Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Nature ; 606(7913): 382-388, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35614220

RESUMO

Mitochondria are epicentres of eukaryotic metabolism and bioenergetics. Pioneering efforts in recent decades have established the core protein componentry of these organelles1 and have linked their dysfunction to more than 150 distinct disorders2,3. Still, hundreds of mitochondrial proteins lack clear functions4, and the underlying genetic basis for approximately 40% of mitochondrial disorders remains unresolved5. Here, to establish a more complete functional compendium of human mitochondrial proteins, we profiled more than 200 CRISPR-mediated HAP1 cell knockout lines using mass spectrometry-based multiomics analyses. This effort generated approximately 8.3 million distinct biomolecule measurements, providing a deep survey of the cellular responses to mitochondrial perturbations and laying a foundation for mechanistic investigations into protein function. Guided by these data, we discovered that PIGY upstream open reading frame (PYURF) is an S-adenosylmethionine-dependent methyltransferase chaperone that supports both complex I assembly and coenzyme Q biosynthesis and is disrupted in a previously unresolved multisystemic mitochondrial disorder. We further linked the putative zinc transporter SLC30A9 to mitochondrial ribosomes and OxPhos integrity and established RAB5IF as the second gene harbouring pathogenic variants that cause cerebrofaciothoracic dysplasia. Our data, which can be explored through the interactive online MITOMICS.app resource, suggest biological roles for many other orphan mitochondrial proteins that still lack robust functional characterization and define a rich cell signature of mitochondrial dysfunction that can support the genetic diagnosis of mitochondrial diseases.


Assuntos
Mitocôndrias , Proteínas Mitocondriais , Proteínas de Transporte de Cátions , Proteínas de Ciclo Celular , Metabolismo Energético , Humanos , Espectrometria de Massas , Mitocôndrias/genética , Mitocôndrias/metabolismo , Doenças Mitocondriais/genética , Doenças Mitocondriais/metabolismo , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Fatores de Transcrição , Proteínas rab5 de Ligação ao GTP
2.
Blood ; 139(21): 3111-3126, 2022 05 26.
Artigo em Inglês | MEDLINE | ID: mdl-35213692

RESUMO

The congenital bone marrow failure syndrome Diamond-Blackfan anemia (DBA) is typically associated with variants in ribosomal protein (RP) genes impairing erythroid cell development. Here we report multiple individuals with biallelic HEATR3 variants exhibiting bone marrow failure, short stature, facial and acromelic dysmorphic features, and intellectual disability. These variants destabilize a protein whose yeast homolog is known to synchronize the nuclear import of RPs uL5 (RPL11) and uL18 (RPL5), which are both critical for producing ribosomal subunits and for stabilizing the p53 tumor suppressor when ribosome biogenesis is compromised. Expression of HEATR3 variants or repression of HEATR3 expression in primary cells, cell lines of various origins, and yeast models impairs growth, differentiation, pre-ribosomal RNA processing, and ribosomal subunit formation reminiscent of DBA models of large subunit RP gene variants. Consistent with a role of HEATR3 in RP import, HEATR3-depleted cells or patient-derived fibroblasts display reduced nuclear accumulation of uL18. Hematopoietic progenitor cells expressing HEATR3 variants or small-hairpin RNAs knocking down HEATR3 synthesis reveal abnormal acceleration of erythrocyte maturation coupled to severe proliferation defects that are independent of p53 activation. Our study uncovers a new pathophysiological mechanism leading to DBA driven by biallelic HEATR3 variants and the destabilization of a nuclear import protein important for ribosome biogenesis.


Assuntos
Anemia de Diamond-Blackfan , Proteínas , Transporte Ativo do Núcleo Celular/genética , Anemia de Diamond-Blackfan/metabolismo , Humanos , Mutação , Proteínas/genética , Proteínas/metabolismo , Proteínas de Ligação a RNA/genética , Proteínas Ribossômicas/genética , Proteínas Ribossômicas/metabolismo , Ribossomos/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
3.
J Stroke Cerebrovasc Dis ; 30(9): 105997, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34303089

RESUMO

OBJECTIVES: To identify the underlying genetic defect for a consanguineous family with an unusually high number of members affected by cerebral small vessel disease. MATERIALS AND METHODS: A total of 6 individuals, of whom 3 are severely affected, from the family were clinically and radiologically evaluated. SNP genotyping was performed in multiple members to demonstrate genome-wide runs-of-homozygosity. Coding variants in the most likely candidate gene, HTRA1 were explored by Sanger sequencing. Published HTRA1-related phenotypes were extensively reviewed to explore the effect of number of affected alleles on phenotypic expression. RESULTS: Genome-wide homozygosity mapping identified a 3.2 Mbp stretch on chromosome 10q26.3 where HTRA1 gene is located. HTRA1 sequencing revealed an evolutionarily conserved novel homozygous c.824C>T (p.Pro275Leu) mutation, affecting the serine protease domain of HtrA1. Early-onset of cognitive and motor deterioration in homozygotes are in consensus with CARASIL. However, there was a clear phenotypic variability between homozygotes which includes alopecia, a suggested hallmark of CARASIL. All heterozygotes, presenting as CADASIL type 2, had spinal disk degeneration and several neuroimaging findings, including leukoencephalopathy and microhemorrhage despite a lack of severe clinical presentation. CONCLUSION: Here, we clearly demonstrate that CARASIL and CADASIL type 2 are two clinical consequences of the same disorder with different severities thorough the evaluation of the largest collection of homozygotes and heterozygotes segregating in a family. Considering the semi-dominant inheritance of HTRA1-related phenotypes, genetic testing and clinical follow-up must be offered for all members of a family with HTRA1 mutations regardless of symptoms.


Assuntos
Alopecia/genética , CADASIL/genética , Infarto Cerebral/genética , Serina Peptidase 1 de Requerimento de Alta Temperatura A/genética , Leucoencefalopatias/genética , Mutação , Doenças da Coluna Vertebral/genética , Adulto , Idade de Início , Alopecia/diagnóstico , Alopecia/fisiopatologia , CADASIL/diagnóstico , CADASIL/fisiopatologia , Infarto Cerebral/diagnóstico , Infarto Cerebral/fisiopatologia , Consanguinidade , Análise Mutacional de DNA , Feminino , Predisposição Genética para Doença , Hereditariedade , Heterozigoto , Homozigoto , Humanos , Leucoencefalopatias/diagnóstico , Leucoencefalopatias/fisiopatologia , Masculino , Pessoa de Meia-Idade , Linhagem , Fenótipo , Índice de Gravidade de Doença , Doenças da Coluna Vertebral/diagnóstico , Doenças da Coluna Vertebral/fisiopatologia
4.
J Clin Invest ; 127(11): 4090-4103, 2017 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-28972538

RESUMO

Shwachman-Diamond syndrome (SDS) (OMIM #260400) is a rare inherited bone marrow failure syndrome (IBMFS) that is primarily characterized by neutropenia and exocrine pancreatic insufficiency. Seventy-five to ninety percent of patients have compound heterozygous loss-of-function mutations in the Shwachman-Bodian-Diamond syndrome (sbds) gene. Using trio whole-exome sequencing (WES) in an sbds-negative SDS family and candidate gene sequencing in additional SBDS-negative SDS cases or molecularly undiagnosed IBMFS cases, we identified 3 independent patients, each of whom carried a de novo missense variant in srp54 (encoding signal recognition particle 54 kDa). These 3 patients shared congenital neutropenia linked with various other SDS phenotypes. 3D protein modeling revealed that the 3 variants affect highly conserved amino acids within the GTPase domain of the protein that are critical for GTP and receptor binding. Indeed, we observed that the GTPase activity of the mutated proteins was impaired. The level of SRP54 mRNA in the bone marrow was 3.6-fold lower in patients with SRP54-mutations than in healthy controls. Profound reductions in neutrophil counts and chemotaxis as well as a diminished exocrine pancreas size in a SRP54-knockdown zebrafish model faithfully recapitulated the human phenotype. In conclusion, autosomal dominant mutations in SRP54, a key member of the cotranslation protein-targeting pathway, lead to syndromic neutropenia with a Shwachman-Diamond-like phenotype.


Assuntos
Doenças da Medula Óssea/genética , Insuficiência Pancreática Exócrina/genética , Lipomatose/genética , Neutropenia/congênito , Partícula de Reconhecimento de Sinal/genética , Animais , Criança , Síndrome Congênita de Insuficiência da Medula Óssea , Análise Mutacional de DNA , Feminino , Estudos de Associação Genética , Humanos , Lactente , Masculino , Modelos Moleculares , Neutropenia/genética , Pâncreas Exócrino/metabolismo , Fenótipo , Domínios Proteicos , Síndrome de Shwachman-Diamond , Partícula de Reconhecimento de Sinal/química , Peixe-Zebra
5.
Eur J Hum Genet ; 25(10): 1118-1125, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28832566

RESUMO

Microcephalic primordial dwarfism (MPD) is a group of autosomal recessive inherited single-gene disorders with intrauterine and postnatal global growth failure. Seckel syndrome is the most common form of the MPD. Ten genes are known with Seckel syndrome. Using genome-wide SNP genotyping and homozygosity mapping we mapped a Seckel syndrome gene to chromosomal region 4q28.1-q28.3 in a Turkish family. Direct sequencing of PLK4 (polo-like kinase 4) revealed a homozygous splicing acceptor site transition (c.31-3 A>G) that results in a premature translation termination (p.[=,Asp11Profs*14]) causing deletion of all known functional domains of the protein. PLK4 is a master regulator of centriole biogenesis and its deficiency has recently been associated with Seckel syndrome. However, the role of PLK4 in genomic stability and the DNA damage response is unclear. Evaluation of the PLK4-Seckel fibroblasts obtained from patient revealed the expected impaired centriole biogenesis, disrupted mitotic morphology, G2/M delay, and extended cell doubling time. Analysis of the PLK4-Seckel cells indicated that PLK4 is also essential for genomic stability and DNA damage response. These findings provide mechanistic insight into the pathogenesis of the severe growth failure associated with PLK4-deficiency.


Assuntos
Centrossomo/metabolismo , Dano ao DNA , Nanismo/genética , Microcefalia/genética , Mutação , Proteínas Serina-Treonina Quinases/genética , Adulto , Células Cultivadas , Criança , Pré-Escolar , Cromossomos Humanos Par 4/genética , Nanismo/patologia , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Instabilidade Genômica , Humanos , Lactente , Masculino , Microcefalia/patologia , Mitose , Linhagem , Splicing de RNA/genética
6.
J Allergy Clin Immunol ; 137(3): 879-88.e2, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26476480

RESUMO

BACKGROUND: Coronin-1A (CORO1A) is a regulator of actin dynamics important for T-cell homeostasis. CORO1A deficiency causes T(-)B(+) natural killer-positive severe combined immunodeficiency or T-cell lymphopenia with severe viral infections. However, because all known human mutations in CORO1A abrogate protein expression, the role of the protein's functional domains in host immunity is unknown. OBJECTIVE: We sought to identify the cause of the primary immunodeficiency in 2 young adult siblings with a history of disseminated varicella, cutaneous warts, and CD4(+) T-cell lymphopenia. METHODS: We performed immunologic, genetic, and biochemical studies in the patients, family members, and healthy control subjects. RESULTS: Both patients had CD4(+) T-cell lymphopenia and decreased lymphocyte proliferation to mitogens. IgG, IgM, IgA, and specific antibody responses were normal. Whole-genome sequencing identified a homozygous frameshift mutation in CORO1A disrupting the last 2 C-terminal domains by replacing 61 amino acids with a novel 91-amino-acid sequence. The CORO1A(S401fs) mutant was expressed in the patients' lymphocytes at a level comparable with that of wild-type CORO1A in normal lymphocytes but did not oligomerize and had impaired cytoskeletal association. CORO1A(S401fs) was associated with increased filamentous actin accumulation in T cells, severely defective thymic output, and impaired T-cell survival but normal calcium flux and cytotoxicity, demonstrating the importance of CORO1A oligomerization and subcellular localization in T-cell homeostasis. CONCLUSIONS: We describe a truncating mutation in CORO1A that permits protein expression and survival into young adulthood. Our studies demonstrate the importance of intact CORO1A C-terminal domains in thymic egress and T-cell survival, as well as in defense against viral pathogens.


Assuntos
Citoesqueleto/metabolismo , Homozigoto , Proteínas dos Microfilamentos/genética , Mutação , Multimerização Proteica , Viroses/etiologia , Viroses/metabolismo , Actinas/química , Actinas/metabolismo , Adolescente , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Degranulação Celular/genética , Degranulação Celular/imunologia , Sobrevivência Celular/genética , Análise Mutacional de DNA , Feminino , Mutação da Fase de Leitura , Humanos , Imunoglobulinas/sangue , Imunoglobulinas/imunologia , Contagem de Linfócitos , Linfopenia , Masculino , Camundongos , Proteínas dos Microfilamentos/química , Proteínas dos Microfilamentos/metabolismo , Linhagem , Fenótipo , Multimerização Proteica/genética , Transporte Proteico , Irmãos , Transdução de Sinais , Dermatopatias/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Viroses/diagnóstico , Verrugas/patologia
7.
Hum Mol Genet ; 24(8): 2267-73, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25561690

RESUMO

Colobomatous macrophthalmia with microcornea syndrome (MACOM, Online Mendelian Inheritance in Man (OMIM) 602499) is an autosomal dominantly inherited malformation of the eye, which is characterized by microcornea with increased axial length, coloboma of the iris and of the optic disc, and severe myopia. We performed whole-exome sequencing (WES) in two affected individuals from the 2p23-p16-linked MACOM family, which includes 13 affected individuals in 3 generations. As no shared novel variation was found on the linked haplotype, we performed copy number variation (CNV) analysis by comparing the coverage of all exons in the WES data sets of the 2 patients with the coverage of 26 control exomes. We identified a heterozygous deletion predicted to span 22 kb including exons 14-17 of CRIM1 (cysteine-rich transmembrane bone morphogenetic protein (BMP) regulator 1). Quantitative PCR (qPCR) analysis confirmed the deletion, which was present in 11 affected individuals. Split-read analysis of WES data followed by breakpoint PCR and Sanger sequencing determined both breakpoints flanked by a 4-bp microhomology (CTTG). In the mouse, Crim1 is a growth-factor-binding protein with pleiotropic roles in the development of multiple organs, including the eye. To investigate the role of Crim1 during eye development in mice, we crossed a Crim1(flox) mouse line with the Ap2α-cre mouse line, which expresses Cre in the head surface ectoderm. Strikingly, we observed alterations of eye development in homozygous mice leading to severe anatomical and morphological changes overlapping with the anomalies observed in MACOM patients. Taken together, these findings identify CRIM1 as the causative gene for MACOM syndrome and emphasize the importance of CRIM1 in eye development.


Assuntos
Receptores de Proteínas Morfogenéticas Ósseas/metabolismo , Doenças da Córnea/genética , Anormalidades do Olho/genética , Olho/crescimento & desenvolvimento , Haploinsuficiência , Proteínas de Membrana/metabolismo , Adulto , Animais , Sequência de Bases , Receptores de Proteínas Morfogenéticas Ósseas/genética , Doenças da Córnea/metabolismo , Doenças da Córnea/fisiopatologia , Variações do Número de Cópias de DNA , Éxons , Olho/anatomia & histologia , Olho/metabolismo , Anormalidades do Olho/metabolismo , Anormalidades do Olho/fisiopatologia , Feminino , Homozigoto , Humanos , Masculino , Proteínas de Membrana/genética , Camundongos , Dados de Sequência Molecular , Linhagem , Adulto Jovem
8.
Am J Hum Genet ; 95(6): 649-59, 2014 Dec 04.
Artigo em Inglês | MEDLINE | ID: mdl-25466284

RESUMO

Mesoaxial synostotic syndactyly, Malik-Percin type (MSSD) (syndactyly type IX) is a rare autosomal-recessive nonsyndromic digit anomaly with only two affected families reported so far. We previously showed that the trait is genetically distinct from other syndactyly types, and through autozygosity mapping we had identified a locus on chromosome 17p13.3 for this unique limb malformation. Here, we extend the number of independent pedigrees from various geographic regions segregating MSSD to a total of six. We demonstrate that three neighboring missense mutations affecting the highly conserved DNA-binding region of the basic helix-loop-helix A9 transcription factor (BHLHA9) are associated with this phenotype. Recombinant BHLHA9 generated by transient gene expression is shown to be located in the cytoplasm and the cell nucleus. Transcription factors 3, 4, and 12, members of the E protein (class I) family of helix-loop-helix transcription factors, are highlighted in yeast two-hybrid analysis as potential dimerization partners for BHLHA9. In the presence of BHLHA9, the potential of these three proteins to activate expression of an E-box-regulated target gene is reduced considerably. BHLHA9 harboring one of the three substitutions detected in MSSD-affected individuals eliminates entirely the transcription activation by these class I bHLH proteins. We conclude that by dimerizing with other bHLH protein monomers, BHLHA9 could fine tune the expression of regulatory factors governing determination of central limb mesenchyme cells, a function made impossible by altering critical amino acids in the DNA binding domain. These findings identify BHLHA9 as an essential player in the regulatory network governing limb morphogenesis in humans.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Dedos/anormalidades , Mutação de Sentido Incorreto , Sindactilia/genética , Dedos do Pé/anormalidades , Sequência de Aminoácidos , Sítios de Ligação , Análise Mutacional de DNA , Dimerização , Feminino , Genes Reporter , Genótipo , Haplótipos , Humanos , Itália , Masculino , Pessoa de Meia-Idade , Paquistão , Linhagem , Fenótipo , Ligação Proteica , Estrutura Terciária de Proteína , Alinhamento de Sequência , Turquia , Adulto Jovem
9.
Br J Ophthalmol ; 98(6): 832-40, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24568872

RESUMO

AIM: This study aimed to identify the underlying genetic defect responsible for anophthalmia/microphthalmia. METHODS: In total, two Turkish families with a total of nine affected individuals were included in the study. Affymetrix 250 K single nucleotide polymorphism genotyping and homozygosity mapping were used to identify the localisation of the genetic defect in question. Coding region of the ALDH1A3 gene was screened via direct sequencing. cDNA samples were generated from primary fibroblast cell cultures for expression analysis. Reverse transcriptase PCR (RT-PCR) analysis was performed using direct sequencing of the obtained fragments. RESULTS: The causative genetic defect was mapped to chromosome 15q26.3. A homozygous G>A substitution (c.666G>A) at the last nucleotide of exon 6 in the ALDH1A3 gene was identified in the first family. Further cDNA sequencing of ALDH1A3 showed that the c.666G>A mutation caused skipping of exon 6, which predicted in-frame loss of 43 amino acids (p.Trp180_Glu222del). A novel missense c.1398C>A mutation in exon 12 of ALDH1A3 that causes the substitution of a conserved asparagine by lysine at amino acid position 466 (p.Asn466Lys) was observed in the second family. No extraocular findings-except for nevus flammeus in one affected individual and a variant of Dandy-Walker malformation in another affected individual-were observed. Autistic-like behaviour and mental retardation were observed in three cases. CONCLUSIONS: In conclusion, novel ALDH1A3 mutations identified in the present study confirm the pivotal role of ALDH1A3 in human eye development. Autistic features, previously reported as an associated finding, were considered to be the result of social deprivation and inadequate parenting during early infancy in the presented families.


Assuntos
Aldeído Oxirredutases/genética , Anoftalmia/genética , Microftalmia/genética , Mutação de Sentido Incorreto , Sítios de Splice de RNA , Adolescente , Sequência de Bases , Criança , Cromossomos Humanos Par 15/genética , Análise Mutacional de DNA , Feminino , Genes Recessivos/genética , Genótipo , Humanos , Masculino , Dados de Sequência Molecular , Linhagem , Reação em Cadeia da Polimerase , Polimorfismo de Nucleotídeo Único , Reação em Cadeia da Polimerase Via Transcriptase Reversa
10.
Mol Genet Genomic Med ; 1(4): 223-37, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-24498618

RESUMO

We have characterized a novel autosomal recessive Crouzon-like craniosynostosis syndrome in a 12-affected member family from Antakya, Turkey, the presenting features of which include: multiple suture synostosis, midface hypoplasia, variable degree of exophthalmos, relative prognathism, a beaked nose, and conductive hearing loss. Homozygosity mapping followed by targeted next-generation sequencing identified a c.479+6T>G mutation in the interleukin 11 receptor alpha gene (IL11RA) on chromosome 9p21. This donor splice-site mutation leads to a high percentage of aberrant IL11RA mRNA transcripts in an affected individual and altered mRNA splicing determined by in vitro exon trapping. An extended IL11RA mutation screen was performed in a cohort of 79 patients with an initial clinical diagnosis of Crouzon syndrome, pansynostosis, or unclassified syndromic craniosynostosis. We identified mutations segregating with the disease in five families: a German patient of Turkish origin and a Turkish family with three affected sibs all of whom were homozygous for the previously identified IL11RA c.479+6T>G mutation; a family with pansynostosis with compound heterozygous missense mutations, p.Pro200Thr and p.Arg237Pro; and two further Turkish families with Crouzon-like syndrome carrying the homozygous nonsense mutations p.Tyr232* and p.Arg292*. Using transient coexpression in HEK293T and COS7 cells, we demonstrated dramatically reduced IL11-mediated STAT3 phosphorylation for all mutations. Immunofluorescence analysis of mouse Il11ra demonstrated specific protein expression in cranial mesenchyme which was localized around the coronal suture tips and in the lambdoidal suture. In situ hybridization analysis of adult zebrafish also detected zfil11ra expression in the coronal suture between the overlapping frontal and parietal plates. This study demonstrates that mutations in the IL11RA gene cause an autosomal recessive Crouzon-like craniosynostosis.

11.
Am J Hum Genet ; 90(4): 661-74, 2012 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-22482805

RESUMO

Bone morphogenetic protein 1 (BMP1) is an astacin metalloprotease with important cellular functions and diverse substrates, including extracellular-matrix proteins and antagonists of some TGFß superfamily members. Combining whole-exome sequencing and filtering for homozygous stretches of identified variants, we found a homozygous causative BMP1 mutation, c.34G>C, in a consanguineous family affected by increased bone mineral density and multiple recurrent fractures. The mutation is located within the BMP1 signal peptide and leads to impaired secretion and an alteration in posttranslational modification. We also characterize a zebrafish bone mutant harboring lesions in bmp1a, demonstrating conservation of BMP1 function in osteogenesis across species. Genetic, biochemical, and histological analyses of this mutant and a comparison to a second, similar locus reveal that Bmp1a is critically required for mature-collagen generation, downstream of osteoblast maturation, in bone. We thus define the molecular and cellular bases of BMP1-dependent osteogenesis and show the importance of this protein for bone formation and stability.


Assuntos
Proteína Morfogenética Óssea 1/fisiologia , Osteogênese/genética , Osteogênese/fisiologia , Animais , Sequência de Bases , Conservadores da Densidade Óssea/uso terapêutico , Proteína Morfogenética Óssea 1/genética , Proteína Morfogenética Óssea 1/metabolismo , Osso e Ossos/metabolismo , Diferenciação Celular , Pré-Escolar , Colágeno/biossíntese , Difosfonatos/uso terapêutico , Exoma , Feminino , Fraturas Ósseas/tratamento farmacológico , Fraturas Ósseas/prevenção & controle , Loci Gênicos , Proteínas de Choque Térmico , Humanos , Masculino , Dados de Sequência Molecular , Mutação , Osteoblastos/efeitos dos fármacos , Osteoblastos/fisiologia , Osteogênese/efeitos dos fármacos , Fragmentos de Peptídeos , Processamento de Proteína Pós-Traducional , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
12.
Am J Hum Genet ; 90(1): 76-85, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22197489

RESUMO

The autosomal-recessive form of popliteal pterygium syndrome, also known as Bartsocas-Papas syndrome, is a rare, but frequently lethal disorder characterized by marked popliteal pterygium associated with multiple congenital malformations. Using Affymetrix 250K SNP array genotyping and homozygosity mapping, we mapped this malformation syndrome to chromosomal region 21q22.3. Direct sequencing of RIPK4 (receptor-interacting serine/threonine kinase protein 4) showed a homozygous transversion (c.362T>A) that causes substitution of a conserved isoleucine with asparagine at amino acid position 121 (p.Ile121Asn) in the serine/threonine kinase domain of the protein. Additional pathogenic mutations-a homozygous transition (c.551C>T) that leads to a missense substitution (p.Thr184Ile) at a conserved position and a homozygous one base-pair insertion mutation (c.777_778insA) predicted to lead to a premature stop codon (p.Arg260ThrfsX14) within the kinase domain-were observed in two families. Molecular modeling of the kinase domain showed that both the Ile121 and Thr184 positions are critical for the protein's stability and kinase activity. Luciferase reporter assays also demonstrated that these mutations are critical for the catalytic activity of RIPK4. RIPK4 mediates activation of the nuclear factor-κB (NF-κB) signaling pathway and is required for keratinocyte differentiation and craniofacial and limb development. The phenotype of Ripk4(-/-) mice is consistent with the human phenotype presented herein. Additionally, the spectrum of malformations observed in the presented families is similar, but less severe than the conserved helix-loop-helix ubiquitous kinase (CHUK)-deficient human fetus phenotype; known as Cocoon syndrome; this similarity indicates that RIPK4 and CHUK might function via closely related pathways to promote keratinocyte differentiation and epithelial growth.


Assuntos
Cromossomos Humanos Par 21/genética , Fenda Labial/genética , Fissura Palatina/genética , Mutação , Proteínas Serina-Treonina Quinases/genética , Pterígio/congênito , Adolescente , Animais , Sequência de Bases , Análise Mutacional de DNA , Feminino , Genes Recessivos , Humanos , Lactente , Recém-Nascido , Masculino , Camundongos , Dados de Sequência Molecular , NF-kappa B/metabolismo , Polimorfismo de Nucleotídeo Único , Pterígio/genética , Anormalidades da Pele
13.
Am J Hum Genet ; 89(1): 139-47, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21722858

RESUMO

Idiopathic nephrotic syndrome (INS) is a genetically heterogeneous group of disorders characterized by proteinuria, hypoalbuminemia, and edema. Because it typically results in end-stage kidney disease, the steroid-resistant subtype (SRNS) of INS is especially important when it occurs in children. The present study included 29 affected and 22 normal individuals from 17 SRNS families; genome-wide analysis was performed with Affymetrix 250K SNP arrays followed by homozygosity mapping. A large homozygous stretch on chromosomal region 12p12 was identified in one consanguineous family with two affected siblings. Direct sequencing of protein tyrosine phosphatase receptor type O (PTPRO; also known as glomerular epithelial protein-1 [GLEPP1]) showed homozygous c.2627+1G>T donor splice-site mutation. This mutation causes skipping of the evolutionarily conserved exon 16 (p.Glu854_Trp876del) at the RNA level. Immunohistochemistry with GLEPP1 antibody showed a similar staining pattern in the podocytes of the diseased and control kidney tissues. We used a highly polymorphic intragenic DNA marker-D12S1303-to search for homozygosity in 120 Turkish and 13 non-Turkish individuals in the PodoNet registry. This analysis yielded 17 candidate families, and a distinct homozygous c.2745+1G>A donor splice-site mutation in PTPRO was further identified via DNA sequencing in a second Turkish family. This mutation causes skipping of exon 19, and this introduces a premature stop codon at the very beginning of exon 20 (p.Asn888Lysfs*3) and causes degradation of mRNA via nonsense-mediated decay. Immunohistochemical analysis showed complete absence of immunoreactive PTPRO. Ultrastructural alterations, such as diffuse foot process fusion and extensive microvillus transformation of podocytes, were observed via electron microscopy in both families. The present study introduces mutations in PTPRO as another cause of autosomal-recessive nephrotic syndrome.


Assuntos
Síndrome Nefrótica/congênito , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Adolescente , Idade de Início , Sequência de Aminoácidos , Criança , Pré-Escolar , Cromossomos Humanos Par 12 , Códon sem Sentido/genética , Consanguinidade , Éxons , Feminino , Genes Recessivos , Estudo de Associação Genômica Ampla/métodos , Homozigoto , Humanos , Masculino , Dados de Sequência Molecular , Síndrome Nefrótica/genética , Linhagem , Polimorfismo de Nucleotídeo Único , Sítios de Splice de RNA , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo
14.
Am J Hum Genet ; 87(6): 834-41, 2010 Dec 10.
Artigo em Inglês | MEDLINE | ID: mdl-21109228

RESUMO

Limb-girdle muscular dystrophy (LGMD) is a genetically heterogeneous group of inherited muscular disorders manifesting symmetric, proximal, and slowly progressive muscle weakness. Using Affymetrix 250K SNP Array genotyping and homozygosity mapping, we mapped an autosomal-recessive LGMD phenotype to the telomeric portion of chromosome 8q in a consanguineous Turkish family with three affected individuals. DNA sequence analysis of PLEC identified a homozygous c.1_9del mutation containing an initiation codon in exon 1f, which is an isoform-specific sequence of plectin isoform 1f. The same homozygous mutation was also detected in two additional families during the analysis of 72 independent LGMD2-affected families. Moreover, we showed that the expression of PLEC was reduced in the patient's muscle and that there was almost no expression for plectin 1f mRNA as a result of the mutation. In addition to dystrophic changes in muscle, ultrastructural alterations, such as membrane duplications, an enlarged space between the membrane and sarcomere, and misalignment of Z-disks, were observed by transmission electron microscopy. Unlike the control skeletal muscle, no sarcolemmal staining of plectin was detected in the patient's muscle. We conclude that as a result of plectin 1f deficiency, the linkage between the sarcolemma and sarcomere is broken, which could affect the structural organization of the myofiber. Our data show that one of the isoforms of plectin plays a key role in skeletal muscle function and that disruption of the plectin 1f can cause the LGMD2 phenotype without any dermatologic component as was previously reported with mutations in constant exons of PLEC.


Assuntos
Éxons , Genes Recessivos , Distrofia Muscular do Cíngulo dos Membros/genética , Mutação , Plectina/genética , Isoformas de Proteínas/genética , Consanguinidade , Feminino , Humanos , Masculino , Linhagem
15.
Am J Med Genet A ; 152A(10): 2628-33, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20814945

RESUMO

We report a family with Marie Unna hereditary hypotrichosis (MUHH) from Turkey. MUHH is a distinct form of scalp and body hair loss characterized by the absence or scarcity of scalp hair, eyebrows, and eyelashes at birth. Coarse wiry hair begins to grow during childhood. Around puberty, progressive hair loss occurs in the affected patients. Recently, mutations were identified in U2HR, an inhibitory upstream open reading frame in the 5'-untranslated region of the human hairless gene (HR) as the underlying cause of MUHH. We are presenting hair loss of eyebrows in a Turkish family comprising eight affected and seven unaffected individuals. The pedigree is compatible with autosomal dominant inheritance. Linkage and haplotype analyses confirmed linkage of this family to the MUHH locus at cytoband 8p21. By sequencing U2HR, we identified the mutation c.2T>C (M1T) in all affected family members. We concluded that there may be considerable clinical variations in MUHH, and that eyebrow loss is an important clue for accurate diagnosis.


Assuntos
Doenças do Cabelo/genética , Cabelo/anormalidades , Hipotricose/genética , Adolescente , Diagnóstico Diferencial , Sobrancelhas/anormalidades , Família , Feminino , Marcadores Genéticos , Cabelo/ultraestrutura , Humanos , Hipotricose/diagnóstico , Masculino , Microscopia Eletrônica de Varredura , Linhagem , Fatores de Transcrição/genética , Turquia
16.
J Craniofac Surg ; 21(5): 1460-4, 2010 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-20818260

RESUMO

BACKGROUND: Craniofacial structures have an intimate relationship with the central nervous system in the embryologic development period and the developmental abnormalities of the face and skull that are frequently associated with malformations of the central nervous system. Additional intracranial and extracranial malformations in a patient with craniofacial deformity may negatively affect the outcome of the surgery and the quality of life. PATIENTS AND METHODS: A retrospective analysis of a total of 123 patients with craniofacial anomalies was performed. Physical examination notes, ophthalmologic findings, computed tomography, and magnetic resonance imaging reports were retrospectively analyzed, and intracranial and extracranial malformations and ophthalmologic problems in each group were categorized. RESULTS: Of the patients with nonsyndromic craniosynostosis, 29% had intracranial and extracranial malformations. Of them, 17% had ophthalmologic problems. Of the patients with syndromic craniosynostosis, 34% had intracranial and 31% had extracranial malformations. In the patients with craniofacial cleft, 60% had intracranial and 30% had extracranial malformations. The most common intracranial malformations are hydrocephaly, Chiari type 1 malformation, and corpus callosum disorders. CONCLUSIONS: A multidisciplinary approach is essential in the evaluation and follow-up of individuals with craniofacial abnormalities. Conventional radiography and three-dimensional computed tomography of the bony skeleton and axial scanning of the soft tissues is our first-step routine. Brain magnetic resonance imaging should be performed in patients with multiple-suture synostosis, syndromic synostosis, and craniofacial clefts to rule out central nervous system and soft tissue malformations. During the postoperative first year, conventional x-rays are sufficient to evaluate the craniofacial area. Central nervous system disorders may cause major headaches, muscle weakness, hearing problems, extreme fatigue, poor motor coordination, and cognitive and social disabilities even when their intelligence quotient is normal. Therefore, every effort should be performed to search and treat additional malformations. Prevention of additional morbidities improves surgical and social outcomes.


Assuntos
Anormalidades Múltiplas/epidemiologia , Anormalidades Craniofaciais/patologia , Adolescente , Adulto , Agenesia do Corpo Caloso , Anoftalmia/epidemiologia , Malformação de Arnold-Chiari/epidemiologia , Criança , Pré-Escolar , Fenda Labial/epidemiologia , Fissura Palatina/epidemiologia , Anormalidades Craniofaciais/epidemiologia , Craniossinostoses/epidemiologia , Encefalocele/epidemiologia , Feminino , Humanos , Hidrocefalia/epidemiologia , Lactente , Imageamento por Ressonância Magnética , Masculino , Exame Físico , Estudos Retrospectivos , Tomografia Computadorizada por Raios X
17.
Am J Hum Genet ; 86(5): 789-96, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20451171

RESUMO

We present an autosomal-recessive frontonasal dysplasia (FND) characterized by bilateral extreme microphthalmia, bilateral oblique facial cleft, complete cleft palate, hypertelorism, wide nasal bridge with hypoplasia of the ala nasi, and low-set, posteriorly rotated ears in two distinct families. Using Affymetrix 250K SNP array genotyping and homozygosity mapping, we mapped this clinical entity to chromosome 12q21. In one of the families, three siblings were affected, and CNV analysis of the critical region showed a homozygous 3.7 Mb deletion containing the ALX1 (CART1) gene, which encodes the aristaless-like homeobox 1 transcription factor. In the second family we identified a homozygous donor-splice-site mutation (c.531+1G > A) in the ALX1 gene, providing evidence that complete loss of function of ALX1 protein causes severe disruption of early craniofacial development. Unlike loss of its murine ortholog, loss of human ALX1 does not result in neural-tube defects; however, it does severely affect the orchestrated fusion between frontonasal, nasomedial, nasolateral, and maxillary processes during early-stage embryogenesis. This study further expands the spectrum of the recently recognized autosomal-recessive ALX-related FND phenotype in humans.


Assuntos
Fissura Palatina/genética , Proteínas de Homeodomínio/genética , Microftalmia/genética , Anormalidades Musculoesqueléticas/genética , Mutação , Orelha/anormalidades , Face/anormalidades , Homozigoto , Humanos , Fenótipo , Sítios de Splice de RNA/genética , Deleção de Sequência/genética
18.
Hum Mutat ; 31(1): 81-9, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19847792

RESUMO

Werner mesomelic syndrome (WMS) is an autosomal dominant disorder with unknown molecular etiology characterized by hypo- or aplasia of the tibiae in addition to the preaxial polydactyly (PPD) of the hands and feet and/or five-fingered hand with absence of thumbs. We show that point mutations of a specific nucleotide within the sonic hedgehog (SHH) regulatory region (ZRS) cause WMS. In a previously unpublished WMS family, we identified the causative G>A transition at position 404 of the ZRS, and in six affected family members of a second WMS family we found a 404G>C mutation of the ZRS. The 404G>A ZRS mutation is known as the "Cuban mutation" of PPD type II (PPD2). Interestingly, the index patient of that family had tibial hypoplasia as well. These data provide the first evidence that WMS is caused by a specific ZRS mutation, which leads to strong ectopic SHH expression. In contrast, we show that complete duplications of the ZRS region lead to type Haas polysyndactyly or triphalangeal thumb-polysyndactyly syndrome, but do not affect lower limb development. We suggest the term "ZRS-associated syndromes" and a clinical subclassification for the continuum of limb malformations caused by different molecular alterations of the ZRS.


Assuntos
Elementos Facilitadores Genéticos/genética , Proteínas Hedgehog/genética , Deformidades Congênitas dos Membros/genética , Mutação Puntual , Polidactilia/genética , Sindactilia/genética , Polegar/anormalidades , Adulto , Feminino , Falanges dos Dedos da Mão/anormalidades , Predisposição Genética para Doença , Humanos , Masculino , Síndrome , Tíbia/anormalidades
19.
Am J Med Genet A ; 143A(12): 1308-12, 2007 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-17506091

RESUMO

Colobomatous macrophthalmia with microcornea syndrome (OMIM 602499) is a rare, autosomal dominant malformation characterized by microcornea, uveal coloboma, axial enlargement of the globe, and myopia. Using what is currently the largest described pedigree and candidate localization approach, we first excluded the candidate genes PAX2, PAX3, PAX6, and PAX9. Subsequently, the chromosome 14q24 region containing the CHX10, SIX1, and SIX4 genes were also excluded. Positive LOD scores were obtained with the DNA markers selected from the 2p23-p16 region. A maximum pairwise LOD score of 3.61 (Theta = 0) was noted with the DNA marker D2S1788. Haplotype analysis positioned the locus between DNA markers D2S2263 and D2S1352 within a 22 Mb physical interval. This region contains major candidate genes, such as SIX2, SIX3, and CYP1B1; however, mutation analysis did not identify a causative mutation in these genes. Macrophthalmia, colobomatous, with microcornea (MACOM) is proposed as the gene symbol for this malformation linked to 2p23-p16.


Assuntos
Cromossomos Humanos Par 2/genética , Coloboma/genética , Doenças da Córnea/genética , Coloboma/patologia , Doenças da Córnea/patologia , Genes Dominantes , Haplótipos/genética , Humanos , Escore Lod , Linhagem , Turquia
20.
Am J Med Genet A ; 134(4): 404-8, 2005 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-15779011

RESUMO

Previously we have described a novel and distinct form of non-syndromic osseous syndactyly segregating in an autosomal recessive pattern in a consanguineous Pakistani family. The limb findings include mesoaxial reduction of the fingers, synostoses of the third and fourth metacarpals with associated single phalanges, fifth finger clinodactyly, and preaxial webbing of toes. We identified another published report of this phenotype in a large, inbred Turkish family. In the present study we mapped the phenotype in the Pakistani and Turkish families to chromosome 17p13.3 (multipoint LOD score 5.1). The identification of a single locus for this complex limb malformation in two families with distinct ethnic backgrounds supports the hypothesis that this is a distinct form of syndactyly. Since this form of syndactyly is phenotypically distinct from the previously described eight types, we propose to name this phenotype mesoaxial synostotic syndactyly with phalangeal reduction (MSSD, type IX syndactyly, Malik-Percin type).


Assuntos
Anormalidades Múltiplas/genética , Cromossomos Humanos Par 17/genética , Genes Recessivos/genética , Deformidades Congênitas dos Membros/patologia , Sindactilia/patologia , Sinostose/patologia , Anormalidades Múltiplas/patologia , Mapeamento Cromossômico , Saúde da Família , Feminino , Dedos/anormalidades , Ligação Genética , Genoma Humano , Humanos , Escore Lod , Masculino , Repetições de Microssatélites , Paquistão , Linhagem , Turquia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...