornos do Crescimento/patologia , Perda Auditiva Neurossensorial/patologia , Humanos , Lactente , Deficiência Intelectual/patologia , Masculino , Anormalidades Musculoesqueléticas/patologia , Mutação de Sentido Incorreto/genética , Adulto Jovem
18.
J Mol Neurosci ; 70(1): 1-8, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31428919

RESUMO

This study presents two Chinese siblings with a rare neurodevelopmental disorder (NDD) caused by biallelic INTS1 mutations and investigates the clinical features of this disease by means of in silico analysis. Two siblings, an 11-year-old brother and a 5-year-old sister, visited our hospital due to physical retardation and profound intellectual disability. Whole-exome sequencing (WES) was performed for the girl, and Sanger sequencing was used to validate the identified variants. Phenotype correlation analysis and in silico genetic interaction network analysis were performed to investigate genes that could lead to diseases similar to the rare disease in the patients. Growth retardation, distinct intellectual disability, hypertelorism, mild cataract, uneven teeth, abnormal palmar and plantar creases, and dubious genitalia were noted in the sister. No neurological features related to neuropathy were found. The brother showed features and growth delay similar to his sister. Heterozygous novel variants of c.1645A>G,p.Met549Val and c.5881C>T,p.Gln1961* in INTS1 were considered a candidate etiology. Sanger sequencing demonstrated that the variants were inherited from the grandfather and (maternal) grandmother. Phenotype correlation analysis revealed that CTDP1 mutation-induced congenital cataracts-facial dysmorphism-neuropathy (CCFDN) mostly overlapped with the performance of our patients. In silico analysis of the genetic interaction network showed that INTS1 is highly associated with INTS8 and CTDP1. Our study further validated that biallelic INTS1 mutations could bring about the onset of a novel neurodevelopmental disorder.


Assuntos
Deficiência Intelectual/genética , Anormalidades Musculoesqueléticas/genética , Mutação , Transtornos do Neurodesenvolvimento/genética , Proteína Wnt1/genética , Criança , Feminino , Humanos , Deficiência Intelectual/patologia , Masculino , Anormalidades Musculoesqueléticas/patologia , Transtornos do Neurodesenvolvimento/patologia , Fenótipo , Irmãos , Síndrome
19.
Eur Spine J ; 28(9): 1977-1986, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31079250

RESUMO

PURPOSE: To determine the relationship between the bone formation-related functions of GPR126 and the structural asymmetry of spine in adolescent idiopathic scoliosis (AIS). METHODS: Vertebral body samples were obtained from 51 AIS patients during spinal surgery between October 2014 and November 2017, and the expression pattern of GPR126 in the convex/concave sides of AIS spine was identified by RT-qPCR. Next, we explored the bone formation-related functions of GPR126 by knocking down and overexpressing GPR126 in human mesenchymal stem cells (hMSC) and further performing osteogenic differentiation. We also applied overexpression of N-terminal fragments derived from GPR126 (GPR126-NTFs) and osteogenic differentiation experiments to determine the functional part of GPR126 in skeletal development. RESULTS: We provided evidence that GPR126 showed a marked convex/concave asymmetric expression in the spine of AIS. Further RNA detection found that exon6-included transcripts of GPR126 (GPR126-exon6in) were significantly higher expressed in the convex side of AIS patients. Knocking down of GPR126 accelerated ossification of hMSCs during osteogenic differentiation, and overexpression of GPR126-exon6in delayed this process. Overexpression of GPR126-NTFs revealed that NTF is a functional fragment and exon6-included NTF (NTF-exon6in) delayed ossification of hMSCs. CONCLUSION: Our findings indicated that GPR126-NTFs play a role in skeletal development, and the inclusion/exclusion of exon6 may regulate the bone formation-related functions of GPR126. The convex/concave asymmetric expression of GPR126-exon6in may be an important factor in abnormal bone formation of AIS. These slides can be retrieved under Electronic Supplementary Material.


Assuntos
Anormalidades Musculoesqueléticas/metabolismo , Receptores Acoplados a Proteínas G/biossíntese , Escoliose/metabolismo , Adolescente , Diferenciação Celular/fisiologia , Células Cultivadas , Criança , Feminino , Regulação da Expressão Gênica/fisiologia , Técnicas de Silenciamento de Genes , Humanos , Masculino , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Anormalidades Musculoesqueléticas/genética , Anormalidades Musculoesqueléticas/patologia , Osteogênese/genética , Osteogênese/fisiologia , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/fisiologia , Escoliose/genética , Escoliose/patologia , Escoliose/cirurgia , Coluna Vertebral/patologia , Coluna Vertebral/fisiopatologia
20.
Am J Hum Genet ; 104(3): 422-438, 2019 03 07.
Artigo em Inglês | MEDLINE | ID: mdl-30773277

RESUMO

SPONASTRIME dysplasia is an autosomal-recessive spondyloepimetaphyseal dysplasia characterized by spine (spondylar) abnormalities, midface hypoplasia with a depressed nasal bridge, metaphyseal striations, and disproportionate short stature. Scoliosis, coxa vara, childhood cataracts, short dental roots, and hypogammaglobulinemia have also been reported in this disorder. Although an autosomal-recessive inheritance pattern has been hypothesized, pathogenic variants in a specific gene have not been discovered in individuals with SPONASTRIME dysplasia. Here, we identified bi-allelic variants in TONSL, which encodes the Tonsoku-like DNA repair protein, in nine subjects (from eight families) with SPONASTRIME dysplasia, and four subjects (from three families) with short stature of varied severity and spondylometaphyseal dysplasia with or without immunologic and hematologic abnormalities, but no definitive metaphyseal striations at diagnosis. The finding of early embryonic lethality in a Tonsl-/- murine model and the discovery of reduced length, spinal abnormalities, reduced numbers of neutrophils, and early lethality in a tonsl-/- zebrafish model both support the hypomorphic nature of the identified TONSL variants. Moreover, functional studies revealed increased amounts of spontaneous replication fork stalling and chromosomal aberrations, as well as fewer camptothecin (CPT)-induced RAD51 foci in subject-derived cell lines. Importantly, these cellular defects were rescued upon re-expression of wild-type (WT) TONSL; this rescue is consistent with the hypothesis that hypomorphic TONSL variants are pathogenic. Overall, our studies in humans, mice, zebrafish, and subject-derived cell lines confirm that pathogenic variants in TONSL impair DNA replication and homologous recombination-dependent repair processes, and they lead to a spectrum of skeletal dysplasia phenotypes with numerous extra-skeletal manifestations.


Assuntos
Instabilidade Cromossômica , Dano ao DNA , Variação Genética , Anormalidades Musculoesqueléticas/patologia , NF-kappa B/genética , Osteocondrodisplasias/patologia , Adolescente , Adulto , Alelos , Animais , Células Cultivadas , Criança , Pré-Escolar , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Estudos de Associação Genética , Humanos , Camundongos , Camundongos Knockout , Anormalidades Musculoesqueléticas/genética , Osteocondrodisplasias/genética , Sequenciamento do Exoma , Adulto Jovem , Peixe-Zebra
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA