Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Genes (Basel) ; 14(4)2023 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-37107635

RESUMO

SHOX deficiency is a common genetic cause of short stature of variable degree. SHOX haploinsufficiency causes Leri-Weill dyschondrosteosis (LWD) as well as nonspecific short stature. SHOX haploinsufficiency is known to result from heterozygous loss-of-function variants with pseudo-autosomal dominant inheritance, while biallelic SHOX loss-of-function variants cause the more severe skeletal dysplasia, Langer mesomelic dyschondrosteosis (LMD). Here we report for the first time the pseudo-autosomal recessive inheritance of LWD in two siblings caused by a novel homozygous non-canonical, leaky splice-site variant in intron 3 of SHOX: c.544+5G>C. Transcript analyses in patient-derived fibroblasts showed homozygous patients to produce approximately equal amounts of normally spliced mRNA and mRNA with the abnormal retention of intron 3 and containing a premature stop codon (p.Val183Glyfs*31). The aberrant transcript was shown to undergo nonsense-mediated mRNA decay, and thus resulting in SHOX haploinsufficiency in the homozygous patient. Six healthy relatives who are of normal height are heterozygous for this variant and fibroblasts from a heterozygote for the c.544+5G>C variant produced wild-type transcript amounts comparable to healthy control. The unique situation reported here highlights the fact that the dosage of SHOX determines the clinical phenotype rather than the Mendelian inheritance pattern of SHOX variants. This study extends the molecular and inheritance spectrum of SHOX deficiency disorder and highlights the importance of functional testing of SHOX variants of unknown significance in order to allow appropriate counseling and precision medicine for each family individual.


Assuntos
Nanismo , Osteocondrodisplasias , Humanos , Proteínas de Homeodomínio/genética , Proteína de Homoeobox de Baixa Estatura/genética , Transtornos do Crescimento/genética , Osteocondrodisplasias/genética , Osteocondrodisplasias/complicações , Nanismo/genética
2.
Hepatol Commun ; 6(7): 1611-1619, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35271763

RESUMO

Wilson disease (WD) is caused by biallelic pathogenic variants in adenosine triphosphatase copper-transporting beta (ATP7B); however, genetic testing identifies only one or no pathogenic ATP7B variant in a number of patients with WD. Synonymous single-nucleotide sequence variants have been recognized as pathogenic in individual families. The aim of the present study was to evaluate the prevalence and disease mechanism of the synonymous variant c.2292C>T (p.Phe764=) in WD. A cohort of 280 patients with WD heterozygous for a single ATP7B variant was investigated for the presence of c.2292C>T (p.Phe764=). In this cohort of otherwise genetically unexplained WD, the allele frequency of c.2292C>T (p.Phe764=) was 2.5% (14 of 560) compared to 7.1 × 10-6 in the general population (2 of 280,964 in the Genome Aggregation Database; p < 10-5 ; Fisher exact test). In an independent United Kingdom (UK) cohort, 2 patients with WD homozygous for p.Phe764= were identified. RNA analysis of ATP7B transcripts from patients homozygous or heterozygous for c.2292C>T and control fibroblasts showed that this variant caused high expression of an ATP7B transcript variant lacking exon 8. Conclusion: The synonymous ATP7B variant c.2292C>T (p.Phe764=) causes abnormal messenger RNA processing of ATP7B transcripts and is associated with WD in compound heterozygotes and homozygotes.


Assuntos
Degeneração Hepatolenticular , Cobre/metabolismo , ATPases Transportadoras de Cobre/genética , Éxons/genética , Degeneração Hepatolenticular/genética , Humanos , Mutação/genética , Mutação Silenciosa
3.
Genes (Basel) ; 12(11)2021 10 20.
Artigo em Inglês | MEDLINE | ID: mdl-34828254

RESUMO

Biallelic truncating FAM149B1 variants result in cilia dysfunction and have been reported in four infants with Joubert syndrome and orofaciodigital syndrome type VI, respectively. We report here on three adult siblings, 18 to 40 years of age, homozygous for the known FAM149B1 c.354_357delinsCACTC (p.Gln118Hisfs*20) variant. Detailed clinical examinations were performed including ocular and gait analyses, skeletal- and neuroimaging. All three patients presented with neurological and oculomotor symptoms since birth and mild skeletal dysplasia in infancy resulting in characteristic gait abnormalities. We document mild skeletal dysplasia, abnormal gait with increased hip rotation and increased external foot rotation, ataxia, variable polydactyly, ocular Duane syndrome, progressive ophthalmoplegia, nystagmus, situs inversus of the retinal vessels, olfactory bulb aplasia, and corpus callosal dysgenesis as novel features in FAM149B1-ciliopathy. We show that intellectual disability is mild to moderate and retinal, renal and liver function is normal in these affected adults. Our study thus expands the FAM149B1-related Joubert syndrome to a mainly neurological and skeletal ciliopathy phenotype with predominant oculomotor dysfunction but otherwise stable outcome in adults. Diagnosis of FAM149B1-related disorder was impeded by segregation of multiple neurogenetic disorders in the same family, highlighting the importance of extended clinical and genetic studies in families with complex phenotypes.


Assuntos
Anormalidades Múltiplas/genética , Cerebelo/anormalidades , Ciliopatias/genética , Proteínas do Citoesqueleto/genética , Anormalidades do Olho/genética , Doenças Renais Císticas/genética , Malformações do Sistema Nervoso/genética , Retina/anormalidades , Anormalidades Múltiplas/diagnóstico , Adolescente , Adulto , Ciliopatias/diagnóstico , Consanguinidade , Síndrome da Retração Ocular/complicações , Síndrome da Retração Ocular/diagnóstico , Síndrome da Retração Ocular/genética , Anormalidades do Olho/complicações , Feminino , Humanos , Doenças Renais Císticas/complicações , Masculino , Malformações do Sistema Nervoso/complicações , Malformações do Sistema Nervoso/diagnóstico , Fenótipo , Arábia Saudita , Irmãos , Adulto Jovem
4.
Clin Transl Gastroenterol ; 12(11): e00427, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34797252

RESUMO

INTRODUCTION: Gain-of-function mutations in guanylyl cyclase C (GCC) result in persistent diarrhea with perinatal onset. We investigated a specific GCC inhibitor, SSP2518, for its potential to treat this disorder. METHODS: We investigated the effect of SSP2518 on GCC-mediated intracellular cyclic guanosine monophosphate (cGMP) levels and on GCC-mediated chloride secretion in intestinal organoids from 3 patients with distinct activating GCC mutations and from controls, with and without stimulation of GCC with heat-stable enterotoxin. RESULTS: Patient-derived organoids had significantly higher basal cGMP levels than control organoids, which were lowered by SSP2518 to levels found in control organoids. In addition, SSP2518 significantly reduced cGMP levels and chloride secretion in patient-derived and control organoids (P < 0.05 for all comparisons) after heat-stable enterotoxin stimulation. DISCUSSION: We reported in this study that the GCC inhibitor SSP2518 normalizes cGMP levels in intestinal organoids derived from patients with GCC gain-of-function mutations and markedly reduces cystic fibrosis transmembrane conductance regulator-dependent chloride secretion, the driver of persistent diarrhea.


Assuntos
Anormalidades Múltiplas/tratamento farmacológico , Anormalidades Múltiplas/genética , Diarreia/congênito , Erros Inatos do Metabolismo/tratamento farmacológico , Erros Inatos do Metabolismo/genética , Receptores de Enterotoxina/antagonistas & inibidores , Anormalidades Múltiplas/metabolismo , GMP Cíclico/metabolismo , Diarreia/tratamento farmacológico , Diarreia/genética , Diarreia/metabolismo , Mutação com Ganho de Função , Compostos Heterocíclicos de 4 ou mais Anéis/uso terapêutico , Humanos , Erros Inatos do Metabolismo/metabolismo , Receptores de Enterotoxina/genética
5.
Hepatology ; 74(5): 2424-2435, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34048062

RESUMO

BACKGROUND AND AIMS: High serum ferritin is frequent among patients with chronic liver disease and commonly associated with hepatic iron overload. Genetic causes of high liver iron include homozygosity for the p.Cys282Tyr variant in homeostatic iron regulator (HFE) and rare variants in non-HFE genes. The aims of the present study were to describe the landscape and frequency of mutations in hemochromatosis genes and determine whether patient selection by noninvasive hepatic iron quantification using MRI improves the diagnostic yield of next-generation sequencing (NGS) in patients with hyperferritinemia. APPROACH AND RESULTS: A cohort of 410 unselected liver clinic patients with high serum ferritin (defined as ≥200 µg/L for women and ≥300 µg/L for men) was investigated by HFE genotyping and abdominal MRI R2*. Forty-one (10%) patients were homozygous for the p.Cys282Tyr variant in HFE. Of the remaining 369 patients, 256 (69%) had high transferrin saturation (TSAT; ≥45%) and 199 (53%) had confirmed hepatic iron overload (liver R2* ≥70 s-1 ). NGS of hemochromatosis genes was carried out in 180 patients with hepatic iron overload, and likely pathogenic variants were identified in 68 of 180 (38%) patients, mainly in HFE (79%), ceruloplasmin (25%), and transferrin receptor 2 (19%). Low spleen iron (R2* <50 s-1 ), but not TSAT, was significantly associated with the presence of mutations. In 167 patients (93%), no monogenic cause of hepatic iron overload could be identified. CONCLUSIONS: In patients without homozygosity for p.Cys282Tyr, coincident pathogenic variants in HFE and non-HFE genes could explain hyperferritinemia with hepatic iron overload in a subset of patients. Unlike HFE hemochromatosis, this type of polygenic hepatic iron overload presents with variable TSAT. High ferritin in blood is an indicator of the iron storage disease, hemochromatosis. A simple genetic test establishes this diagnosis in the majority of patients affected. MRI of the abdomen can guide further genetic testing.


Assuntos
Proteína da Hemocromatose/genética , Hemocromatose/diagnóstico por imagem , Hemocromatose/genética , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Ferro/metabolismo , Hepatopatias/diagnóstico por imagem , Hepatopatias/genética , Imageamento por Ressonância Magnética/métodos , Seleção de Pacientes , Fenótipo , Adulto , Idoso , Ceruloplasmina/genética , Feminino , Ferritinas/sangue , Seguimentos , Testes Genéticos , Genótipo , Hemocromatose/sangue , Homozigoto , Humanos , Fígado/diagnóstico por imagem , Fígado/metabolismo , Fígado/patologia , Hepatopatias/sangue , Masculino , Pessoa de Meia-Idade , Mutação , Receptores da Transferrina/genética , Estudos Retrospectivos
6.
J Crohns Colitis ; 15(10): 1679-1685, 2021 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-33770165

RESUMO

BACKGROUND: Congenital chloride diarrhoea [CLD] is a rare autosomal recessive disease caused by mutations in the solute family carrier 26 member 3 [SLC26A3] gene. Patients suffer from life-long watery diarrhoea and chloride loss. Inflammatory bowel disease [IBD] has been reported in individual patients with CLD and in scl26a3-deficient mice. METHODS: We performed an international multicentre analysis to build a CLD cohort and to identify cases with IBD. We assessed clinical and genetic characteristics of subjects and studied the cumulative incidence of CLD-associated IBD. RESULTS: In a cohort of 72 patients with CLD caused by 17 different SLC26A3 mutations, we identified 12 patients [17%] diagnosed with IBD. Nine patients had Crohn's disease, two ulcerative colitis and one IBD-unclassified [IBD-U]. The prevalence of IBD in our cohort of CLD was higher than the highest prevalence of IBD in Europe [p < 0.0001]. The age of onset was variable [13.5 years, interquartile range: 8.5-23.5 years]. Patients with CLD and IBD had lower z-score for height than those without IBD. Four of 12 patients had required surgery [ileostomy formation n = 2, ileocaecal resection due to ileocaecal valve stenosis n = 1 and colectomy due to stage II transverse colon cancer n = 1]. At last follow-up, 5/12 were on biologics [adalimumab, infliximab or vedolizumab], 5/12 on immunosuppressants [azathioprine or mercaptopurine], one on 5-ASA and one off-treatment. CONCLUSIONS: A substantial proportion of patients with CLD develop IBD. This suggests the potential involvement of SL26A3-mediated anion transport in IBD pathogenesis. Patients with CLD-associated IBD may require surgery for treatment failure or colon cancer.


Assuntos
Diarreia/congênito , Doenças Inflamatórias Intestinais/epidemiologia , Erros Inatos do Metabolismo/epidemiologia , Adolescente , Adulto , Criança , Antiportadores de Cloreto-Bicarbonato/genética , Estudos de Coortes , Diarreia/epidemiologia , Diarreia/genética , Europa (Continente)/epidemiologia , Feminino , Humanos , Masculino , Erros Inatos do Metabolismo/genética , Mutação , Prevalência , Transportadores de Sulfato/genética , Adulto Jovem
7.
Int J Hyperthermia ; 37(1): 1074-1081, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32954876

RESUMO

BACKGROUND AND AIMS: Surgical resection is currently the cornerstone of liver tumor treatment in children. In adults radiofrequency ablation (RFA) is an established minimally invasive treatment option for small focal liver tumors. Multiprobe stereotactic RFA (SRFA) with intraoperative image fusion to confirm ablation margins allows treatment for large lesions. We describe our experience with SRFA in children with liver masses. METHODS: SRFA was performed in 10 patients with a median age of 14 years (range 0.5-17.0 years) suffering from liver adenoma (n = 3), hepatocellular carcinoma (n = 1), hepatoblastoma (n = 2), myofibroblastic tumor (n = 1), hepatic metastases of extrahepatic tumors (n = 2) and infiltrative hepatic cysts associated with alveolar echinococcosis (n = 1). Overall, 15 lesions with a mean lesion size of 2.6 cm (range 0.7-9.5 cm) were treated in 11 sessions. RESULTS: The technical success rate was 100%, as was the survival rate. No transient adverse effects higher than grade II (Clavien and Dindo) were encountered after interventions. The median hospital stay was 5 d (range 2-33 d). In two patients who subsequently underwent transplant hepatectomy complete ablation was histologically confirmed. Follow-up imaging studies (median 55 months, range 18-129 months) revealed no local or distant recurrence of disease in any patient. CONCLUSIONS: SRFA is an effective minimal-invasive treatment option in pediatric patients with liver tumors of different etiologies.


Assuntos
Carcinoma Hepatocelular , Ablação por Cateter , Neoplasias Hepáticas , Ablação por Radiofrequência , Adolescente , Adulto , Carcinoma Hepatocelular/cirurgia , Criança , Pré-Escolar , Humanos , Lactente , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/cirurgia , Recidiva Local de Neoplasia/cirurgia , Estudos Retrospectivos , Resultado do Tratamento
8.
Clin Genet ; 98(3): 282-287, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32557621

RESUMO

Biallelic loss-of-function mutations in the centrosomal pericentrin gene (PCNT) cause microcephalic osteodysplastic primordial dwarfism type II (MOPDII), which is characterized by extreme growth retardation, microcephaly, skeletal dysplasia, and dental anomalies. Life expectancy is reduced due to a high risk of cerebral vascular anomalies. Here, we report two siblings with MOPDII and attenuated growth restriction, and pachygyria. Compound heterozygosity for two novel truncated PCNT variants was identified. Both truncated PCNT proteins were expressed in patient's fibroblasts, with a reduced total protein amount compared to control. Patient's fibroblasts showed impaired cell cycle progression. As a novel finding, 20% of patient's fibroblasts were shown to express PCNT comparable to control. This was associated with normal mitotic morphology and normal co-localization of mutated PCNT with centrosome-associated proteins γ-tubulin and centrin 3, suggesting some residual function of truncated PCNT proteins. These data expand the clinical and molecular spectrum of MOPDII and indicate that residual PCNT function might be associated with attenuated growth restriction in MOPDII.


Assuntos
Antígenos/genética , Nanismo/genética , Retardo do Crescimento Fetal/genética , Predisposição Genética para Doença , Lisencefalia/genética , Microcefalia/genética , Osteocondrodisplasias/genética , Adolescente , Adulto , Alelos , Centrossomo/metabolismo , Criança , Pré-Escolar , Nanismo/patologia , Feminino , Retardo do Crescimento Fetal/patologia , Fibroblastos/metabolismo , Humanos , Lisencefalia/patologia , Mutação com Perda de Função/genética , Masculino , Microcefalia/patologia , Osteocondrodisplasias/patologia , Irmãos , Tubulina (Proteína)/genética , Adulto Jovem
9.
Hum Genet ; 139(10): 1247-1259, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32306098

RESUMO

Congenital diarrheal disorders (CDD) comprise > 50 monogenic entities featuring chronic diarrhea of early-onset, including defects in nutrient and electrolyte absorption, enterocyte polarization, enteroendocrine cell differentiation, and epithelial integrity. Diarrhea is also a predominant symptom in many immunodeficiencies, congenital disorders of glycosylation, and in some defects of the vesicular sorting and transporting machinery. We set out to identify the etiology of an intractable diarrhea in 2 consanguineous families by whole-exome sequencing, and identified two novel AP1S1 mutations, c.269T>C (p.Leu90Pro) and c.346G>A (p.Glu116Lys). AP1S1 encodes the small subunit of the adaptor protein 1 complex (AP-1), which plays roles in clathrin coat-assembly and trafficking between trans-Golgi network, endosomes and the plasma membrane. An AP1S1 knock-out (KO) of a CaCo2 intestinal cell line was generated to characterize intestinal AP1S1 deficiency as well as identified mutations by stable expression in KO background. Morphology and prototype transporter protein distribution were comparable between parental and KO cells. We observed altered localization of tight-junction proteins ZO-1 and claudin 3, decreased transepithelial electrical resistance and an increased dextran permeability of the CaCo2-AP1S1-KO monolayer. In addition, lumen formation in 3D cultures of these cells was abnormal. Re-expression of wild-type AP1S1 in CaCo2-AP1S1-KO cells reverted these abnormalities, while expression of AP1S1 containing either missense mutation did not. Our data indicate that loss of AP1S1 function causes an intestinal epithelial barrier defect, and that AP1S1 mutations can cause a non-syndromic form of congenital diarrhea, whereas 2 reported truncating AP1S1 mutations caused MEDNIK syndrome, characterized by mental retardation, enteropathy, deafness, neuropathy, ichthyosis, and keratodermia.


Assuntos
Complexo 1 de Proteínas Adaptadoras/genética , Subunidades sigma do Complexo de Proteínas Adaptadoras/genética , Surdez/genética , Diarreia/genética , Ictiose/genética , Deficiência Intelectual/genética , Ceratodermia Palmar e Plantar/genética , Mutação de Sentido Incorreto , Complexo 1 de Proteínas Adaptadoras/deficiência , Subunidades sigma do Complexo de Proteínas Adaptadoras/deficiência , Sequência de Bases , Células CACO-2 , Claudina-3/genética , Claudina-3/metabolismo , Consanguinidade , Surdez/diagnóstico , Surdez/metabolismo , Surdez/patologia , Diarreia/diagnóstico , Diarreia/metabolismo , Diarreia/patologia , Feminino , Expressão Gênica , Técnicas de Inativação de Genes , Teste de Complementação Genética , Humanos , Ictiose/diagnóstico , Ictiose/metabolismo , Ictiose/patologia , Lactente , Recém-Nascido , Deficiência Intelectual/diagnóstico , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Ceratodermia Palmar e Plantar/diagnóstico , Ceratodermia Palmar e Plantar/metabolismo , Ceratodermia Palmar e Plantar/patologia , Linhagem , Permeabilidade , Sequenciamento do Exoma , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
10.
Hum Mutat ; 41(3): 655-667, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31705726

RESUMO

Congenital disorders of glycosylation (CDGs) comprise a large number of inherited metabolic defects that affect the biosynthesis and attachment of glycans. CDGs manifest as a broad spectrum of disease, most often including neurodevelopmental and skeletal abnormalities and skin laxity. Two patients with biallelic CSGALNACT1 variants and a mild skeletal dysplasia have been described previously. We investigated two unrelated patients presenting with short stature with advanced bone age, facial dysmorphism, and mild language delay, in whom trio-exome sequencing identified novel biallelic CSGALNACT1 variants: compound heterozygosity for c.1294G>T (p.Asp432Tyr) and the deletion of exon 4 that includes the start codon in one patient, and homozygosity for c.791A>G (p.Asn264Ser) in the other patient. CSGALNACT1 encodes CSGalNAcT-1, a key enzyme in the biosynthesis of sulfated glycosaminoglycans chondroitin and dermatan sulfate. Biochemical studies demonstrated significantly reduced CSGalNAcT-1 activity of the novel missense variants, as reported previously for the p.Pro384Arg variant. Altered levels of chondroitin, dermatan, and heparan sulfate moieties were observed in patients' fibroblasts compared to controls. Our data indicate that biallelic loss-of-function mutations in CSGALNACT1 disturb glycosaminoglycan synthesis and cause a mild skeletal dysplasia with advanced bone age, CSGALNACT1-CDG.


Assuntos
Defeitos Congênitos da Glicosilação/diagnóstico , Defeitos Congênitos da Glicosilação/genética , Estudos de Associação Genética , Predisposição Genética para Doença , Anormalidades Musculoesqueléticas/diagnóstico , Anormalidades Musculoesqueléticas/genética , Mutação , N-Acetilgalactosaminiltransferases/genética , Sequência de Aminoácidos , Osso e Ossos/anormalidades , Osso e Ossos/diagnóstico por imagem , Fácies , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Recém-Nascido , Mutação com Perda de Função , Masculino , Mutação de Sentido Incorreto , Linhagem , Fenótipo
11.
J Pediatr Gastroenterol Nutr ; 68(1): e1-e6, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30589726

RESUMO

Mutations in the nuclear gene DGUOK, encoding deoxyguanosine kinase, cause an infantile hepatocerebral type of mitochondrial depletion syndrome (MDS). We report 6 MDS patients harboring bi-allelic DGUOK mutations, of which 3 are novel, including a large intragenic Austrian founder deletion. One patient was diagnosed with hepatocellular carcinoma aged 6 months, supporting a link between mitochondrial DNA depletion and tumorigenesis; liver transplantation proved beneficial with regard to both tumor treatment and psychomotor development.


Assuntos
Doenças Mitocondriais/genética , Áustria , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/cirurgia , DNA Mitocondrial/genética , Feminino , Humanos , Lactente , Recém-Nascido , Fígado/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/cirurgia , Transplante de Fígado , Masculino , Doenças Mitocondriais/patologia , Doenças Mitocondriais/cirurgia , Mutação
12.
Traffic ; 18(7): 453-464, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28407399

RESUMO

Microvillus inclusion disease (MVID) is a congenital enteropathy characterized by accumulation of vesiculo-tubular endomembranes in the subapical cytoplasm of enterocytes, historically termed "secretory granules." However, neither their identity nor pathophysiological significance is well defined. Using immunoelectron microscopy and tomography, we studied biopsies from MVID patients (3× Myosin 5b mutations and 1× Syntaxin3 mutation) and compared them to controls and genome-edited CaCo2 cell models, harboring relevant mutations. Duodenal biopsies from 2 patients with novel Myosin 5b mutations and typical clinical symptoms showed unusual ultrastructural phenotypes: aberrant subapical vesicles and tubules were prominent in the enterocytes, though other histological hallmarks of MVID were almost absent (ectopic intra-/intercellular microvilli, brush border atrophy). We identified these enigmatic vesiculo-tubular organelles as Rab11-Rab8-positive recycling compartments of altered size, shape and location harboring the apical SNARE Syntaxin3, apical transporters sodium-hydrogen exchanger 3 (NHE3) and cystic fibrosis transmembrane conductance regulator. Our data strongly indicate that in MVID disrupted trafficking between cargo vesicles and the apical plasma membrane is the primary cause of a defect of epithelial polarity and subsequent facultative loss of brush border integrity, leading to malabsorption. Furthermore, they support the notion that mislocalization of transporters, such as NHE3 substantially contributes to the reported sodium loss diarrhea.


Assuntos
Enterócitos/metabolismo , Síndromes de Malabsorção/metabolismo , Microvilosidades/patologia , Mucolipidoses/metabolismo , Vesículas Secretórias/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Células CACO-2 , Membrana Celular/metabolismo , Enterócitos/ultraestrutura , Humanos , Síndromes de Malabsorção/genética , Masculino , Microvilosidades/genética , Microvilosidades/metabolismo , Mucolipidoses/genética , Mutação , Miosina Tipo V/genética , Transporte Proteico , Proteínas Qa-SNARE/genética , Vesículas Secretórias/ultraestrutura
13.
Hum Mutat ; 38(4): 365-372, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28181337

RESUMO

We identified two unrelated consanguineous families with three children affected by the rare association of congenital nephrotic syndrome (CNS) diagnosed in the first days of life, of hypogonadism, and of prenatally detected adrenal calcifications, associated with congenital adrenal insufficiency in one case. Using exome sequencing and targeted Sanger sequencing, two homozygous truncating mutations, c.1513C>T (p.Arg505*) and c.934delC (p.Leu312Phefs*30), were identified in SGPL1-encoding sphingosine-1-phosphate (S1P) lyase 1. SGPL1 catalyzes the irreversible degradation of endogenous and dietary S1P, the final step of sphingolipid catabolism, and of other phosphorylated long-chain bases. S1P is an intracellular and extracellular signaling molecule involved in angiogenesis, vascular maturation, and immunity. The levels of SGPL1 substrates, S1P, and sphingosine were markedly increased in the patients' blood and fibroblasts, as determined by liquid chromatography-tandem mass spectrometry. Vascular alterations were present in a patient's renal biopsy, in line with changes seen in Sgpl1 knockout mice that are compatible with a developmental defect in vascular maturation. In conclusion, loss of SGPL1 function is associated with CNS, adrenal calcifications, and hypogonadism.


Assuntos
Doenças das Glândulas Suprarrenais/genética , Aldeído Liases/genética , Calcinose/genética , Mutação , Síndrome Nefrótica/genética , Doenças das Glândulas Suprarrenais/congênito , Doenças das Glândulas Suprarrenais/enzimologia , Adulto , Aldeído Liases/deficiência , Animais , Sequência de Bases , Calcinose/enzimologia , Consanguinidade , Feminino , Humanos , Lactente , Lisofosfolipídeos/sangue , Lisofosfolipídeos/metabolismo , Masculino , Camundongos Knockout , Síndrome Nefrótica/congênito , Síndrome Nefrótica/enzimologia , Linhagem , Análise de Sequência de DNA/métodos , Esfingosina/análogos & derivados , Esfingosina/sangue , Esfingosina/metabolismo
14.
Hepatology ; 66(1): 286-288, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28073151

RESUMO

A 52-year old patient presented with lymphedema, protein loosing enteropathy, and sclerosing cholangitis and was diagnosed with lymphedema cholestasis syndrome (LCS). Cholangioscopy revealed dilated lymphatic vessels obstructing the bile duct and compound heterozygosity for collagen and calcium-binding epidermal growth factor domain-containing protein 1 (CCBE1) mutations was identified defining a novel type of LCS. (Hepatology 2017;66:286-288).


Assuntos
Proteínas de Ligação ao Cálcio/genética , Colangite Esclerosante/genética , Colestase/diagnóstico por imagem , Predisposição Genética para Doença , Linfedema/diagnóstico por imagem , Proteínas Supressoras de Tumor/genética , Biópsia por Agulha , Colangiografia/métodos , Colangite Esclerosante/diagnóstico por imagem , Colangite Esclerosante/patologia , Colestase/terapia , Humanos , Imuno-Histoquímica , Linfedema/terapia , Imageamento por Ressonância Magnética/métodos , Pessoa de Meia-Idade , Mutação , Doenças Raras , Recidiva , Índice de Gravidade de Doença
15.
Hum Mutat ; 38(1): 34-38, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27599773

RESUMO

Mutations in genes encoding enzymes responsible for the biosynthesis and structural diversity of glycosaminoglycans (GAGs) cause a variety of disorders affecting bone and connective tissues, including Desbuquois dysplasia (DD). In an infant with prenatal-onset disproportionate short stature, joint laxity, and radiographic findings typical for DD compound-heterozygosity for a large intragenic deletion, and a p.Pro384Arg missense mutation in CSGALNACT1 was found. CSGALNACT1 encodes chondroitin sulfate N-acetylgalactosaminyltransferase-1 (CSGalNAcT-1, ChGn-1), which initiates chondroitin sulfate (CS) chain biosynthesis on the so-called GAG-protein linker region tetrasaccharide. Biochemical studies revealed a reduced GalNAc-transferase activity of the Arg-384 mutant protein, whereas no differences in proteoglycan synthesis in fibroblasts and the GAG content in the urine were found between patient and controls. This is the first description of bi-allelic loss-of-function mutations in CSGALNACT1 that produce a skeletal dysplasia reminiscent of the skeletal dysplasia of Csgalnact1-/- mice, and adds to the genetic heterogeneity of DD.


Assuntos
Instabilidade Articular/diagnóstico , Instabilidade Articular/genética , Anormalidades Musculoesqueléticas/diagnóstico , Anormalidades Musculoesqueléticas/genética , N-Acetilgalactosaminiltransferases/deficiência , Pré-Escolar , Análise Mutacional de DNA , Ativação Enzimática , Éxons , Feminino , Expressão Gênica , Heterozigoto , Humanos , Lactente , Mutação , N-Acetilgalactosaminiltransferases/genética , N-Acetilgalactosaminiltransferases/metabolismo , Fenótipo , Radiografia , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Índice de Gravidade de Doença
16.
J Invest Dermatol ; 137(4): 845-854, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28017832

RESUMO

In this paper, we report three patients with severe palmoplantar keratoderma associated with ichthyosis and sensorineural deafness. Biallelic mutations were found in VPS33B, encoding VPS33B, a Sec1/Munc18 family protein that interacts with Rab11a and Rab25 proteins and is involved in trafficking of the collagen-modifying enzyme LH3. Two patients were homozygous for the missense variant p.Gly131Glu, whereas one patient was compound heterozygous for p.Gly131Glu and the splice site mutation c.240-1G>C, previously reported in patients with arthrogryposis renal dysfunction and cholestasis syndrome. We demonstrated the pathogenicity of variant p.Gly131Glu by assessing the interactions of the mutant VPS33B construct and its ability to traffic LH3. Compared with wild-type VPS33B, the p.Gly131Glu mutant VPS33B had reduced coimmunoprecipitation and colocalization with Rab11a and Rab25 and did not rescue LH3 trafficking. Confirming the cell-based experiments, we found deficient LH3-specific collagen lysine modifications in patients' urine and skin fibroblasts. Additionally, the epidermal ultrastructure of the p.Gly131Glu patients mirrored defects in tamoxifen-inducible VPS33B-deficient Vps33bfl/fl-ERT2 mice. Both patients and murine models revealed an impaired epidermal structure, ascribed to aberrant secretion of lamellar bodies, which are essential for epidermal barrier formation. Our results demonstrate that p.Gly131Glu mutant VPS33B causes an autosomal recessive keratoderma-ichthyosis-deafness syndrome.


Assuntos
Perda Auditiva Neurossensorial/genética , Ictiose Lamelar/genética , Ceratodermia Palmar e Plantar/genética , Mutação , Proteínas de Transporte Vesicular/genética , Adolescente , Adulto , Animais , Colágeno/metabolismo , Perda Auditiva Neurossensorial/diagnóstico , Humanos , Ictiose Lamelar/diagnóstico , Ceratodermia Palmar e Plantar/diagnóstico , Masculino , Camundongos , Prognóstico , Doenças Raras , Estudos de Amostragem , Síndrome , Proteínas rab de Ligação ao GTP/genética
17.
Nat Commun ; 7: 11920, 2016 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-27380894

RESUMO

Osteogenesis imperfecta (OI) is a collagen-related bone dysplasia. We identified an X-linked recessive form of OI caused by defects in MBTPS2, which encodes site-2 metalloprotease (S2P). MBTPS2 missense mutations in two independent kindreds with moderate/severe OI cause substitutions at highly conserved S2P residues. Mutant S2P has normal stability, but impaired functioning in regulated intramembrane proteolysis (RIP) of OASIS, ATF6 and SREBP transcription factors, consistent with decreased proband secretion of type I collagen. Further, hydroxylation of the collagen lysine residue (K87) critical for crosslinking is reduced in proband bone tissue, consistent with decreased lysyl hydroxylase 1 in proband osteoblasts. Reduced collagen crosslinks presumptively undermine bone strength. Also, proband osteoblasts have broadly defective differentiation. These mutations provide evidence that RIP plays a fundamental role in normal bone development.


Assuntos
Membrana Celular/patologia , Colágeno Tipo I/genética , Metaloendopeptidases/genética , Mutação de Sentido Incorreto , Osteoblastos/metabolismo , Osteogênese Imperfeita/genética , Fator 6 Ativador da Transcrição/genética , Fator 6 Ativador da Transcrição/metabolismo , Adulto , Idoso , Diferenciação Celular , Membrana Celular/metabolismo , Colágeno Tipo I/deficiência , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Regulação da Expressão Gênica , Genes Recessivos , Humanos , Hidroxilação , Masculino , Metaloendopeptidases/metabolismo , Pessoa de Meia-Idade , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Osteoblastos/patologia , Osteogênese Imperfeita/metabolismo , Osteogênese Imperfeita/patologia , Linhagem , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/genética , Pró-Colágeno-Lisina 2-Oxoglutarato 5-Dioxigenase/metabolismo , Proteólise , Índice de Gravidade de Doença , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo
18.
J Pediatr Gastroenterol Nutr ; 63(2): 170-6, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-26835907

RESUMO

Congenital diarrheal disorders (CDDs) represent a group of challenging clinical conditions for pediatricians because of the severity of the presentation and the broad range of possible differential diagnoses. CDDs arise from alterations in the transport of nutrients and electrolytes across the intestinal mucosa, from enterocyte and enteroendocrine cell differentiation and/or polarization defects, and from the modulation of the intestinal immune response. Advances were made recently in deciphering the etiology and pathophysiology of one of these disorders, congenital sodium diarrhea (CSD). CSD refers to an intractable diarrhea of intrauterine onset with high fecal sodium loss. CSD is clinically and genetically heterogeneous. A syndromic form of CSD features choanal and intestinal atresias as well as recurrent corneal erosions. Small bowel histology frequently detects an epithelial "tufting" dysplasia. It is autosomal recessively inherited, and caused by SPINT2 mutations. The nonsyndromic form of CSD can be caused by dominant activating mutations in GUCY2C, encoding intestinal receptor guanylate cyclase C (GC-C), and by autosomal recessive SLC9A3 loss-of-function mutations. SLC9A3 encodes Na/H antiporter 3, the major intestinal brush border Na/H exchanger, and a downstream target of GC-C. A number of patients with GUCY2C and SLC9A3 mutations developed inflammatory bowel disease. Both the number of recognized CDD forms as well as the number of underlying disease genes are gradually increasing. Knowledge of these CDD genes enables noninvasive, next-generation gene panel-based testing to facilitate an early diagnosis in CDD. Primary Na/H antiporter 3 and GC-C malfunction is implicated as a predisposition for inflammatory bowel disease in subset of patients.


Assuntos
Anormalidades Múltiplas/diagnóstico , Diarreia/congênito , Erros Inatos do Metabolismo/diagnóstico , Anormalidades Múltiplas/etiologia , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/fisiopatologia , Diagnóstico Diferencial , Diarreia/diagnóstico , Diarreia/etiologia , Diarreia/genética , Diarreia/fisiopatologia , Marcadores Genéticos , Predisposição Genética para Doença , Humanos , Doenças Inflamatórias Intestinais/etiologia , Erros Inatos do Metabolismo/etiologia , Erros Inatos do Metabolismo/genética , Erros Inatos do Metabolismo/fisiopatologia , Mutação , Síndrome
19.
J Cell Biol ; 211(3): 587-604, 2015 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-26553929

RESUMO

Mutations in the motor protein Myosin Vb (Myo5B) or the soluble NSF attachment protein receptor Syntaxin 3 (Stx3) disturb epithelial polarity and cause microvillus inclusion disease (MVID), a lethal hereditary enteropathy affecting neonates. To understand the molecular mechanism of Myo5B and Stx3 interplay, we used genome editing to introduce a defined Myo5B patient mutation in a human epithelial cell line. Our results demonstrate a selective role of Myo5B and Stx3 for apical cargo exocytosis in polarized epithelial cells. Apical exocytosis of NHE3, CFTR (cystic fibrosis transmembrane conductance regulator), and GLUT5 required an interaction cascade of Rab11, Myo5B, Slp4a, Munc18-2, and Vamp7 with Stx3, which cooperate in the final steps of this selective apical traffic pathway. The brush border enzymes DPPIV and sucrase-isomaltase still correctly localize at the apical plasma membrane independent of this pathway. Hence, our work demonstrates how Myo5B, Stx3, Slp4a, Vamp7, Munc18-2, and Rab8/11 cooperate during selective apical cargo trafficking and exocytosis in epithelial cells and thereby provides further insight into MVID pathophysiology.


Assuntos
Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Exocitose/fisiologia , Cadeias Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Proteínas Qa-SNARE/metabolismo , Proteínas R-SNARE/metabolismo , Proteínas de Transporte Vesicular/metabolismo , Células CACO-2 , Linhagem Celular , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Polaridade Celular/fisiologia , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Transportador de Glucose Tipo 5/metabolismo , Células HEK293 , Humanos , Síndromes de Malabsorção/metabolismo , Microvilosidades/metabolismo , Microvilosidades/patologia , Mucolipidoses/metabolismo , Proteínas Munc18/metabolismo , Mutação/fisiologia , Transdução de Sinais/fisiologia , Trocador 3 de Sódio-Hidrogênio , Trocadores de Sódio-Hidrogênio/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo
20.
Proc Natl Acad Sci U S A ; 112(40): 12408-13, 2015 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-26392529

RESUMO

Microvillus inclusion disease (MVID) is a rare intestinal enteropathy with an onset within a few days to months after birth, resulting in persistent watery diarrhea. Mutations in the myosin Vb gene (MYO5B) have been identified in the majority of MVID patients. However, the exact pathophysiology of MVID still remains unclear. To address the specific role of MYO5B in the intestine, we generated an intestine-specific conditional Myo5b-deficient (Myo5bfl/fl;Vil-CreERT2) mouse model. We analyzed intestinal tissues and cultured organoids of Myo5bfl/fl;Vil-CreERT2 mice by electron microscopy, immunofluorescence, and immunohistochemistry. Our data showed that Myo5bfl/fl;Vil-CreERT2 mice developed severe diarrhea within 4 d after tamoxifen induction. Periodic Acid Schiff and alkaline phosphatase staining revealed subapical accumulation of intracellular vesicles in villus enterocytes. Analysis by electron microscopy confirmed an almost complete absence of apical microvilli, the appearance of microvillus inclusions, and enlarged intercellular spaces in induced Myo5bfl/fl;Vil-CreERT2 intestines. In addition, we determined that MYO5B is involved not only in apical but also basolateral trafficking of proteins. The analysis of the intestine during the early onset of the disease revealed that subapical accumulation of secretory granules precedes occurrence of microvillus inclusions, indicating involvement of MYO5B in early differentiation of epithelial cells. By comparing our data with a novel MVID patient, we conclude that our mouse model completely recapitulates the intestinal phenotype of human MVID. This includes severe diarrhea, loss of microvilli, occurrence of microvillus inclusions, and subapical secretory granules. Thus, loss of MYO5B disturbs both apical and basolateral trafficking of proteins and causes MVID in mice.


Assuntos
Síndromes de Malabsorção/metabolismo , Microvilosidades/patologia , Mucolipidoses/metabolismo , Miosina Tipo V/metabolismo , Animais , Modelos Animais de Doenças , Enterócitos/metabolismo , Enterócitos/patologia , Enterócitos/ultraestrutura , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Células Epiteliais/ultraestrutura , Feminino , Humanos , Imuno-Histoquímica , Mucosa Intestinal/metabolismo , Intestinos/patologia , Intestinos/ultraestrutura , Síndromes de Malabsorção/induzido quimicamente , Masculino , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Knockout , Camundongos Transgênicos , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Microvilosidades/metabolismo , Microvilosidades/ultraestrutura , Mucolipidoses/induzido quimicamente , Miosina Tipo V/genética , Técnicas de Cultura de Órgãos , Transporte Proteico/genética , Transporte Proteico/fisiologia , Tamoxifeno
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA