Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
1.
Differentiation ; 133: 60-76, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37481904

RESUMO

Mutations in SHH and several other genes encoding components of the Hedgehog signaling pathway have been associated with holoprosencephaly syndromes, with craniofacial anomalies ranging in severity from cyclopia to facial cleft to midfacial and mandibular hypoplasia. Studies in animal models have revealed that SHH signaling plays crucial roles at multiple stages of craniofacial morphogenesis, from cranial neural crest cell survival to growth and patterning of the facial primordia to organogenesis of the palate, mandible, tongue, tooth, and taste bud formation and homeostasis. This article provides a summary of the major findings in studies of the roles of SHH signaling in craniofacial development, with emphasis on recent advances in the understanding of the molecular and cellular mechanisms regulating the SHH signaling pathway activity and those involving SHH signaling in the formation and patterning of craniofacial structures.


Assuntos
Proteínas Hedgehog , Holoprosencefalia , Animais , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Crista Neural/metabolismo , Holoprosencefalia/genética , Holoprosencefalia/metabolismo , Morfogênese/genética , Transdução de Sinais/genética
2.
J Med Genet ; 59(4): 358-365, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-33820834

RESUMO

BACKGROUND: Holoprosencephaly is a spectrum of developmental disorder of the embryonic forebrain in which there is failed or incomplete separation of the prosencephalon into two cerebral hemispheres. To date, dominant mutations in sonic hedgehog (SHH) pathway genes are the predominant Mendelian causes, and have marked interfamilial and intrafamilial phenotypical variabilities. METHODS: We describe two families in which offspring had holoprosencephaly spectrum and homozygous predicted-deleterious variants in phospholipase C eta-1 (PLCH1). Immunocytochemistry was used to examine the expression pattern of PLCH1 in human embryos. We used SHH as a marker of developmental stage and of early embryonic anatomy. RESULTS: In the first family, two siblings had congenital hydrocephalus, significant developmental delay and a monoventricle or fused thalami with a homozygous PLCH1 c.2065C>T, p.(Arg689*) variant. In the second family, two siblings had alobar holoprosencephaly and cyclopia with a homozygous PLCH1 c.4235delA, p.(Cys1079ValfsTer16) variant. All parents were healthy carriers, with no holoprosencephaly spectrum features. We found that the subcellular localisation of PLCH1 is cytoplasmic, but the p.(Cys1079ValfsTer16) variant was predominantly nuclear. Human embryo immunohistochemistry showed PLCH1 to be expressed in the notorcord, developing spinal cord (in a ventral to dorsal gradient), dorsal root ganglia, cerebellum and dermatomyosome, all tissues producing or responding to SHH. Furthermore, the embryonic subcellular localisation of PLCH1 was exclusively cytoplasmic, supporting protein mislocalisation contributing to the pathogenicity of the p.(Cys1079ValfsTer16) variant. CONCLUSION: Our data support the contention that PLCH1 has a role in prenatal mammalian neurodevelopment, and deleterious variants cause a clinically variable holoprosencephaly spectrum phenotype.


Assuntos
Holoprosencefalia , Fosfolipases Tipo C , Animais , Proteínas Hedgehog/genética , Holoprosencefalia/genética , Holoprosencefalia/metabolismo , Humanos , Mamíferos/metabolismo , Mutação , Fenótipo , Fosfolipases Tipo C/genética
3.
Int J Mol Sci ; 22(18)2021 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-34576017

RESUMO

The Hedgehog (HH) signalling pathway is one of the major pathways controlling cell differentiation and proliferation during human development. This pathway is complex, with HH function influenced by inhibitors, promotors, interactions with other signalling pathways, and non-genetic and cellular factors. Many aspects of this pathway are not yet clarified. The main features of Sonic Hedgehog (SHH) signalling are discussed in relation to its function in human development. The possible role of SHH will be considered using examples of holoprosencephaly and short-rib polydactyly (SRP) syndromes. In these syndromes, there is wide variability in phenotype even with the same genetic mutation, so that other factors must influence the outcome. SHH mutations were the first identified genetic causes of holoprosencephaly, but many other genes and environmental factors can cause malformations in the holoprosencephaly spectrum. Many patients with SRP have genetic defects affecting primary cilia, structures found on most mammalian cells which are thought to be necessary for canonical HH signal transduction. Although SHH signalling is affected in both these genetic conditions, there is little overlap in phenotype. Possible explanations will be canvassed, using data from published human and animal studies. Implications for the understanding of SHH signalling in humans will be discussed.


Assuntos
Proteínas Hedgehog/metabolismo , Holoprosencefalia/etiologia , Síndrome de Costela Curta e Polidactilia/etiologia , Animais , Cílios/metabolismo , Ciliopatias/etiologia , Ciliopatias/metabolismo , Holoprosencefalia/metabolismo , Humanos , Síndrome de Costela Curta e Polidactilia/metabolismo , Transdução de Sinais
4.
J Clin Endocrinol Metab ; 105(9)2020 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32403133

RESUMO

CONTEXT: In human, Sonic hedgehog (SHH) haploinsufficiency is the predominant cause of holoprosencephaly, a structural malformation of the forebrain midline characterized by phenotypic heterogeneity and incomplete penetrance. The NOTCH signaling pathway has recently been associated with holoprosencephaly in humans, but the precise mechanism involving NOTCH signaling during early brain development remains unknown. OBJECTIVE: The aim of this study was to evaluate the relationship between SHH and NOTCH signaling to determine the mechanism by which NOTCH dysfunction could cause midline malformations of the forebrain. DESIGN: In this study, we have used a chemical inhibition approach in the chick model and a genetic approach in the mouse model. We also reported results obtained from the clinical diagnosis of a cohort composed of 141 holoprosencephaly patients. RESULTS: We demonstrated that inhibition of NOTCH signaling in chick embryos as well as in mouse embryos induced a specific downregulation of SHH in the anterior hypothalamus. Our data in the mouse also revealed that the pituitary gland was the most sensitive tissue to Shh insufficiency and that haploinsufficiency of the SHH and NOTCH signaling pathways synergized to produce a malformed pituitary gland. Analysis of a large holoprosencephaly cohort revealed that some patients possessed multiple heterozygous mutations in several regulators of both pathways. CONCLUSIONS: These results provided new insights into molecular mechanisms underlying the extreme phenotypic variability observed in human holoprosencephaly. They showed how haploinsufficiency of the SHH and NOTCH activity could contribute to specific congenital hypopituitarism that was associated with a sella turcica defect.


Assuntos
Proteínas Hedgehog/genética , Holoprosencefalia/genética , Sistema Hipotálamo-Hipofisário/metabolismo , Receptores Notch/genética , Animais , Células Cultivadas , Embrião de Galinha , Estudos de Coortes , Modelos Animais de Doenças , Embrião de Mamíferos , Feminino , Haploinsuficiência/genética , Proteínas Hedgehog/metabolismo , Holoprosencefalia/metabolismo , Holoprosencefalia/patologia , Holoprosencefalia/fisiopatologia , Humanos , Sistema Hipotálamo-Hipofisário/patologia , Masculino , Camundongos , Camundongos Transgênicos , Gravidez , Receptores Notch/deficiência , Estudos Retrospectivos , Transdução de Sinais/genética
5.
Birth Defects Res ; 111(19): 1561-1563, 2019 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-31419067

RESUMO

BACKGROUND: The extraction and processing of copper and cobalt in the African Copperbelt in the Democratic Republic of Congo have led to substantial environmental pollution, causing concerns about possible adverse effects on human health, including birth defects. CASES: We report three neonates with clinically diagnosed holoprosencephaly who were part of a case-control study performed in Lubumbashi between February 2013 and February 2015. One mother had a high concentration of uranium in urine, and high manganese concentrations were found in blood of another mother and in cord blood of one infant. Two of the three fathers had a mining-related job. DISCUSSION: We hypothesize that these cases of holoprosencephaly were connected to mining-related pollution, possibly via epigenetic alterations induced by paternal occupational exposure to toxic metals.


Assuntos
Cobre/efeitos adversos , Holoprosencefalia/etiologia , Manganês/efeitos adversos , Urânio/efeitos adversos , Adulto , Estudos de Casos e Controles , República Democrática do Congo , Exposição Ambiental/efeitos adversos , Feminino , Holoprosencefalia/metabolismo , Humanos , Lactente , Recém-Nascido , Masculino , Mineração , Exposição Ocupacional/efeitos adversos
6.
Eur J Hum Genet ; 27(7): 1113-1120, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30787447

RESUMO

Hartsfield syndrome (HS) is an ultrarare developmental disorder mainly featuring holoprosencephaly and ectrodactyly. It is caused by heterozygous or biallelic variants in FGFR1. Recently, a dominant-negative effect was suggested for FGFR1 variants associated with HS. Here, exome sequencing analysis in a 12-year-old boy with HS disclosed a novel de novo heterozygous variant c.1934C>T in FGFR1 predicted to cause the p.(Ala645Val) amino-acid substitution. In order to evaluate whether the variant, changing a highly conserved residue of the kinase domain, affects FGFR1 function, biochemical studies were employed. We measured the FGFR1 receptor activity in FGF2-treated cell lines exogenously expressing wild-type or Ala645Val FGFR1 by monitoring the activation status of FGF2/FGFR1 downstream pathways. Our analysis highlighted that RAS/ERK1/2 signaling was significantly perturbed in cells expressing mutated FGFR1, in comparison with control cells. We also provided preliminary evidence showing a modulation of the autophagic process in cells expressing mutated FGFR1. This study expands the FGFR1 mutational spectrum associated with HS, provides functional evidence further supporting a dominant-negative effect of this category of FGFR1 variants and offers initial insights on dysregulation of autophagy in HS.


Assuntos
Fenda Labial , Fissura Palatina , Dedos/anormalidades , Deformidades Congênitas da Mão , Holoprosencefalia , Deficiência Intelectual , Sistema de Sinalização das MAP Quinases , Mutação de Sentido Incorreto , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Substituição de Aminoácidos , Fenda Labial/genética , Fenda Labial/metabolismo , Fenda Labial/patologia , Fissura Palatina/genética , Fissura Palatina/metabolismo , Fissura Palatina/patologia , Feminino , Dedos/patologia , Genes Dominantes , Deformidades Congênitas da Mão/genética , Deformidades Congênitas da Mão/metabolismo , Deformidades Congênitas da Mão/patologia , Holoprosencefalia/genética , Holoprosencefalia/metabolismo , Holoprosencefalia/patologia , Humanos , Deficiência Intelectual/genética , Deficiência Intelectual/metabolismo , Deficiência Intelectual/patologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
7.
Hum Mutat ; 39(10): 1416-1427, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-29992659

RESUMO

Here, we applied targeted capture to examine 153 genes representative of all the major vertebrate developmental pathways among 333 probands to rank their relative significance as causes for holoprosencephaly (HPE). We now show that comparisons of variant transmission versus nontransmission among 136 HPE Trios indicates some reported genes now lack confirmation, while novel genes are implicated. Furthermore, we demonstrate that variation of modest intrinsic effect can synergize with these driver mutations as gene modifiers.


Assuntos
Fatores de Crescimento de Fibroblastos/metabolismo , Predisposição Genética para Doença , Proteínas Hedgehog/metabolismo , Holoprosencefalia/genética , Holoprosencefalia/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Fatores de Crescimento de Fibroblastos/genética , Frequência do Gene , Estudos de Associação Genética , Genótipo , Proteínas Hedgehog/genética , Holoprosencefalia/diagnóstico , Humanos , Padrões de Herança , Mutação , Fenótipo , Síndrome , Fator de Crescimento Transformador beta/genética
8.
Am J Med Genet C Semin Med Genet ; 178(2): 128-139, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29749689

RESUMO

Holoprosencephaly (HPE) is a frequent human forebrain developmental disorder with both genetic and environmental causes. Multiple loci have been associated with HPE in humans, and potential causative genes at 14 of these loci have been identified. Although TGIF1 (originally TGIF, for Thymine Guanine-Interacting Factor) is among the most frequently screened genes in HPE patients, an understanding of how mutations in this gene contribute to the pathogenesis of HPE has remained elusive. However, mouse models based on loss of function of Tgif1, and the related Tgif2 gene, have shed some light on how human TGIF1 variants might cause HPE. Functional analyses of TGIF proteins and of TGIF1 single nucleotide variants from HPE patients, combined with analysis of forebrain development in mouse embryos lacking both Tgif1 and Tgif2, suggest that TGIFs regulate the transforming growth factor ß/Nodal signaling pathway and sonic hedgehog (SHH) signaling independently. Although, some developmental processes that are regulated by TGIFs may be Nodal-dependent, it appears that the forebrain patterning defects and HPE in Tgif mutant mouse embryos is primarily due to altered signaling via the Shh pathway.


Assuntos
Encéfalo/embriologia , Encéfalo/metabolismo , Suscetibilidade a Doenças , Holoprosencefalia/etiologia , Holoprosencefalia/metabolismo , Proteínas de Homeodomínio/genética , Organogênese/genética , Proteínas Repressoras/genética , Animais , Padronização Corporal , Encéfalo/anormalidades , Encéfalo/ultraestrutura , Anormalidades Craniofaciais/etiologia , Anormalidades Craniofaciais/metabolismo , Deficiências do Desenvolvimento/etiologia , Deficiências do Desenvolvimento/metabolismo , Modelos Animais de Doenças , Deleção de Genes , Regulação da Expressão Gênica , Variação Genética , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Proteína Nodal/genética , Proteína Nodal/metabolismo , Prosencéfalo/embriologia , Prosencéfalo/metabolismo , Proteínas Repressoras/metabolismo , Transdução de Sinais , Transcrição Gênica
9.
Am J Med Genet C Semin Med Genet ; 178(2): 140-150, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29749693

RESUMO

Holoprosencephaly (HPE) is a common developmental defect caused by failure to define the midline of the forebrain and/or midface. HPE is associated with heterozygous mutations in Nodal and Sonic hedgehog (SHH) pathway components, but clinical presentation is highly variable, and many mutation carriers are unaffected. It is therefore thought that such mutations interact with more common modifiers, genetic and/or environmental, to produce severe patterning defects. Modifiers are difficult to identify, as their effects are context-dependent and occur within the complex genetic and environmental landscapes that characterize human populations. This has made a full understanding of HPE etiology challenging. We discuss here the use of mice, a genetically tractable model sensitive to teratogens, as a system to address this challenge. Mice carrying mutations in human HPE genes often display wide variations in phenotypic penetrance and expressivity when placed on different genetic backgrounds, demonstrating the existence of silent HPE modifier genes. Studies with mouse lines carrying SHH pathway mutations on appropriate genetic backgrounds have led to identification of both genetic and environmental modifiers that synergize with the mutations to produce a spectrum of HPE phenotypes. These models favor a scenario in which multiple modifying influences-both genetic and environmental, sensitizing and protective-interact with bona fide HPE mutations to grade phenotypic outcomes. Despite the complex interplay of HPE risk factors, mouse models have helped establish some clear concepts in HPE etiology. A combination of mouse and human cohort studies should improve our understanding of this fascinating and medically important issue.


Assuntos
Holoprosencefalia/etiologia , Modelos Biológicos , Herança Multifatorial , Animais , Biomarcadores , Modelos Animais de Doenças , Epistasia Genética , Interação Gene-Ambiente , Estudos de Associação Genética , Predisposição Genética para Doença , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Holoprosencefalia/diagnóstico , Holoprosencefalia/metabolismo , Humanos , Camundongos , Camundongos Knockout , Mutação , Proteína Nodal/genética , Proteína Nodal/metabolismo , Fenótipo , Transdução de Sinais
10.
Am J Med Genet C Semin Med Genet ; 178(2): 246-257, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29761634

RESUMO

Nonchromosomal, nonsyndromic holoprosencephaly (NCNS-HPE) has traditionally been considered as a condition of brain and craniofacial maldevelopment. In this review, we present the results of a comprehensive literature search supporting a wide spectrum of extracephalic manifestations identified in patients with NCNS-HPE. These manifestations have been described in case reports and in large cohorts of patients with "single-gene" mutations, suggesting that the NCNS-HPE phenotype can be more complex than traditionally thought. Likely, a complex network of interacting genetic variants and environmental factors is responsible for these systemic abnormalities that deviate from the usual brain and craniofacial findings in NCNS-HPE. In addition to the systemic consequences of pituitary dysfunction (as a direct result of brain midline defects), here we describe a number of extracephalic findings of NCNS-HPE affecting various organ systems. It is our goal to provide a guide of extracephalic features for clinicians given the important clinical implications of these manifestations for the management and care of patients with HPE and their mutation-positive relatives. The health risks associated with some manifestations (e.g., fatty liver disease) may have historically been neglected in affected families.


Assuntos
Suscetibilidade a Doenças , Holoprosencefalia/diagnóstico , Holoprosencefalia/etiologia , Fenótipo , Anormalidades Múltiplas/diagnóstico , Anormalidades Múltiplas/etiologia , Anormalidades Múltiplas/metabolismo , Biomarcadores , Doenças do Sistema Endócrino/congênito , Predisposição Genética para Doença , Proteínas Hedgehog/genética , Proteínas Hedgehog/metabolismo , Holoprosencefalia/metabolismo , Humanos , Mutação , Transdução de Sinais
11.
Biochem Biophys Res Commun ; 496(2): 575-581, 2018 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-29355528

RESUMO

Human protein TGIF1 is an essential regulator of cell fate with broad roles in different tissues, and has been implicated in holoprosencephaly (HPE) and many cancers. The function of TGIF1 in transcriptional regulation depends on its three-amino acid loop extension (TALE) type of homeodomain (HD). Two missense mutations that led to P192A and R219C substitutions in TGIF1-HD were previously found in HPE patients and suggested to be the causes for these cases. However, how these mutations affected TGIF1 function has not been investigated from a structural view. Here, we investigated the roles of P192 and R219 in TGIF1-HD structure packing through determining the NMR structure of TGIF1-HD. Surprisingly, P192 and R219 were found to play roles in packing α1 and α2 to α3 together with A190 and F215 through side-chain interactions. Circular dichroism (CD) showed that P192A and R219C mutants displayed structural change and less folding compared with wild-type TGIF1-HD, and 1H-15N HSQC spectrum of P192A mutant exhibited chemical shift perturbations in all three helices of TGIF1-HD. Thus, it is suggested that P192A and R219C mutations led to structure disturbances of TGIF1-HD, which subsequently reduced the DNA-binding affinity of TGIF1-HD by 23-fold and 10-fold respectively, as revealed by the isothermal titration calorimetry (ITC) experiments. Our study provides structural insights of the probable pathogenesis mechanism of two TGIF1-related HPE cases, and evidences for the roles of P192 and R219 in HD folding.


Assuntos
Holoprosencefalia/genética , Proteínas de Homeodomínio/química , Proteínas de Homeodomínio/genética , Mutação Puntual , Proteínas Repressoras/química , Proteínas Repressoras/genética , Sequência de Aminoácidos , DNA/metabolismo , Holoprosencefalia/metabolismo , Proteínas de Homeodomínio/metabolismo , Humanos , Modelos Moleculares , Mutação de Sentido Incorreto , Ressonância Magnética Nuclear Biomolecular , Conformação Proteica , Dobramento de Proteína , Proteínas Repressoras/metabolismo
12.
Congenit Anom (Kyoto) ; 58(1): 29-32, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28670735

RESUMO

Holoprosencephaly (HPE) is failure of the forebrain to divide completely during embryogenesis. Incomplete penetrance has not been reported previously in SIX3 whole gene deletions, which are known to cause HPE. Both chromosomal microarray and whole exome sequencing (WES) were used to evaluate families with inherited HPE. Two families showed inherited deletions that contain SIX3 and were incompletely penetrant for HPE. Using WES, we ruled out parental mosaicism, a SIX3 hypomorph, and clinically significant variants in genes that are known to interact with SIX3 as causes of incomplete penetrance. We demonstrate the importance of molecular cascade testing in families with HPE and we answer important questions about incomplete penetrance.


Assuntos
Proteínas do Olho/genética , Deleção de Genes , Holoprosencefalia/genética , Proteínas de Homeodomínio/genética , Proteínas do Tecido Nervoso/genética , Prosencéfalo/anormalidades , Adulto , Pré-Escolar , Expressão Gênica , Holoprosencefalia/diagnóstico , Holoprosencefalia/metabolismo , Holoprosencefalia/patologia , Humanos , Lactente , Recém-Nascido , Análise em Microsséries , Proteínas do Tecido Nervoso/deficiência , Penetrância , Prosencéfalo/metabolismo , Sequenciamento do Exoma , Proteína Homeobox SIX3
13.
Dev Biol ; 430(1): 156-165, 2017 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-28800946

RESUMO

Tctn1, Tctn2, and Tctn3 are membrane proteins that localize at the transition zone of primary cilia. Tctn1 and Tctn2 mutations have been reported in both humans and mice, but Tctn3 mutations have been reported only in humans. It is also not clear whether the three Tctn proteins are functionally conserved with respect to ciliogenesis and Hedgehog (Hh) signaling. In the present study, we report that loss of Tctn3 gene function in mice results in a decrease in ciliogenesis and Hh signaling. Consistent with this, Tctn3 mutant mice exhibit holoprosencephaly and randomized heart looping and lack the floor plate in the neural tube, the phenotypes similar to those of Tctn1 and Tctn2 mutants. We also show that overexpression of Tctn3, but not Tctn1 or Tctn2, can rescue ciliogenesis in Tctn3 mutant cells. Similarly, replacement of Tctn3 with Tctn1 or Tctn2 in the Tctn3 gene locus results in reduced ciliogenesis and Hh signaling, holoprosencephaly, and randomized heart looping. Surprisingly, however, the neural tube patterning and the proteolytic processing of Gli3 (a transcription regulator for Hh signaling) into a repressor, both of which are usually impaired in ciliary gene mutants, are normal. These results suggest that Tctn1, Tctn2, and Tctn3 are functionally divergent with respect to their role in ciliogenesis and Hh signaling but conserved in neural tube patterning and Gli3 processing.


Assuntos
Padronização Corporal , Cílios/metabolismo , Proteínas Hedgehog/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas de Membrana/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Tubo Neural/embriologia , Tubo Neural/metabolismo , Organogênese , Proteínas Adaptadoras de Transdução de Sinal , Animais , Proteínas Reguladoras de Apoptose , Padronização Corporal/genética , Sequência Conservada , Embrião de Mamíferos/metabolismo , Desenvolvimento Embrionário , Fibroblastos/metabolismo , Deleção de Genes , Inativação Gênica , Holoprosencefalia/metabolismo , Holoprosencefalia/patologia , Homozigoto , Camundongos , Organogênese/genética , Transdução de Sinais/genética , Proteína Gli3 com Dedos de Zinco
14.
Hum Mutat ; 38(11): 1464-1470, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-28677295

RESUMO

Holoprosencephaly (HPE), a common developmental defect of the forebrain and midface, has a complex etiology. Heterozygous, loss-of-function mutations in the sonic hedgehog (SHH) pathway are associated with HPE. However, mutation carriers display highly variable clinical presentation, leading to an "autosomal dominant with modifier" model, in which the penetrance and expressivity of a predisposing mutation is graded by genetic or environmental modifiers. Such modifiers have not been identified. Boc encodes a SHH coreceptor and is a silent HPE modifier gene in mice. Here, we report the identification of missense BOC variants in HPE patients. Consistent with these alleles functioning as HPE modifiers, individual variant BOC proteins had either loss- or gain-of-function properties in cell-based SHH signaling assays. Therefore, in addition to heterozygous loss-of-function mutations in specific SHH pathway genes and an ill-defined environmental component, our findings identify a third variable in HPE: low-frequency modifier genes, BOC being the first identified.


Assuntos
Genes Modificadores , Holoprosencefalia/genética , Imunoglobulina G/genética , Receptores de Superfície Celular/genética , Animais , Expressão Gênica , Variação Genética , Holoprosencefalia/metabolismo , Humanos , Imunoglobulina G/química , Imunoglobulina G/metabolismo , Camundongos , Modelos Moleculares , Mutação , Conformação Proteica , Domínios Proteicos , Receptores de Superfície Celular/química , Receptores de Superfície Celular/metabolismo
15.
Pflugers Arch ; 469(7-8): 907-916, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28497274

RESUMO

Megalin (or LRP2) is an endocytic receptor that plays a central role in embryonic development and adult tissue homeostasis. Loss of this receptor in congenital or acquired diseases results in multiple organ dysfunctions, including forebrain malformation (holoprosencephaly) and renal reabsorption defects (renal Fanconi syndrome). Here, we describe current concepts of the mode of receptor action that include co-receptors and a repertoire of different ligands, and we discuss how these interactions govern functional integrity of the kidney and the brain, and cause disease when defective.


Assuntos
Síndrome de Fanconi/metabolismo , Holoprosencefalia/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/metabolismo , Animais , Encéfalo/crescimento & desenvolvimento , Encéfalo/metabolismo , Endocitose , Síndrome de Fanconi/genética , Holoprosencefalia/genética , Humanos , Túbulos Renais Proximais/crescimento & desenvolvimento , Túbulos Renais Proximais/metabolismo , Proteína-2 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Reabsorção Renal
16.
Proc Natl Acad Sci U S A ; 113(40): E5866-E5875, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27647915

RESUMO

The Hedgehog cell-cell signaling pathway is crucial for animal development, and its misregulation is implicated in numerous birth defects and cancers. In unstimulated cells, pathway activity is inhibited by the tumor suppressor membrane protein, Patched. Hedgehog signaling is triggered by the secreted Hedgehog ligand, which binds and inhibits Patched, thus setting in motion the downstream events in signal transduction. Despite its critical importance, the mechanism by which Hedgehog antagonizes Patched has remained unknown. Here, we show that vertebrate Patched1 inhibition is caused by direct, palmitate-dependent interaction with the Sonic Hedgehog ligand. We find that a short palmitoylated N-terminal fragment of Sonic Hedgehog binds Patched1 and, strikingly, is sufficient to inhibit it and to activate signaling. The rest of Sonic Hedgehog confers high-affinity Patched1 binding and internalization through a distinct binding site, but, surprisingly, it is not absolutely required for signaling. The palmitate-dependent interaction with Patched1 is specifically impaired in a Sonic Hedgehog mutant causing human holoprosencephaly, the most frequent congenital brain malformation, explaining its drastically reduced potency. The palmitate-dependent interaction is also abolished in constitutively inhibited Patched1 point mutants causing the Gorlin cancer syndrome, suggesting that they might adopt a conformation distinct from the wild type. Our data demonstrate that Sonic Hedgehog signals via the palmitate-dependent arm of a two-pronged contact with Patched1. Furthermore, our results suggest that, during Hedgehog signaling, ligand binding inhibits Patched by trapping it in an inactive conformation, a mechanism that explains the dramatically reduced activity of oncogenic Patched1 mutants.


Assuntos
Proteínas Hedgehog/metabolismo , Receptores Patched/metabolismo , Transdução de Sinais , Células 3T3 , Animais , Endocitose/efeitos dos fármacos , Holoprosencefalia/metabolismo , Holoprosencefalia/patologia , Humanos , Lipoilação , Camundongos , Modelos Moleculares , Mutação/genética , Oncogenes , Ácido Palmítico/metabolismo , Peptídeos/farmacologia , Ligação Proteica/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
17.
PLoS One ; 10(3): e0120517, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25793997

RESUMO

The Hedgehog (Hh) signaling pathway mediates multiple spatiotemporally-specific aspects of brain and face development. Genetic and chemical disruptions of the pathway are known to result in an array of structural malformations, including holoprosencephaly (HPE), clefts of the lip with or without cleft palate (CL/P), and clefts of the secondary palate only (CPO). Here, we examined patterns of dysmorphology caused by acute, stage-specific Hh signaling inhibition. Timed-pregnant wildtype C57BL/6J mice were administered a single dose of the potent pathway antagonist vismodegib at discrete time points between gestational day (GD) 7.0 and 10.0, an interval approximately corresponding to the 15th to 24th days of human gestation. The resultant pattern of facial and brain dysmorphology was dependent upon stage of exposure. Insult between GD7.0 and GD8.25 resulted in HPE, with peak incidence following exposure at GD7.5. Unilateral clefts of the lip extending into the primary palate were also observed, with peak incidence following exposure at GD8.875. Insult between GD9.0 and GD10.0 resulted in CPO and forelimb abnormalities. We have previously demonstrated that Hh antagonist-induced cleft lip results from deficiency of the medial nasal process and show here that CPO is associated with reduced growth of the maxillary-derived palatal shelves. By defining the critical periods for the induction of HPE, CL/P, and CPO with fine temporal resolution, these results provide a mechanism by which Hh pathway disruption can result in "non-syndromic" orofacial clefting, or HPE with or without co-occurring clefts. This study also establishes a novel and tractable mouse model of human craniofacial malformations using a single dose of a commercially available and pathway-specific drug.


Assuntos
Anilidas/efeitos adversos , Fenda Labial/patologia , Fissura Palatina/patologia , Proteínas Hedgehog/antagonistas & inibidores , Holoprosencefalia/patologia , Piridinas/efeitos adversos , Transdução de Sinais , Animais , Fenda Labial/induzido quimicamente , Fenda Labial/metabolismo , Fissura Palatina/induzido quimicamente , Fissura Palatina/metabolismo , Face/anormalidades , Feminino , Proteínas Hedgehog/metabolismo , Holoprosencefalia/induzido quimicamente , Holoprosencefalia/metabolismo , Camundongos Endogâmicos C57BL , Morfogênese/efeitos dos fármacos , Fenótipo , Gravidez , Transdução de Sinais/efeitos dos fármacos
18.
Clin Endocrinol (Oxf) ; 82(5): 728-38, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25327282

RESUMO

INTRODUCTION: The Gli family of zinc finger (GLI) transcription factors mediates the sonic hedgehog signalling pathway (HH) essential for CNS, early pituitary and ventral forebrain development in mice. Human mutations in this pathway have been described in patients with holoprosencephaly (HPE), isolated congenital hypopituitarism (CH) and cranial/midline facial abnormalities. Mutations in Sonic hedgehog (SHH) have been associated with HPE but not CH, despite murine studies indicating involvement in pituitary development. OBJECTIVES/METHODS: We aimed to establish the role of the HH pathway in the aetiology of hypothalamo-pituitary disorders by screening our cohort of patients with midline defects and/or CH for mutations in SHH, GLI2, Shh brain enhancer 2 (SBE2) and growth-arrest specific 1 (GAS1). RESULTS: Two variants and a deletion of GLI2 were identified in three patients. A novel variant at a highly conserved residue in the zinc finger DNA-binding domain, c.1552G > A [pE518K], was identified in a patient with growth hormone deficiency and low normal free T4. A nonsynonymous variant, c.2159G > A [p.R720H], was identified in a patient with a short neck, cleft palate and hypogonadotrophic hypogonadism. A 26·6 Mb deletion, 2q12·3-q21·3, encompassing GLI2 and 77 other genes, was identified in a patient with short stature and impaired growth. Human embryonic expression studies and molecular characterisation of the GLI2 mutant p.E518K support the potential pathogenicity of GLI2 mutations. No mutations were identified in GAS1 or SBE2. A novel SHH variant, c.1295T>A [p.I432N], was identified in two siblings with variable midline defects but normal pituitary function. CONCLUSIONS: Our data suggest that mutations in SHH, GAS1 and SBE2 are not associated with hypopituitarism, although GLI2 is an important candidate for CH.


Assuntos
Regulação da Expressão Gênica , Proteínas Hedgehog/genética , Hipopituitarismo/sangue , Transdução de Sinais , Adolescente , Animais , Proteínas de Ciclo Celular/genética , Criança , Pré-Escolar , Estudos de Coortes , Elementos Facilitadores Genéticos/genética , Feminino , Proteínas Ligadas por GPI/genética , Deleção de Genes , Variação Genética , Heterozigoto , Holoprosencefalia/metabolismo , Humanos , Hipopituitarismo/congênito , Hipopituitarismo/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Masculino , Camundongos , Mutação , Células NIH 3T3 , Proteínas Nucleares/genética , Fenótipo , Análise de Sequência de DNA , Proteína Gli2 com Dedos de Zinco , Dedos de Zinco
19.
Brain Dev ; 36(6): 463-71, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24529974

RESUMO

BACKGROUND: Genetic programming of cerebral development involves tissue morphogenesis and also timing of developmental processes. Precocious synaptogenesis in the neocortical plate was previously demonstrated in 5 of 6 fetuses of 20-31 weeks gestation. MATERIALS AND METHODS: Neuropathological examination was performed of a 13-week-5-day fetus with trisomy-13, a lobar holoprosencephaly, hydrocephalus, cyclopia and absence of ears. Immunocytochemical demonstration of the synaptic vesicle protein synaptophysin was performed in the brain and retina, along with other neuronal markers. RESULTS: Synaptophysin reactivity in the cortical plate was patchy and precocious. Radial glial fibres, demonstrated by vimentin, were oriented parallel to the cortical plate rather than perpendicular, probably because of hydrocephalus. A corpus striatum was not identified, but the poorly formed thalamus exhibited synaptophysin reactivity around many neurones. The cyclopean eye had ocular features of maturational delay including persistent hyaloid artery; ganglion cells were reduced in number, but retinal synaptophysin reactivity was paradoxically precocious. CONCLUSIONS: Holoprosencephaly exhibits abnormal patchy synapse distribution in the neocortex and retina; synaptogenesis was precocious, as we previously described in older fetuses. Too soon an onset of synapse formation may promote early epileptic circuitry, leading to severe infantile epilepsies postnatally. The visual system is the last of the special sensory systems to mature, yet in this case showed too early synapse formation. In HPE, cyclopia and in trisomy 13, total absence of external ears has not been reported; it results from faulty craniofacial induction by neural crest.


Assuntos
Córtex Cerebral/patologia , Holoprosencefalia/patologia , Retina/patologia , Sinapses/patologia , Córtex Cerebral/embriologia , Córtex Cerebral/metabolismo , Feminino , Feto , Holoprosencefalia/metabolismo , Humanos , Imuno-Histoquímica , Fotomicrografia , Retina/embriologia , Retina/metabolismo , Sinapses/metabolismo , Sinaptofisina/metabolismo
20.
Clin Exp Obstet Gynecol ; 40(3): 460-2, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24283191

RESUMO

Cyclopia is a rare type of holoprosencephaly and a congenital disorder characterized by the failure of the embryonic forebrain to properly divide the orbits of the eye into two cavities (the embryonic forebrain is normally responsible for inducing the development of the orbits). As a result a birth defect in which there is only one eye is developed. This eye is centrally placed in the area normally occupied by the root of the nose. As a rule, there is a missing nose or a non-functioning nose in the form of a proboscis (a tubular appendage) located above the central eye. In this report the macroscopic, radiographic, and immunohistochemical findings of a case of true cyclopia in a female fetus are described. Cyclopia is a lethal condition that is associated with dramatic symmetric deformities of the nose, skull, orbits, and brain.


Assuntos
Anormalidades Múltiplas , Holoprosencefalia , Polidactilia , Evolução Fatal , Feminino , Feto/patologia , Proteínas Hedgehog/metabolismo , Holoprosencefalia/genética , Holoprosencefalia/metabolismo , Holoprosencefalia/patologia , Humanos , Imuno-Histoquímica , Polidactilia/complicações , Natimorto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA