Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
2.
Am J Kidney Dis ; 77(3): 410-419, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33039432

RESUMO

Primary cilia are specialized sensory organelles that protrude from the apical surface of most cell types. During the past 2 decades, they have been found to play important roles in tissue development and signal transduction, with mutations in ciliary-associated proteins resulting in a group of diseases collectively known as ciliopathies. Many of these mutations manifest as renal ciliopathies, characterized by kidney dysfunction resulting from aberrant cilia or ciliary functions. This group of overlapping and genetically heterogeneous diseases includes polycystic kidney disease, nephronophthisis, and Bardet-Biedl syndrome as the main focus of this review. Renal ciliopathies are characterized by the presence of kidney cysts that develop due to uncontrolled epithelial cell proliferation, growth, and polarity, downstream of dysregulated ciliary-dependent signaling. Due to cystic-associated kidney injury and systemic inflammation, cases result in kidney failure requiring dialysis and transplantation. Of the handful of pharmacologic treatments available, none are curative. It is important to determine the molecular mechanisms that underlie the involvement of the primary cilium in cyst initiation, expansion, and progression for the development of novel and efficacious treatments. This review updates research progress in defining key genes and molecules central to ciliogenesis and renal ciliopathies.


Assuntos
Síndrome de Bardet-Biedl/genética , Cílios/metabolismo , Ciliopatias/genética , Doenças Renais Policísticas/genética , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transporte Vesicular/genética , Síndrome de Bardet-Biedl/metabolismo , Síndrome de Bardet-Biedl/fisiopatologia , Cerebelo/anormalidades , Cerebelo/metabolismo , Cerebelo/fisiopatologia , Chaperoninas/genética , Cílios/fisiologia , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/fisiopatologia , Ciliopatias/metabolismo , Ciliopatias/fisiopatologia , Proteínas do Citoesqueleto/genética , Encefalocele/genética , Encefalocele/metabolismo , Encefalocele/fisiopatologia , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Anormalidades do Olho/fisiopatologia , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/fisiopatologia , Amaurose Congênita de Leber/genética , Amaurose Congênita de Leber/metabolismo , Amaurose Congênita de Leber/fisiopatologia , Proteínas de Membrana/genética , Proteínas Associadas aos Microtúbulos/genética , Atrofias Ópticas Hereditárias/genética , Atrofias Ópticas Hereditárias/metabolismo , Atrofias Ópticas Hereditárias/fisiopatologia , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/fisiopatologia , Proteínas/genética , Retina/anormalidades , Retina/metabolismo , Retina/fisiopatologia , Retinose Pigmentar/genética , Retinose Pigmentar/metabolismo , Retinose Pigmentar/fisiopatologia , Canais de Cátion TRPP/genética
3.
Sci Rep ; 9(1): 1069, 2019 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-30705305

RESUMO

Transmembrane protein 67 (TMEM67) is mutated in Meckel Gruber Syndrome type 3 (MKS3) resulting in a pleiotropic phenotype with hydrocephalus and renal cystic disease in both humans and rodent models. The precise pathogenic mechanisms remain undetermined. Herein it is reported for the first time that a point mutation of TMEM67 leads to a gene dose-dependent hydrocephalic phenotype in the Wistar polycystic kidney (Wpk) rat. Animals with TMEM67 heterozygous mutations manifest slowly progressing hydrocephalus, observed during the postnatal period and continuing into adulthood. These animals have no overt renal phenotype. The TMEM67 homozygous mutant rats have severe ventriculomegaly as well as severe polycystic kidney disease and die during the neonatal period. Protein localization in choroid plexus epithelial cells indicates that aquaporin 1 and claudin-1 both remain normally polarized in all genotypes. The choroid plexus epithelial cells may have selectively enhanced permeability as evidenced by increased Na+, K+ and Cl- in the cerebrospinal fluid of the severely hydrocephalic animals. Collectively, these results suggest that TMEM67 is required for the regulation of choroid plexus epithelial cell fluid and electrolyte homeostasis. The Wpk rat model, orthologous to human MKS3, provides a unique platform to study the development of both severe and mild hydrocephalus.


Assuntos
Transtornos da Motilidade Ciliar/metabolismo , Encefalocele/metabolismo , Hidrocefalia/metabolismo , Proteínas de Membrana/metabolismo , Doenças Renais Policísticas/metabolismo , Retinose Pigmentar/metabolismo , Animais , Encéfalo/metabolismo , Cloretos/líquido cefalorraquidiano , Plexo Corióideo/metabolismo , Transtornos da Motilidade Ciliar/genética , Encefalocele/genética , Feminino , Hidrocefalia/genética , Proteínas de Membrana/genética , Mutação/genética , Doenças Renais Policísticas/genética , Potássio/líquido cefalorraquidiano , Ratos , Retinose Pigmentar/genética , Sódio/líquido cefalorraquidiano
4.
FASEB J ; 33(4): 4688-4702, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30592646

RESUMO

Folate deficiency in early development leads to disturbance in multiple processes, including neurogenesis during which fibroblast growth factor (FGF) pathway is one of the crucial pathways. Whether folic acid (FA) directly affects FGF pathways to influence neurodevelopment and the possible mechanism remains unclear. In this study, we presented evidence that in human FA-insufficient encephalocele, the FGF pathway was interfered. Furthermore, in Brachyury knockout mice devoid of such T-box transcription factors regulating embryonic neuromesodermal bipotency and a key component of FGF pathway, change in expression of Brachyury downstream targets, activator Fgf8 and suppressor dual specificity phosphatase 6 was detected, along with the reduction in expression of other key FGF pathway genes. By using a FA-deficient cell model, we further demonstrated that decrease in Brachyury expression was through alteration in hypermethylation at the Brachyury promoter region under FA deficiency conditions, and suppression of Brachyury promoted the inactivation of the FGF pathway. Correspondingly, FA supplementation partially reverses the effects seen in FA-deficient embryoid bodies. Lastly, in mice with maternal folate-deficient diets, aberrant FGF pathway activity was found in fetal brain dysplasia. Taken together, our findings highlight the effect of FA on FGF pathways during neurogenesis, and the mechanism may be due to the low expression of Brachyury gene via hypermethylation under FA-insufficient conditions.-Chang, S., Lu, X., Wang, S., Wang, Z., Huo, J., Huang, J., Shangguan, S., Li, S., Zou, J., Bao, Y., Guo, J., Wang, F., Niu, B., Zhang, T., Qiu, Z., Wu, J., Wang, L. The effect of folic acid deficiency on FGF pathway via Brachyury regulation in neural tube defects.


Assuntos
Proteínas Fetais/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Deficiência de Ácido Fólico/metabolismo , Ácido Fólico/uso terapêutico , Defeitos do Tubo Neural/tratamento farmacológico , Defeitos do Tubo Neural/metabolismo , Proteínas com Domínio T/metabolismo , Animais , Apoptose/efeitos dos fármacos , Western Blotting , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Imunoprecipitação da Cromatina , Encefalocele/metabolismo , Feminino , Deficiência de Ácido Fólico/fisiopatologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Regiões Promotoras Genéticas , Transdução de Sinais/efeitos dos fármacos , Sulfitos/farmacologia
5.
Matrix Biol ; 57-58: 55-75, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27746220

RESUMO

Collagen XVIII is a ubiquitous basement membrane (BM) proteoglycan produced in three tissue-specific isoforms that differ in their N-terminal non-collagenous sequences, but share collagenous and C-terminal non-collagenous domains. The collagenous domain provides flexibility to the large collagen XVIII molecules on account of multiple interruptions in collagenous sequences. Each isoform has a complex multi-domain structure that endows it with an ability to perform various biological functions. The long isoform contains a frizzled-like (Fz) domain with Wnt-inhibiting activity and a unique domain of unknown function (DUF959), which is also present in the medium isoform. All three isoforms share an N-terminal laminin-G-like/thrombospondin-1 sequence whose specific functions still remain unconfirmed. The proteoglycan nature of the isoforms further increases the functional diversity of collagen XVIII. An anti-angiogenic domain termed endostatin resides in the C-terminus of collagen XVIII and is proteolytically cleaved from the parental molecule during the BM breakdown for example in the process of tumour progression. Recombinant endostatin can efficiently reduce tumour angiogenesis and growth in experimental models by inhibiting endothelial cell migration and proliferation or by inducing their death, but its efficacy against human cancers is still a subject of debate. Mutations in the COL18A1 gene result in Knobloch syndrome, a genetic disorder characterised mainly by severe eye defects and encephalocele and, occasionally, other symptoms. Studies with gene-modified mice have elucidated some aspects of this rare disease, highlighting in particular the importance of collagen XVIII in the development of the eye. Research with model organisms have also helped in determining other structural and biological functions of collagen XVIII, such as its requirement in the maintenance of BM integrity and its emerging roles in regulating cell survival, stem or progenitor cell maintenance and differentiation and inflammation. In this review, we summarise current knowledge on the properties and endogenous functions of collagen XVIII in normal situations and tissue dysregulation. When data is available, we discuss the functions of the distinct isoforms and their specific domains.


Assuntos
Membrana Basal/efeitos dos fármacos , Colágeno Tipo VIII/genética , Encefalocele/genética , Neoplasias/genética , Neovascularização Patológica/prevenção & controle , Descolamento Retiniano/congênito , Animais , Membrana Basal/metabolismo , Membrana Basal/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Colágeno Tipo VIII/metabolismo , Colágeno Tipo XVIII , Encefalocele/metabolismo , Encefalocele/patologia , Endostatinas/farmacologia , Células Endoteliais/efeitos dos fármacos , Regulação da Expressão Gênica , Homeostase/genética , Humanos , Neoplasias/metabolismo , Neoplasias/patologia , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/patologia , Domínios Proteicos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteólise , Proteínas Recombinantes/farmacologia , Degeneração Retiniana , Descolamento Retiniano/genética , Descolamento Retiniano/metabolismo , Descolamento Retiniano/patologia
6.
PLoS Genet ; 11(10): e1005575, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26485645

RESUMO

Ciliopathies are a group of human disorders caused by dysfunction of primary cilia, ubiquitous microtubule-based organelles involved in transduction of extra-cellular signals to the cell. This function requires the concentration of receptors and channels in the ciliary membrane, which is achieved by complex trafficking mechanisms, in part controlled by the small GTPase RAB8, and by sorting at the transition zone located at the entrance of the ciliary compartment. Mutations in the transition zone gene CC2D2A cause the related Joubert and Meckel syndromes, two typical ciliopathies characterized by central nervous system malformations, and result in loss of ciliary localization of multiple proteins in various models. The precise mechanisms by which CC2D2A and other transition zone proteins control protein entrance into the cilium and how they are linked to vesicular trafficking of incoming cargo remain largely unknown. In this work, we identify the centrosomal protein NINL as a physical interaction partner of CC2D2A. NINL partially co-localizes with CC2D2A at the base of cilia and ninl knockdown in zebrafish leads to photoreceptor outer segment loss, mislocalization of opsins and vesicle accumulation, similar to cc2d2a-/- phenotypes. Moreover, partial ninl knockdown in cc2d2a-/- embryos enhances the retinal phenotype of the mutants, indicating a genetic interaction in vivo, for which an illustration is found in patients from a Joubert Syndrome cohort. Similar to zebrafish cc2d2a mutants, ninl morphants display altered Rab8a localization. Further exploration of the NINL-associated interactome identifies MICAL3, a protein known to interact with Rab8 and to play an important role in vesicle docking and fusion. Together, these data support a model where CC2D2A associates with NINL to provide a docking point for cilia-directed cargo vesicles, suggesting a mechanism by which transition zone proteins can control the protein content of the ciliary compartment.


Assuntos
Cerebelo/anormalidades , Transtornos da Motilidade Ciliar/genética , Encefalocele/genética , Proteínas Associadas aos Microtúbulos/metabolismo , Oxigenases de Função Mista/genética , Proteínas Nucleares/metabolismo , Doenças Renais Policísticas/genética , Proteínas/genética , Retina/anormalidades , Proteínas rab de Ligação ao GTP/genética , Anormalidades Múltiplas/genética , Anormalidades Múltiplas/metabolismo , Anormalidades Múltiplas/patologia , Animais , Cerebelo/metabolismo , Cerebelo/patologia , Cílios/genética , Cílios/metabolismo , Cílios/patologia , Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/patologia , Proteínas do Citoesqueleto , Encefalocele/metabolismo , Encefalocele/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/metabolismo , Anormalidades do Olho/patologia , Técnicas de Silenciamento de Genes , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/metabolismo , Doenças Renais Císticas/patologia , Proteínas Associadas aos Microtúbulos/genética , Oxigenases de Função Mista/metabolismo , Mutação , Proteínas Nucleares/genética , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Transporte Proteico/genética , Proteínas/metabolismo , Retina/metabolismo , Retina/patologia , Retinose Pigmentar , Transdução de Sinais , Peixe-Zebra , Proteínas rab de Ligação ao GTP/metabolismo
7.
Hum Mol Genet ; 24(17): 4997-5014, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26071364

RESUMO

Agenesis of the corpus callosum (AgCC) is a frequent brain disorder found in over 80 human congenital syndromes including ciliopathies. Here, we report a severe AgCC in Ftm/Rpgrip1l knockout mouse, which provides a valuable model for Meckel-Grüber syndrome. Rpgrip1l encodes a protein of the ciliary transition zone, which is essential for ciliogenesis in several cell types in mouse including neuroepithelial cells in the developing forebrain. We show that AgCC in Rpgrip1l(-/-) mouse is associated with a disturbed location of guidepost cells in the dorsomedial telencephalon. This mislocalization results from early patterning defects and abnormal cortico-septal boundary (CSB) formation in the medial telencephalon. We demonstrate that all these defects primarily result from altered GLI3 processing. Indeed, AgCC, together with patterning defects and mispositioning of guidepost cells, is rescued by overexpressing in Rpgrip1l(-/-) embryos, the short repressor form of the GLI3 transcription factor (GLI3R), provided by the Gli3(Δ699) allele. Furthermore, Gli3(Δ699) also rescues AgCC in Rfx3(-/-) embryos deficient for the ciliogenic RFX3 transcription factor that regulates the expression of several ciliary genes. These data demonstrate that GLI3 processing is a major outcome of primary cilia function in dorsal telencephalon morphogenesis. Rescuing CC formation in two independent ciliary mutants by GLI3(Δ699) highlights the crucial role of primary cilia in maintaining the proper level of GLI3R required for morphogenesis of the CC.


Assuntos
Cílios/metabolismo , Corpo Caloso/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Agenesia do Corpo Caloso/embriologia , Agenesia do Corpo Caloso/genética , Agenesia do Corpo Caloso/metabolismo , Animais , Padronização Corporal/genética , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/metabolismo , Corpo Caloso/enzimologia , Corpo Caloso/patologia , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Encefalocele/genética , Encefalocele/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Fatores de Transcrição Kruppel-Like/genética , Camundongos , Camundongos Knockout , Mutação , Neocórtex/embriologia , Neocórtex/metabolismo , Neocórtex/patologia , Proteínas do Tecido Nervoso/genética , Neurônios/metabolismo , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Fatores de Transcrição de Fator Regulador X , Retinose Pigmentar , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Proteína Gli3 com Dedos de Zinco
8.
J Neurosurg Pediatr ; 12(4): 380-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23971635

RESUMO

OBJECT: The authors sought to identify novel biomarkers for early detection of neural tube defects (NTDs) in human fetuses. METHODS: Amniotic fluid and serum were drawn from women in the second trimester of pregnancy. The study group included 2 women pregnant with normal fetuses and 4 with fetuses displaying myelomeningocele (n = 1), anencephaly (n = 1), holoprosencephaly (n = 1), or encephalocele (n = 1). Amniotic fluid stem cells (AFSCs) were isolated and cultured. The cells were immunostained for the stem cell markers Oct4, CD133, and Sox2; the epigenetic biomarkers H3K4me2, H3K4me3, H3K27me2, H3K27me3, H3K9Ac, and H3K18Ac; and the histone modifiers KDM6B (a histone H3K27 demethylase) and Gcn5 (a histone acetyltransferase). The levels of 2 markers for neural tube development, bone morphogenetic protein-4 (BMP4) and sonic hedgehog (Shh), were measured in amniotic fluid and serum using an enzyme-linked immunosorbent assay. RESULTS: The AFSCs from the woman pregnant with a fetus affected by myelomeningocele had higher levels of H3K4me2, H3K4me3, H3K27me2, and H3K27me3 and lower levels of KDM6B than the AFSCs from the women with healthy fetuses. The levels of H3K9ac, H3K18ac, and Gcn5 were also decreased in the woman with the fetus exhibiting myelomeningocele. In AFSCs from the woman carrying an anencephalic fetus, levels of H3K27me3, along with those of H3K9Ac, H3K18ac, and Gcn5, were increased, while that of KDM6B was decreased. Compared with the normal controls, the levels of BMP4 in amniotic fluid and serum from the woman with a fetus with myelomeningocele were increased, whereas levels of Shh were increased in the woman pregnant with a fetus displaying anencephaly. CONCLUSIONS: The levels of epigenetic marks, such as H3K4me, H3K27me3, H3K9Ac, and H3K18A, in cultured AFSCs in combination with levels of key developmental proteins, such as BMP4 and Shh, are potential biomarkers for early detection and identification of NTDs in amniotic fluid and maternal serum.


Assuntos
Líquido Amniótico/metabolismo , Biomarcadores/metabolismo , Defeitos do Tubo Neural/diagnóstico , Defeitos do Tubo Neural/metabolismo , Adulto , Líquido Amniótico/citologia , Anencefalia/diagnóstico , Anencefalia/metabolismo , Biomarcadores/sangue , Proteína Morfogenética Óssea 4/metabolismo , Encefalocele/diagnóstico , Encefalocele/metabolismo , Ensaio de Imunoadsorção Enzimática , Epigênese Genética , Feminino , Proteínas Hedgehog/metabolismo , Humanos , Histona Desmetilases com o Domínio Jumonji/metabolismo , Meningomielocele/diagnóstico , Meningomielocele/metabolismo , Defeitos do Tubo Neural/sangue , Defeitos do Tubo Neural/genética , Gravidez , Segundo Trimestre da Gravidez , Células-Tronco/metabolismo
9.
Dev Biol ; 377(1): 55-66, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23454480

RESUMO

Meckel-Gruber syndrome (MKS) is an embryonic lethal ciliopathy resulting from mutations in genes encoding proteins localising to the primary cilium. Mutations in the basal body protein MKS1 account for 7% of cases of MKS. The condition affects the development of multiple organs, including brain, kidney and skeleton. Here we present a novel Mks1(tm1a(EUCOMM)Wtsi) knockout mouse which accurately recapitulates the human condition, consistently developing pre-axial polydactyly, complex posterior fossa defects (including the Dandy-Walker malformation), and renal cystic dysplasia. TOPFlash Wnt reporter assays in mouse embryonic fibroblasts (MEFs) showed general de-regulated high levels of canonical Wnt/ß-catenin signalling in Mks1(-/-) cells. In addition to these signalling defects, we also observed ectopic high proliferation in the brain and kidney of mutant animals at mid- to late-gestation. The specific role of Mks1 in regulating cell proliferation was confirmed in Mks1 siRNA knockdown experiments which showed increased levels of proliferation after knockdown, an effect not seen after knockdown of other ciliopathy genes. We suggest that this is a result of the de-regulation of multiple signalling pathways (Wnt, mTOR and Hh) in the absence of functional Mks1. This novel model system offers insights into the role of MKS1 in Wnt signalling and proliferation, and the impact of deregulation of these processes on brain and kidney development in MKS, as well as expanding our understanding of the role of Mks1 in multiple signalling pathways.


Assuntos
Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/patologia , Modelos Animais de Doenças , Encefalocele/metabolismo , Encefalocele/patologia , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Via de Sinalização Wnt , Animais , Western Blotting , Encéfalo/embriologia , Encéfalo/metabolismo , Encéfalo/patologia , Proliferação de Células , Transtornos da Motilidade Ciliar/genética , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Encefalocele/genética , Éxons/genética , Fibroblastos/metabolismo , Fibroblastos/patologia , Imunofluorescência , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Hidrocefalia/embriologia , Hidrocefalia/patologia , Camundongos , Microftalmia/embriologia , Microftalmia/patologia , Doenças Renais Policísticas/genética , Proteínas/genética , Proteínas/metabolismo , Retinose Pigmentar
10.
Am J Hum Genet ; 92(3): 468-74, 2013 Mar 07.
Artigo em Inglês | MEDLINE | ID: mdl-23472759

RESUMO

Cobblestone brain malformation (COB) is a neuronal migration disorder characterized by protrusions of neurons beyond the first cortical layer at the pial surface of the brain. It is usually seen in association with dystroglycanopathy types of congenital muscular dystrophies (CMDs) and ocular abnormalities termed muscle-eye-brain disease. Here we report homozygous deleterious mutations in LAMB1, encoding laminin subunit beta-1, in two families with autosomal-recessive COB. Affected individuals displayed a constellation of brain malformations including cortical gyral and white-matter signal abnormalities, severe cerebellar dysplasia, brainstem hypoplasia, and occipital encephalocele, but they had less apparent ocular or muscular abnormalities than are typically observed in COB. LAMB1 is localized to the pial basement membrane, suggesting that defective connection between radial glial cells and the pial surface mediated by LAMB1 leads to this malformation.


Assuntos
Encéfalo/anormalidades , Laminina/genética , Distrofias Musculares/genética , Malformações do Sistema Nervoso/genética , Deleção de Sequência , Síndrome de Walker-Warburg/genética , Membrana Basal/metabolismo , Membrana Basal/patologia , Encéfalo/metabolismo , Encéfalo/patologia , Cerebelo/metabolismo , Cerebelo/patologia , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Encefalocele/genética , Encefalocele/metabolismo , Encefalocele/patologia , Feminino , Predisposição Genética para Doença , Homozigoto , Humanos , Masculino , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Malformações do Sistema Nervoso/metabolismo , Malformações do Sistema Nervoso/patologia , Neuroglia/metabolismo , Neuroglia/patologia , Neurônios/metabolismo , Neurônios/patologia , Síndrome de Walker-Warburg/metabolismo , Síndrome de Walker-Warburg/patologia
11.
PLoS One ; 8(3): e59306, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23516626

RESUMO

Ciliopathies lead to multiorgan pathologies that include renal cysts, deafness, obesity and retinal degeneration. Retinal photoreceptors have connecting cilia joining the inner and outer segment that are responsible for transport of molecules to develop and maintain the outer segment process. The present study evaluated meckelin (MKS3) expression during outer segment genesis and determined the consequences of mutant meckelin on photoreceptor development and survival in Wistar polycystic kidney disease Wpk/Wpk rat using immunohistochemistry, analysis of cell death and electron microscopy. MKS3 was ubiquitously expressed throughout the retina at postnatal day 10 (P10) and P21. However, in the mature retina, MKS3 expression was restricted to photoreceptors and the retinal ganglion cell layer. At P10, both the wild type and homozygous Wpk mutant retina had all retinal cell types. In contrast, by P21, cells expressing rod- and cone-specific markers were fewer in number and expression of opsins appeared to be abnormally localized to the cell body. Cell death analyses were consistent with the disappearance of photoreceptor-specific markers and showed that the cells were undergoing caspase-dependent cell death. By electron microscopy, P10 photoreceptors showed rudimentary outer segments with an axoneme, but did not develop outer segment discs that were clearly present in the wild type counterpart. At p21 the mutant outer segments appeared much the same as the P10 mutant outer segments with only a short axoneme, while the wild-type controls had developed outer segments with many well-organized discs. We conclude that MKS3 is not important for formation of connecting cilium and rudimentary outer segments, but is critical for the maturation of outer segment processes.


Assuntos
Transtornos da Motilidade Ciliar/metabolismo , Encefalocele/metabolismo , Proteínas de Membrana/metabolismo , Doenças Renais Policísticas/metabolismo , Retina/metabolismo , Segmento Externo das Células Fotorreceptoras da Retina/metabolismo , Animais , Proteínas de Transporte/metabolismo , Cílios/metabolismo , Cílios/ultraestrutura , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Proteínas de Membrana/genética , Microscopia Eletrônica de Transmissão , Ratos , Ratos Wistar , Retina/ultraestrutura
12.
Hum Mol Genet ; 22(7): 1358-72, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23283079

RESUMO

The ciliopathies are a group of heterogeneous diseases with considerable variations in phenotype for allelic conditions such as Meckel-Gruber syndrome (MKS) and Joubert syndrome (JBTS) even at the inter-individual level within families. In humans, mutations in TMEM67 (also known as MKS3) cause both MKS and JBTS, with TMEM67 encoding the orphan receptor meckelin (TMEM67) that localizes to the ciliary transition zone. We now describe the Tmem67(tm1(Dgen/H)) knockout mouse model that recapitulates the brain phenotypic variability of these human ciliopathies, with categorization of Tmem67 mutant animals into two phenotypic groups. An MKS-like incipient congenic group (F6 to F10) manifested very variable neurological features (including exencephaly, and frontal/occipital encephalocele) that were associated with the loss of primary cilia, diminished Shh signalling and dorsalization of the caudal neural tube. The 'MKS-like' group also had high de-regulated canonical Wnt/ß-catenin signalling associated with hyper-activated Dishevelled-1 (Dvl-1) localized to the basal body. Conversely, a second fully congenic group (F > 10) had less variable features pathognomonic for JBTS (including cerebellar hypoplasia), and retention of abnormal bulbous cilia associated with mild neural tube ventralization. The 'JBTS-like' group had de-regulated low levels of canonical Wnt signalling associated with the loss of Dvl-1 localization to the basal body. Our results suggest that modifier alleles partially determine the variation between MKS and JBTS, implicating the interaction between Dvl-1 and meckelin, or other components of the ciliary transition zone. The Tmem67(tm1(Dgen/H)) line is unique in modelling the variable expressivity of phenotypes in these two ciliopathies.


Assuntos
Doenças Cerebelares/metabolismo , Transtornos da Motilidade Ciliar/metabolismo , Encefalocele/metabolismo , Anormalidades do Olho/metabolismo , Proteínas Hedgehog/metabolismo , Doenças Renais Císticas/metabolismo , Proteínas de Membrana/genética , Doenças Renais Policísticas/metabolismo , Via de Sinalização Wnt , Anormalidades Múltiplas , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Padronização Corporal/genética , Doenças Cerebelares/genética , Doenças Cerebelares/patologia , Cerebelo/anormalidades , Cílios/patologia , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/patologia , Modelos Animais de Doenças , Proteínas Desgrenhadas , Encefalocele/genética , Encefalocele/patologia , Anormalidades do Olho/genética , Anormalidades do Olho/patologia , Regulação da Expressão Gênica , Genes Reporter , Células HEK293 , Humanos , Doenças Renais Císticas/genética , Doenças Renais Císticas/patologia , Luciferases de Vaga-Lume/biossíntese , Luciferases de Vaga-Lume/genética , Proteínas de Membrana/deficiência , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Defeitos do Tubo Neural/patologia , Fenótipo , Fosfoproteínas/metabolismo , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/patologia , Transporte Proteico , Retina/anormalidades , Retina/metabolismo , Retina/patologia , Retinose Pigmentar
13.
Proc Natl Acad Sci U S A ; 109(42): 16951-6, 2012 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-23027964

RESUMO

Joubert syndrome (JS) and Meckel syndrome (MKS) are pleiotropic ciliopathies characterized by severe defects of the cerebellar vermis, ranging from hypoplasia to aplasia. Interestingly, ciliary conditional mutant mice have a hypoplastic cerebellum in which the proliferation of cerebellar granule cell progenitors (GCPs) in response to Sonic hedgehog (SHH) is severely reduced. This suggests that Shh signaling defects could contribute to the vermis hypoplasia observed in the human syndromes. As existing JS/MKS mutant mouse models suggest apparently contradictory hypotheses on JS/MKS etiology, we investigated Shh signaling directly on human fetal samples. First, in an examination of human cerebellar development, we linked the rates of GCP proliferation to the different levels and localizations of active Shh signaling and showed that the GCP possessed a primary cilium with CEP290 at its base. Second, we found that the proliferation of GCPs and their response to SHH were severely impaired in the cerebellum of subjects with JS/MKS and Jeune syndrome. Finally, we showed that the defect in GCP proliferation was similar in the cerebellar vermis and hemispheres in all patients with ciliopathy analyzed, suggesting that the specific cause of vermal hypo-/aplasia precedes this defect. Our results, obtained from the analysis of human samples, show that the hemispheres and the vermis are affected in JS/MKS and provide evidence of a defective cellular mechanism in these pathologic processes.


Assuntos
Doenças Cerebelares/metabolismo , Cerebelo/embriologia , Cerebelo/metabolismo , Transtornos da Motilidade Ciliar/metabolismo , Encefalocele/metabolismo , Anormalidades do Olho/metabolismo , Células Precursoras de Granulócitos/fisiologia , Proteínas Hedgehog/metabolismo , Doenças Renais Císticas/metabolismo , Doenças Renais Policísticas/metabolismo , Transdução de Sinais/fisiologia , Anormalidades Múltiplas , Animais , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Proteínas de Ciclo Celular , Proliferação de Células , Doenças Cerebelares/patologia , Cerebelo/patologia , Transtornos da Motilidade Ciliar/patologia , Proteínas do Citoesqueleto , Encefalocele/patologia , Anormalidades do Olho/patologia , Humanos , Imuno-Histoquímica , Hibridização In Situ , Doenças Renais Císticas/patologia , Camundongos , Microscopia Eletrônica de Transmissão , Microscopia de Fluorescência , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Doenças Renais Policísticas/patologia , Interferência de RNA , Retina/anormalidades , Retina/metabolismo , Retina/patologia , Retinose Pigmentar , Estatísticas não Paramétricas
14.
Endocr J ; 58(3): 193-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21325746

RESUMO

We report a 21-year-old man with severe fatigue due to hypopituitarism. At the age of 6 years, he was diagnosed with short stature due to a GH deficiency accompanied by a sphenoid cystic lesion. Laboratory findings and provocative tests for pituitary hormone function revealed ACTH, LH, FSH, TSH, and GH deficiency. Computed tomography and magnetic resonance imaging revealed transsphenoidal cephalocele due to a defect in the floor of the sella turcica. At 6 years, he only had severe GH deficiency and poor response of LH to LHRH. Hypothalamic-pituitary dysfunction and pituitary herniation have progressed subsequently; we observed a longitudinal progression of hypothalamic-pituitary dysfunction caused by transsphenoidal cephalocele. This dysfunction requires the selection of a treatment that will not aggravate the condition further.


Assuntos
Encefalocele/epidemiologia , Encefalocele/fisiopatologia , Hipopituitarismo/epidemiologia , Hipopituitarismo/fisiopatologia , Sistema Hipotálamo-Hipofisário/fisiopatologia , Hormônios Hipofisários/deficiência , Hormônio Adrenocorticotrópico/deficiência , Comorbidade , Encefalocele/metabolismo , Hormônio Foliculoestimulante/deficiência , Hormônio do Crescimento/deficiência , Humanos , Hipopituitarismo/metabolismo , Estudos Longitudinais , Hormônio Luteinizante/deficiência , Masculino , Tireotropina/deficiência , Adulto Jovem
15.
Nephron Exp Nephrol ; 117(2): e31-8, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-20693816

RESUMO

BACKGROUND: Meckel syndrome (MKS) is a fatal autosomal recessive condition with prominent renal cystic pathology. Renal protein misexpression was evaluated in the Wpk rat model of human MKS3 gene disease to identify biomarkers for the staging of renal cystic progression. METHODS: Misexpressed proteins were compared between early and late stages of MKS renal cystic disease using proteomic analysis (two-dimensional gel electrophoresis with LC-MS/MS identification) followed by Western blot analysis. RESULTS: A proteomic analysis identified 76 proteins with statistically different, normalized abundance in at least one group. Subsequently, Western blot was used to confirm differential expression in several of these and polycystic kidney disease (PKD)-associated proteins. Galectin-1 and vimentin were identified as overexpressed proteins, which have been previously found in the jck mouse model of nephronophthisis 9. Ciliopathic PKD proteins, polycystins 1 & 2, and fibrocystin were also differentially expressed in Wpk kidney. CONCLUSION: In the Wpk rat, misexpressed proteins were identified that were also implicated in other forms of cystic disease. Numerous proteins were either over- or underexpressed in late-stage disease. Differences in protein expression may serve as biomarkers of cystic disease and its progression.


Assuntos
Rim/metabolismo , Proteoma/metabolismo , Animais , Western Blotting , Transtornos da Motilidade Ciliar/genética , Transtornos da Motilidade Ciliar/metabolismo , Modelos Animais de Doenças , Eletroforese em Gel Bidimensional , Encefalocele/genética , Encefalocele/metabolismo , Proteína 3 Ligante de Ácido Graxo , Proteínas de Ligação a Ácido Graxo/metabolismo , Feminino , Galectinas/metabolismo , Humanos , Laminina/metabolismo , Masculino , Camundongos , Doenças Renais Policísticas/genética , Doenças Renais Policísticas/metabolismo , Proteômica , Ratos , Ratos Mutantes , Receptores de Superfície Celular/metabolismo , Canais de Cátion TRPP/metabolismo , Espectrometria de Massas em Tandem , Vimentina/metabolismo
16.
Dis Model Mech ; 4(1): 43-56, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21045211

RESUMO

Meckel-Gruber syndrome (MKS) is a recessive disorder resulting in multiple birth defects that are associated with mutations affecting ciliogenesis. We recovered a mouse mutant with a mutation in the Mks1 gene (Mks1(del64-323)) that caused a 260-amino-acid deletion spanning nine amino acids in the B9 domain, a protein motif with unknown function conserved in two other basal body proteins. We showed that, in wild-type cells, Mks1 was localized to the mother centriole from which the cilium was generated. However, in mutant Mks1(del64-323) cells, Mks1 was not localized to the centriole, even though it maintained a punctate distribution. Resembling MKS patients, Mks1 mutants had craniofacial defects, polydactyly, congenital heart defects, polycystic kidneys and randomized left-right patterning. These defects reflected disturbance of functions subserved by motile and non-motile cilia. In the kidney, glomerular and tubule cysts were observed along with short cilia, and cilia were reduced in number to a near-complete loss. Underlying the left-right patterning defects were fewer and shorter nodal cilia, and analysis with fluorescent beads showed no directional flow at the embryonic node. In the cochlea, the stereocilia were mal-patterned, with the kinocilia being abnormally positioned. Together, these defects suggested disruption of planar cell polarity, which is known to regulate node, kidney and cochlea development. In addition, we also showed that Shh signaling was disrupted. Thus, in the neural tube, the floor plate was not specified posteriorly even as expression of the Shh mediator Gli2 increased. By contrast, the Shh signaling domain was expanded in the anterior neural tube and anterior limb bud, consistent with reduced Gli3-repressor (Gli3R) function. The latter probably accounted for the preaxial digit duplication exhibited by the Mks1(del64-323) mutants. Overall, these findings indicate that centriole localization of Mks1 is required for ciliogenesis of motile and non-motile cilia, but not for centriole assembly. On the basis of these results, we hypothesize a role for the B9 domain in mother centriole targeting, a possibility that warrants further future investigations.


Assuntos
Anormalidades Múltiplas/patologia , Centríolos/metabolismo , Cílios/patologia , Proteínas/metabolismo , Anormalidades Múltiplas/metabolismo , Sequência de Aminoácidos , Animais , Padronização Corporal , Centríolos/patologia , Cílios/metabolismo , Cílios/ultraestrutura , Transtornos da Motilidade Ciliar/metabolismo , Transtornos da Motilidade Ciliar/patologia , Embrião de Mamíferos/anormalidades , Embrião de Mamíferos/patologia , Encefalocele/metabolismo , Encefalocele/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Fibroblastos/metabolismo , Fibroblastos/patologia , Células Ciliadas Auditivas/patologia , Células Ciliadas Auditivas/ultraestrutura , Proteínas Hedgehog/metabolismo , Doenças Renais Císticas/complicações , Doenças Renais Císticas/patologia , Camundongos , Dados de Sequência Molecular , Mutação/genética , Tubo Neural/anormalidades , Tubo Neural/embriologia , Tubo Neural/patologia , Tubo Neural/ultraestrutura , Especificidade de Órgãos , Doenças Renais Policísticas/metabolismo , Doenças Renais Policísticas/patologia , Transporte Proteico , Proteínas/química , Retinose Pigmentar , Transdução de Sinais
18.
Birth Defects Res A Clin Mol Teratol ; 88(8): 619-25, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20589882

RESUMO

BACKGROUND: Over 200 mouse genes are associated with neural tube defects (NTDs), including Cecr2, the bromodomain-containing subunit of the CERF chromatin remodeling complex. METHODS: Gene-trap mutation Cecr2(Gt45Bic) results in 74% exencephaly (equivalent of human anencephaly) on the BALB/c strain. Gene expression altered during cranial neural tube closure by the Cecr2 mutation was identified through microarray analysis of 11-14 somites stage Cecr2(Gt45Bic)embryos. RESULTS: Analysis of Affymetrix Mouse 430 2.0 chips detected 60 transcripts up-regulated and 54 transcripts down-regulated in the Cecr2(Gt45Bic) embryos (fold > 1.5, p < 0.05). The Cecr2 transcript was reduced only approximately 7- to 14-fold from normal levels, suggesting the Cecr2(Gt45Bic) is a hypomorphic mutation. We therefore generated a novel Cecr2 null allele (Cecr2 (tm1.1Hemc)). Resulting mutants displayed a stronger penetrance of exencephaly than Cecr2(Gt45Bic) in both BALB/c and FVB/N strains, in addition to midline facial clefts and forebrain encephalocele in the FVB/N strain. The Cecr2 transcript is reduced 260-fold in the Cecr2(tm1.1Hemc) line. Subsequent qRT-PCR using Cecr2 (tm1.1Hemc) mutant heads confirmed downregulation of transcription factors Alx1/Cart1, Dlx5, Eya1, and Six1. CONCLUSIONS: As both Alx1/Cart1 and Dlx5 mouse mutations result in exencephaly, we hypothesize that changes in expression of these mesenchymal/ectodermal transcription factors may contribute to NTDs associated with Cecr2.


Assuntos
Ectoderma/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Mesoderma/metabolismo , Mutação , Defeitos do Tubo Neural/genética , Defeitos do Tubo Neural/metabolismo , Fatores de Transcrição/genética , Animais , Regulação para Baixo/genética , Ectoderma/fisiopatologia , Encefalocele/metabolismo , Ossos Faciais/anormalidades , Feminino , Regulação da Expressão Gênica no Desenvolvimento/genética , Mesoderma/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Defeitos do Tubo Neural/fisiopatologia , Gravidez , Prosencéfalo/anormalidades , Fatores de Transcrição/metabolismo , Transcrição Gênica , Regulação para Cima/genética
19.
Pediatr Dev Pathol ; 4(3): 289-97, 2001.
Artigo em Inglês | MEDLINE | ID: mdl-11370267

RESUMO

We present the first report of extramedullary hematopoiesis (EMH) in an encephalocele. The patient was a new-born with semilobar holoprosencephaly, a frontoethmoidal encephalocele, and a large subdural hematoma. The encephalocele appeared as a hemorrhagic mass, protruding from the forehead to cover the right eye, without involvement of the sinuses or nasopharynx. Computerized tomography and magnetic resonance imaging studies ruled out other forms of holoprosencephaly and confirmed the continuity of the brain with the extruded mass. Immunohistochemistry confirmed the presence of an atrophic epithelium covering the mass. Histologic examination of the encephalocele revealed EMH both within and adjacent to malformed cerebral cortex, with a tendency for the hematopoietic cells to line up in columns within malformed cerebral cortex. We propose that a single event during the fourth week of gestation could both interrupt closure of the neural tube, giving rise to the encephalocele, and impair migration of the neural crest, leading to holoprosencephaly secondary to failure of neural crest derivatives to induce basomedial telencephalic differentiation. EMH may have been induced from hematopoietic stem cells in the richly vascular meningeal component of the encephalocele, in response to anemia and hypoxia.


Assuntos
Encefalocele/patologia , Hematopoese Extramedular , Holoprosencefalia/patologia , Biomarcadores/análise , Encéfalo/anormalidades , Encéfalo/metabolismo , Encefalocele/complicações , Encefalocele/metabolismo , Encefalocele/cirurgia , Osso Etmoide/anormalidades , Osso Frontal/anormalidades , Holoprosencefalia/complicações , Holoprosencefalia/metabolismo , Holoprosencefalia/cirurgia , Humanos , Técnicas Imunoenzimáticas , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Tomografia Computadorizada por Raios X , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA