Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Proc Natl Acad Sci U S A ; 119(31): e2116974119, 2022 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-35881792

RESUMO

Ribosomal RNA (rRNA) transcription by RNA polymerase I (Pol I) is a critical rate-limiting step in ribosome biogenesis, which is essential for cell survival. Despite its global function, disruptions in ribosome biogenesis cause tissue-specific birth defects called ribosomopathies, which frequently affect craniofacial development. Here, we describe a cellular and molecular mechanism underlying the susceptibility of craniofacial development to disruptions in Pol I transcription. We show that Pol I subunits are highly expressed in the neuroepithelium and neural crest cells (NCCs), which generate most of the craniofacial skeleton. High expression of Pol I subunits sustains elevated rRNA transcription in NCC progenitors, which supports their high tissue-specific levels of protein translation, but also makes NCCs particularly sensitive to rRNA synthesis defects. Consistent with this model, NCC-specific deletion of Pol I subunits Polr1a, Polr1c, and associated factor Tcof1 in mice cell-autonomously diminishes rRNA synthesis, which leads to p53 protein accumulation, resulting in NCC apoptosis and craniofacial anomalies. Furthermore, compound mutations in Pol I subunits and associated factors specifically exacerbate the craniofacial anomalies characteristic of the ribosomopathies Treacher Collins syndrome and Acrofacial Dysostosis-Cincinnati type. Mechanistically, we demonstrate that diminished rRNA synthesis causes an imbalance between rRNA and ribosomal proteins. This leads to increased binding of ribosomal proteins Rpl5 and Rpl11 to Mdm2 and concomitantly diminished binding between Mdm2 and p53. Altogether, our results demonstrate a dynamic spatiotemporal requirement for rRNA transcription during mammalian cranial NCC development and corresponding tissue-specific threshold sensitivities to disruptions in rRNA transcription in the pathogenesis of congenital craniofacial disorders.


Assuntos
Anormalidades Craniofaciais , RNA Polimerase I , RNA Ribossômico , Proteínas Ribossômicas , Crânio , Transcrição Gênica , Animais , Anormalidades Craniofaciais/genética , Disostose Mandibulofacial/genética , Camundongos , Crista Neural/embriologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , RNA Polimerase I/metabolismo , RNA Ribossômico/genética , Proteínas Ribossômicas/metabolismo , Crânio/embriologia , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
2.
Front Med (Lausanne) ; 8: 636160, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33777979

RESUMO

Healthcare workers (HCWs) are known to be at increased risk of infection with SARS-CoV-2, although whether these risks are equal across all roles is uncertain. Here we report a retrospective analysis of a large real-world dataset obtained from 10 March to 6 July 2020 in an NHS Foundation Trust in England with 17,126 employees. 3,338 HCWs underwent symptomatic PCR testing (14.4% positive, 2.8% of all staff) and 11,103 HCWs underwent serological testing for SARS-CoV-2 IgG (8.4% positive, 5.5% of all staff). Seropositivity was lower than other hospital settings in England but higher than community estimates. Increased test positivity rates were observed in HCWs from BAME backgrounds and residents in areas of higher social deprivation. A multiple logistic regression model adjusting for ethnicity and social deprivation confirmed statistically significant increases in the odds of testing positive in certain occupational groups, most notably domestic services staff, nurses, and health-care assistants. PCR testing of symptomatic HCWs appeared to underestimate overall infection levels, probably due to asymptomatic seroconversion. Clinical outcomes were reassuring, with only a small minority of HCWs with COVID-19 requiring hospitalization (2.3%) or ICU management (0.7%) and with no deaths. Despite a relatively low level of HCW infection compared to other UK cohorts, there were nevertheless important differences in test positivity rates between occupational groups, robust to adjustment for demographic factors such as ethnic background and social deprivation. Quantitative and qualitative studies are needed to better understand the factors contributing to this risk. Robust informatics solutions for HCW exposure data are essential to inform occupational monitoring.

3.
Semin Cell Dev Biol ; 91: 75-83, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-28803895

RESUMO

Development of the secondary palate involves a complex series of embryonic events which, if disrupted, result in the common congenital anomaly cleft palate. The secondary palate forms from paired palatal shelves which grow initially vertically before elevating to a horizontal position above the tongue and fusing together in the midline via the medial edge epithelia. As the epithelia of the vertical palatal shelves are in contact with the mandibular and lingual epithelia, pathological fusions between the palate and the mandible and/or the tongue must be prevented. This function is mediated by the single cell layered periderm which forms in a distinct and reproducible pattern early in embryogenesis, exhibits highly polarised expression of adhesion complexes, and is shed from the outer surface as the epidermis acquires its barrier function. Disruption of periderm formation and/or function underlies a series of birth defects that exhibit multiple inter-epithelial adhesions including the autosomal dominant popliteal pterygium syndrome and the autosomal recessive cocoon syndrome and Bartsocas Papas syndrome. Genetic analyses of these conditions have shown that IRF6, IKKA, SFN, RIPK4 and GRHL3, all of which are under the transcriptional control of p63, play a key role in periderm formation. Despite these observations, the medial edge epithelia must rapidly acquire the capability to fuse if the palatal shelves are not to remain cleft. This process is driven by TGFß3-mediated, down-regulation of p63 in the medial edge epithelia which allows periderm migration out of the midline epithelial seam and reduces the proliferative potential of the midline epithelial seam thereby preventing cleft palate. Together, these findings indicate that periderm plays a transient but fundamental role during embryogenesis in preventing pathological adhesion between intimately apposed, adhesion-competent epithelia.


Assuntos
Fissura Palatina/embriologia , Epiderme/embriologia , Epitélio/embriologia , Palato/embriologia , Animais , Diferenciação Celular/genética , Fissura Palatina/genética , Epiderme/metabolismo , Epitélio/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Palato/citologia , Palato/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética
4.
PLoS Genet ; 13(6): e1006828, 2017 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-28604778

RESUMO

Cleft palate is a common congenital disorder that affects up to 1 in 2500 live births and results in considerable morbidity to affected individuals and their families. The aetiology of cleft palate is complex with both genetic and environmental factors implicated. Mutations in the transcription factor p63 are one of the major individual causes of cleft palate; however, the gene regulatory networks in which p63 functions remain only partially characterized. Our findings demonstrate that p63 functions as an essential regulatory molecule in the spatio-temporal control of palatal epithelial cell fate to ensure appropriate fusion of the palatal shelves. Initially, p63 induces periderm formation and controls its subsequent maintenance to prevent premature adhesion between adhesion-competent, intra-oral epithelia. Subsequently, TGFß3-induced down-regulation of p63 in the medial edge epithelia of the palatal shelves is a pre-requisite for palatal fusion by facilitating periderm migration from, and reducing the proliferative potential of, the midline epithelial seam thereby preventing cleft palate.


Assuntos
Fissura Palatina/genética , Redes Reguladoras de Genes/genética , Fosfoproteínas/genética , Transativadores/genética , Fator de Crescimento Transformador beta3/genética , Animais , Movimento Celular/genética , Proliferação de Células/genética , Fissura Palatina/fisiopatologia , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Camundongos , Mutação , Fosfoproteínas/biossíntese , Transdução de Sinais/genética , Transativadores/biossíntese
5.
Artigo em Inglês | MEDLINE | ID: mdl-28186364

RESUMO

Craniofacial anomalies account for approximately one-third of all congenital birth defects reflecting the complexity of head and facial development. Craniofacial development is dependent upon a multipotent, migratory population of neural crest cells, which generate most of the bone and cartilage of the head and face. In this review, we discuss advances in our understanding of the pathogenesis of a specific array of craniofacial anomalies, termed facial dysostoses, which can be subdivided into mandibulofacial dysostosis, which present with craniofacial defects only, and acrofacial dysostosis, which encompasses both craniofacial and limb anomalies. In particular, we focus on Treacher Collins syndrome, Acrofacial Dysostosis-Cincinnati Type as well as Nager and Miller syndromes, and animal models that provide new insights into the molecular and cellular basis of these congenital syndromes. We emphasize the etiologic and pathogenetic similarities between these birth defects, specifically their unique deficiencies in global processes including ribosome biogenesis, DNA damage repair, and pre-mRNA splicing, all of which affect neural crest cell development and result in similar tissue-specific defects. WIREs Dev Biol 2017, 6:e263. doi: 10.1002/wdev.263 For further resources related to this article, please visit the WIREs website.


Assuntos
Disostose Mandibulofacial/patologia , Crista Neural/patologia , Animais , Humanos , Síndrome
6.
Genet Med ; 18(11): 1158-1162, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-26963285

RESUMO

PURPOSE: We aimed to identify a novel genetic cause of tooth agenesis (TA) and/or orofacial clefting (OFC) by combining whole-exome sequencing (WES) and targeted resequencing in a large cohort of TA and OFC patients. METHODS: WES was performed in two unrelated patients: one with severe TA and OFC and another with severe TA only. After deleterious mutations were identified in a gene encoding low-density lipoprotein receptor-related protein 6 (LRP6), all its exons were resequenced with molecular inversion probes in 67 patients with TA, 1,072 patients with OFC, and 706 controls. RESULTS: We identified a frameshift (c.4594delG, p.Cys1532fs) and a canonical splice-site mutation (c.3398-2A>C, p.?) in LRP6, respectively, in the patient with TA and OFC and in the patient with severe TA only. The targeted resequencing showed significant enrichment of unique LRP6 variants in TA patients but not in nonsyndromic OFC patients. Of the five variants in patients with TA, two affected the canonical splice site and three were missense variants; all variants segregated with the dominant phenotype, and in one case the missense mutation occurred de novo. CONCLUSION: Mutations in LRP6 cause TA in humans.Genet Med 18 11, 1158-1162.


Assuntos
Anodontia/genética , Exoma/genética , Predisposição Genética para Doença , Proteína-6 Relacionada a Receptor de Lipoproteína de Baixa Densidade/genética , Adolescente , Anodontia/patologia , Criança , Feminino , Mutação da Fase de Leitura/genética , Humanos , Masculino , Mutação de Sentido Incorreto/genética , Linhagem , Análise de Sequência de DNA , Via de Sinalização Wnt/genética
7.
Dev Biol ; 415(2): 296-305, 2016 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-26772999

RESUMO

The palate functions as the roof of the mouth in mammals, separating the oral and nasal cavities. Its complex embryonic development and assembly poses unique susceptibilities to intrinsic and extrinsic disruptions. Such disruptions may cause failure of the developing palatal shelves to fuse along the midline resulting in a cleft. In other cases the palate may fuse at an arch, resulting in a vaulted oral cavity, termed high-arched palate. There are many models available for studying the pathogenesis of cleft palate but a relative paucity for high-arched palate. One condition exhibiting either cleft palate or high-arched palate is Treacher Collins syndrome, a congenital disorder characterized by numerous craniofacial anomalies. We quantitatively analyzed palatal perturbations in the Tcof1(+/-) mouse model of Treacher Collins syndrome, which phenocopies the condition in humans. We discovered that 46% of Tcof1(+/-) mutant embryos and new born pups exhibit either soft clefts or full clefts. In addition, 17% of Tcof1(+/-) mutants were found to exhibit high-arched palate, defined as two sigma above the corresponding wild-type population mean for height and angular based arch measurements. Furthermore, palatal shelf length and shelf width were decreased in all Tcof1(+/-) mutant embryos and pups compared to controls. Interestingly, these phenotypes were subsequently ameliorated through genetic inhibition of p53. The results of our study therefore provide a simple, reproducible and quantitative method for investigating models of high-arched palate.


Assuntos
Desenvolvimento Maxilofacial/fisiologia , Proteínas Nucleares/genética , Palato/anormalidades , Fosfoproteínas/genética , Animais , Fissura Palatina/diagnóstico por imagem , Fissura Palatina/embriologia , Fissura Palatina/genética , Cruzamentos Genéticos , Modelos Animais de Doenças , Feminino , Técnicas de Inativação de Genes , Genes p53 , Heterozigoto , Humanos , Imageamento Tridimensional , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Disostose Mandibulofacial/diagnóstico por imagem , Disostose Mandibulofacial/embriologia , Disostose Mandibulofacial/genética , Desenvolvimento Maxilofacial/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Microscopia Confocal , Proteínas Nucleares/fisiologia , Palato/diagnóstico por imagem , Palato/embriologia , Fenótipo , Fosfoproteínas/fisiologia , Especificidade da Espécie
8.
Nat Commun ; 7: 10328, 2016 Jan 21.
Artigo em Inglês | MEDLINE | ID: mdl-26792133

RESUMO

Craniofacial anomalies account for approximately one-third of all birth defects and are a significant cause of infant mortality. Since the majority of the bones, cartilage and connective tissues that comprise the head and face are derived from a multipotent migratory progenitor cell population called the neural crest, craniofacial disorders are typically attributed to defects in neural crest cell development. Treacher Collins syndrome (TCS) is a disorder of craniofacial development and although TCS arises primarily through autosomal dominant mutations in TCOF1, no clear genotype-phenotype correlation has been documented. Here we show that Tcof1 haploinsufficiency results in oxidative stress-induced DNA damage and neuroepithelial cell death. Consistent with this discovery, maternal treatment with antioxidants minimizes cell death in the neuroepithelium and substantially ameliorates or prevents the pathogenesis of craniofacial anomalies in Tcof1(+/-) mice. Thus maternal antioxidant dietary supplementation may provide an avenue for protection against the pathogenesis of TCS and similar neurocristopathies.


Assuntos
Antioxidantes/administração & dosagem , Suplementos Nutricionais/análise , Disostose Mandibulofacial/prevenção & controle , Animais , Modelos Animais de Doenças , Feminino , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Disostose Mandibulofacial/embriologia , Disostose Mandibulofacial/genética , Disostose Mandibulofacial/metabolismo , Fenômenos Fisiológicos da Nutrição Materna , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Gravidez
9.
Hum Genet ; 135(1): 41-59, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26561393

RESUMO

Orofacial clefts (OFCs) represent a large fraction of human birth defects and are one of the most common phenotypes affected by large copy number variants (CNVs). Due to the limited number of CNV patients in individual centers, CNV analyses of a large number of OFC patients are challenging. The present study analyzed 249 genomic deletions and 226 duplications from a cohort of 312 OFC patients reported in two publicly accessible databases of chromosome imbalance and phenotype in humans, DECIPHER and ECARUCA. Genomic regions deleted or duplicated in multiple patients were identified, and genes in these overlapping CNVs were prioritized based on the number of genes encompassed by the region and gene expression in embryonic mouse palate. Our analyses of these overlapping CNVs identified two genes known to be causative for human OFCs, SATB2 and MEIS2, and 12 genes (DGCR6, FGF2, FRZB, LETM1, MAPK3, SPRY1, THBS1, TSHZ1, TTC28, TULP4, WHSC1, WHSC2) that are associated with OFC or orofacial development. Additionally, we report 34 deleted and 24 duplicated genes that have not previously been associated with OFCs but are associated with the BMP, MAPK and RAC1 pathways. Statistical analyses show that the high number of overlapping CNVs is not due to random occurrence. The identified genes are not located in highly variable genomic regions in healthy populations and are significantly enriched for genes that are involved in orofacial development. In summary, we report a CNV analysis pipeline of a large cohort of OFC patients and identify novel candidate OFC genes.


Assuntos
Fenda Labial/genética , Fissura Palatina/genética , Variações do Número de Cópias de DNA , Face/anormalidades , Predisposição Genética para Doença , Humanos , Fenótipo
10.
Exp Dermatol ; 24(2): 114-9, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25387952

RESUMO

Nectins are immunoglobulin-like cell adhesion molecules mainly localized in adherens junctions. The transcription factor p63 is a master regulator of gene expression in stratified epithelia and controls several molecular processes. As mutations in the Pvrl1 and Pvrl4 genes encoding for nectins cause genetic disorders with phenotypes similar to p63-related syndromes, we investigated whether these proteins might be under p63 transcriptional control. Here, we show that in p63-null skin, Pvrl1 gene expression is strongly reduced, whereas Pvrl4 expression is unaffected. In human and mouse primary keratinocytes p63 depletion leads to a specific downregulation of the Pvrl1 gene. Consistent with a direct regulation, chromatin immunoprecipitation experiments (ChIP) indicate that p63 binds to two conserved intronic Pvrl1 enhancer regions. Ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome is a rare autosomal dominant disorder, caused by mutations in p63 gene, mainly characterized by skin fragility. To test whether nectins may be affected in AEC syndrome, their expression was measured in keratinocytes obtained from patients with AEC or from a conditional mouse model for AEC syndrome. Pvrl1 expression was reduced in AEC keratinocytes, consistent with impaired p63 function. Surprisingly, Pvrl4 expression was similarly affected, in parallel with decreased expression of the transcription factor Irf6. Consistent with the well-characterized role of Irf6 in keratinocyte differentiation and its strong downregulation in AEC syndrome, Irf6 depletion caused reduced expression of Pvrl4 in wild-type keratinocytes. Taken together, our results indicate that Pvrl1 is a bona fide target gene of the transcription factor p63, whereas Pvrl4 regulation is linked to epidermal differentiation and is under Irf6 control.


Assuntos
Moléculas de Adesão Celular/metabolismo , Epiderme/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Animais Recém-Nascidos , Células Cultivadas , Epiderme/embriologia , Regulação da Expressão Gênica , Regulação da Expressão Gênica no Desenvolvimento , Genes Reporter , Humanos , Queratinócitos/citologia , Camundongos , Mutação , Nectinas , Fenótipo , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Transcrição Gênica
11.
Infect Drug Resist ; 7: 145-52, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24936131

RESUMO

Antimicrobials are an extremely valuable resource across the spectrum of modern medicine. Their development has been associated with dramatic reductions in communicable disease mortality and has facilitated technological advances in cancer therapy, transplantation, and surgery. However, this resource is threatened by the dwindling supply of new antimicrobials and the global increase in antimicrobial resistance. There is an urgent need for antimicrobial stewardship (AMS) to protect our remaining antimicrobials for future generations. AMS emphasizes sensible, appropriate antimicrobial management for the benefit of the individual and society as a whole. Within the English National Health Service (NHS), a series of recent policy initiatives have focused on all aspects of AMS, including best practice guidelines for antimicrobial prescribing, enhanced surveillance mechanisms for monitoring antimicrobial use across primary and secondary care, and new prescribing competencies for doctors in training. Here we provide a concise summary to clarify the current position and importance of AMS within the NHS and review the evidence base for AMS recommendations. The evidence supports the impact of AMS strategies on modifying prescribing practice in hospitals, with beneficial effects on both antimicrobial resistance and the incidence of Clostridium difficile, and no evidence of increased sepsis-related mortality. There is also a promising role for novel diagnostic technologies in AMS, both in enhancing microbiological diagnosis and improving the specificity of sepsis diagnosis. More work is needed to establish an evidence base for interventions to improve public and patient education regarding the role of antibiotics in common clinical syndromes, such as respiratory tract infection. Future priorities include establishing novel approaches to antimicrobial management (eg, duration of therapy, combination regimens) to protect against resistance and working with the pharmaceutical industry to promote the development of new antimicrobials.

12.
PLoS One ; 8(11): e81167, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24260555

RESUMO

The mammalian ureter contains a water-tight epithelium surrounded by smooth muscle. Key molecules have been defined which regulate ureteric bud initiation and drive the differentiation of ureteric mesenchyme into peristaltic smooth muscle. Less is known about mechanisms underlying the developmental patterning of the multilayered epithelium characterising the mature ureter. In skin, which also contains a multilayered epithelium, cytokeratin 15 (CK15), an acidic intermediate filament protein, marks cells whose progeny contribute to epidermal regeneration following wounding. Moreover, CK15+ precursor cells in skin can give rise to basal cell carcinomas. In the current study, using transcriptome microarrays of embryonic wild type mouse ureters, Krt15, coding for CK15, was detected. Quantitative polymerase chain reaction analyses confirmed the initial finding and demonstrated that Krt15 levels increased during the fetal period when the ureteric epithelium becomes multilayered. CK15 protein was undetectable in the ureteric bud, the rudiment from which the ureter grows. Nevertheless, later in fetal development, CK15 was immunodetected in a subset of basal urothelial cells in the ureteric stalk. Superficial epithelial cells, including those positive for the differentiation marker uroplakin III, were CK15-. Transformation-related protein 63 (P63) has been implicated in epithelial differentiation in murine fetal urinary bladders. In wild type fetal ureters, CK15+ cells were positive for P63, and p63 homozygous null mutant ureters lacked CK15+ cells. In these mutant ureters, sections of the urothelium were monolayered versus the uniform multilayering found in wild type littermates. Human urothelial cell carcinomas account for considerable morbidity and mortality. CK15 was upregulated in a subset of invasive ureteric and urinary bladder cancers. Thus, in ureter development, the absence of CK15 is associated with a structurally simplified urothelium whereas, postnatally, increased CK15 levels feature in malignant urothelial overgrowth. CK15 may be a novel marker for urinary tract epithelial precursor cells.


Assuntos
Carcinoma Basocelular/genética , Células Epiteliais/metabolismo , Regulação Neoplásica da Expressão Gênica , Queratina-15/genética , Ureter/metabolismo , Neoplasias da Bexiga Urinária/genética , Urotélio/metabolismo , Idoso , Animais , Carcinoma Basocelular/metabolismo , Carcinoma Basocelular/patologia , Diferenciação Celular , Embrião de Mamíferos , Células Epiteliais/patologia , Feminino , Feto , Regulação da Expressão Gênica no Desenvolvimento , Homozigoto , Humanos , Queratina-15/metabolismo , Masculino , Camundongos , Pessoa de Meia-Idade , Morfogênese/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Análise Serial de Tecidos , Transativadores/genética , Transativadores/metabolismo , Transcriptoma , Ureter/citologia , Ureter/embriologia , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Uroplaquina III/genética , Uroplaquina III/metabolismo , Urotélio/patologia
13.
Hum Mol Genet ; 22(6): 1206-17, 2013 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-23283078

RESUMO

Hirschsprung disease (HSCR) is a human congenital disorder, defined by the absence of ganglia from variable lengths of the colon. These ganglia comprise the enteric nervous system (ENS) and are derived from migratory neural crest cells (NCCs). The inheritance of HSCR is complex, often non-Mendelian and characterized by variable penetrance. Although extensive research has identified many key players in the pathogenesis of Hirschsprung disease, a large number of cases remain genetically undefined. Therefore, additional unidentified genes or modifiers must contribute to the etiology and pathogenesis of Hirschsprung disease. We have discovered that Tcof1 may be one such modifier. Haploinsufficiency of Tcof1 in mice results in a reduction of vagal NCCs and their delayed migration along the length of the gut during early development. This alone, however, is not sufficient to cause colonic aganglionosis as alterations in the balance of NCC proliferation and differentiation ensures NCC colonize the entire length of the gut of Tcof1(+/-) mice by E18.5. In contrast, Tcof1 haploinsufficiency is able to sensitize Pax3(+/-) mice to colonic aganglionosis. Although, Pax3 heterozygous mice do not show ENS defects, compound Pax3;Tcof1 heterozygous mice exhibit cumulative apoptosis which severely reduces the NCC population that migrates into the foregut. In addition, the proliferative capacity of these NCC is also diminished. Taken together with the opposing effects of Pax3 and Tcof1 on NCC differentiation, the synergistic haploinsufficiency of Tcof1 and Pax3 results in colonic aganglionosis in mice and may contribute to the pathogenesis of Hirschsprung disease.


Assuntos
Sistema Nervoso Entérico/embriologia , Doença de Hirschsprung/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição Box Pareados/metabolismo , Fosfoproteínas/metabolismo , Animais , Movimento Celular , Proliferação de Células , Colo/embriologia , Colo/inervação , Colo/metabolismo , Colo/patologia , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/patologia , Feminino , Doença de Hirschsprung/embriologia , Doença de Hirschsprung/genética , Doença de Hirschsprung/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Knockout , Crista Neural/citologia , Crista Neural/metabolismo , Crista Neural/patologia , Proteínas Nucleares/genética , Fator de Transcrição PAX3 , Fatores de Transcrição Box Pareados/genética , Fosfoproteínas/genética
14.
Hum Mol Genet ; 22(3): 531-43, 2013 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-23108156

RESUMO

Ankyloblepharon, ectodermal defects, cleft lip/palate (AEC) syndrome is a rare autosomal dominant disorder caused by mutations in the p63 gene, essential for embryonic development of stratified epithelia. The most severe cutaneous manifestation of this disorder is the long-lasting skin fragility associated with severe skin erosions after birth. Using a knock-in mouse model for AEC syndrome, we found that skin fragility was associated with microscopic blistering between the basal and suprabasal compartments of the epidermis and reduced desmosomal contacts. Expression of desmosomal cadherins and desmoplakin was strongly reduced in AEC mutant keratinocytes and in newborn epidermis. A similar impairment in desmosome gene expression was observed in human keratinocytes isolated from AEC patients, in p63-depleted keratinocytes and in p63 null embryonic skin, indicating that p63 mutations causative of AEC syndrome have a dominant-negative effect on the wild-type p63 protein. Among the desmosomal components, desmocollin 3, desmoplakin and desmoglein 1 were the most significantly reduced by mutant p63 both at the RNA and protein levels. Chromatin immunoprecipitation experiments and transactivation assays revealed that p63 controls these genes at the transcriptional level. Consistent with reduced desmosome function, AEC mutant and p63-deficient keratinocytes had an impaired ability to withstand mechanical stress, which was alleviated by epidermal growth factor receptor inhibitors known to stabilize desmosomes. Our study reveals that p63 is a crucial regulator of a subset of desmosomal genes and that this function is impaired in AEC syndrome. Reduced mechanical strength resulting from p63 mutations can be alleviated pharmacologically by increasing desmosome adhesion with possible therapeutic implications.


Assuntos
Fenda Labial/genética , Fissura Palatina/genética , Desmossomos/metabolismo , Displasia Ectodérmica/genética , Anormalidades do Olho/genética , Proteínas de Membrana/metabolismo , Animais , Adesão Celular , Células Cultivadas , Imunoprecipitação da Cromatina , Fenda Labial/patologia , Fissura Palatina/patologia , Clonagem Molecular , Desmossomos/genética , Displasia Ectodérmica/patologia , Epiderme/metabolismo , Epiderme/fisiopatologia , Epitélio/metabolismo , Epitélio/fisiopatologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Receptores ErbB/metabolismo , Anormalidades do Olho/patologia , Pálpebras/anormalidades , Pálpebras/patologia , Feminino , Regulação da Expressão Gênica , Humanos , Queratinócitos/metabolismo , Luciferases/análise , Luciferases/metabolismo , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Reação em Cadeia da Polimerase em Tempo Real , Pele/metabolismo , Pele/fisiopatologia
15.
PLoS Genet ; 8(3): e1002566, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22479190

RESUMO

The cerebral cortex is a specialized region of the brain that processes cognitive, motor, somatosensory, auditory, and visual functions. Its characteristic architecture and size is dependent upon the number of neurons generated during embryogenesis and has been postulated to be governed by symmetric versus asymmetric cell divisions, which mediate the balance between progenitor cell maintenance and neuron differentiation, respectively. The mechanistic importance of spindle orientation remains controversial, hence there is considerable interest in understanding how neural progenitor cell mitosis is controlled during neurogenesis. We discovered that Treacle, which is encoded by the Tcof1 gene, is a novel centrosome- and kinetochore-associated protein that is critical for spindle fidelity and mitotic progression. Tcof1/Treacle loss-of-function disrupts spindle orientation and cell cycle progression, which perturbs the maintenance, proliferation, and localization of neural progenitors during cortical neurogenesis. Consistent with this, Tcof1(+/-) mice exhibit reduced brain size as a consequence of defects in neural progenitor maintenance. We determined that Treacle elicits its effect via a direct interaction with Polo-like kinase1 (Plk1), and furthermore we discovered novel in vivo roles for Plk1 in governing mitotic progression and spindle orientation in the developing mammalian cortex. Increased asymmetric cell division, however, did not promote increased neuronal differentiation. Collectively our research has therefore identified Treacle and Plk1 as novel in vivo regulators of spindle fidelity, mitotic progression, and proliferation in the maintenance and localization of neural progenitor cells. Together, Treacle and Plk1 are critically required for proper cortical neurogenesis, which has important implications in the regulation of mammalian brain size and the pathogenesis of congenital neurodevelopmental disorders such as microcephaly.


Assuntos
Proteínas de Ciclo Celular , Córtex Cerebral/crescimento & desenvolvimento , Mitose/genética , Neurogênese/genética , Neurônios/citologia , Proteínas Nucleares , Fosfoproteínas , Proteínas Serina-Treonina Quinases , Proteínas Proto-Oncogênicas , Animais , Pontos de Checagem do Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Proliferação de Células , Centrossomo/metabolismo , Córtex Cerebral/anormalidades , Regulação da Expressão Gênica no Desenvolvimento , Células HeLa , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Pontos de Checagem da Fase M do Ciclo Celular/genética , Disostose Mandibulofacial/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos DBA , Camundongos Mutantes , Neurônios/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Quinase 1 Polo-Like
16.
Hum Mol Genet ; 21(8): 1782-93, 2012 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-22228097

RESUMO

The enteric nervous system (ENS) comprises a complex neuronal network that regulates peristalsis of the gut wall and secretions into the lumen. The ENS is formed from a multipotent progenitor cell population called the neural crest, which is derived from the neuroepithelium. Neural crest cells (NCCs) migrate over incredible distances to colonize the entire length of the gut and during their migration they must survive, proliferate and ultimately differentiate. The absence of an ENS from variable lengths of the colon results in Hirschsprung's disease (HSCR) or colonic aganglionosis. Mutations in about 12 different genes have been identified in HSCR patients but the complex pattern of inheritance and variable penetrance suggests that additional genes or modifiers must be involved in the etiology and pathogenesis of this disease. We discovered that Tcof1 haploinsufficiency in mice models many of the early features of HSCR. Neuroepithelial apoptosis diminished the size of the neural stem cell pool resulting in reduced NCC numbers and their delayed migration along the gut from E10.5 to E14.5. Surprisingly however, we observe continued and complete colonization of the entire colon throughout E14.5-E18.5, a period in which the gut is considered to be non- or less-permissive to NCC. Thus, we reveal for the first time that reduced NCC progenitor numbers and delayed migration do not unequivocally equate with a predisposition for the pathogenesis of HSCR. In fact, these deficiencies can be overcome by balancing NCC intrinsic processes of proliferation and differentiation with extrinsic influences of the gut microenvironment.


Assuntos
Sistema Nervoso Entérico/embriologia , Doença de Hirschsprung , Crista Neural/citologia , Proteínas Nucleares/genética , Proteínas Nucleares/fisiologia , Fosfoproteínas/genética , Fosfoproteínas/fisiologia , Animais , Apoptose , Contagem de Células , Movimento Celular , Proliferação de Células , Microambiente Celular , Modelos Animais de Doenças , Proteínas da Matriz Extracelular/metabolismo , Trato Gastrointestinal/citologia , Trato Gastrointestinal/embriologia , Trato Gastrointestinal/inervação , Haploinsuficiência , Doença de Hirschsprung/embriologia , Doença de Hirschsprung/genética , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Crista Neural/fisiologia , Tubo Neural/citologia , Tubo Neural/embriologia , Neurogênese , Células-Tronco/citologia , Células-Tronco/fisiologia , Nervo Vago/embriologia
17.
EMBO Mol Med ; 4(3): 192-205, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22247000

RESUMO

Ankyloblepharon-ectodermal defects-cleft lip/palate (AEC) syndrome, which is characterized by cleft palate and severe defects of the skin, is an autosomal dominant disorder caused by mutations in the gene encoding transcription factor p63. Here, we report the generation of a knock-in mouse model for AEC syndrome (p63(+/L514F) ) that recapitulates the human disorder. The AEC mutation exerts a selective dominant-negative function on wild-type p63 by affecting progenitor cell expansion during ectodermal development leading to a defective epidermal stem cell compartment. These phenotypes are associated with impairment of fibroblast growth factor (FGF) signalling resulting from reduced expression of Fgfr2 and Fgfr3, direct p63 target genes. In parallel, a defective stem cell compartment is observed in humans affected by AEC syndrome and in Fgfr2b(-/-) mice. Restoring Fgfr2b expression in p63(+/L514F) epithelial cells by treatment with FGF7 reactivates downstream mitogen-activated protein kinase signalling and cell proliferation. These findings establish a functional link between FGF signalling and p63 in the expansion of epithelial progenitor cells and provide mechanistic insights into the pathogenesis of AEC syndrome.


Assuntos
Proliferação de Células , Fenda Labial/metabolismo , Fissura Palatina/metabolismo , Ectoderma/citologia , Displasia Ectodérmica/metabolismo , Anormalidades do Olho/metabolismo , Fatores de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais , Células-Tronco/citologia , Fatores de Transcrição/genética , Proteínas Supressoras de Tumor/genética , Animais , Fenda Labial/genética , Fenda Labial/fisiopatologia , Fissura Palatina/genética , Fissura Palatina/fisiopatologia , Ectoderma/metabolismo , Displasia Ectodérmica/genética , Displasia Ectodérmica/fisiopatologia , Anormalidades do Olho/genética , Anormalidades do Olho/fisiopatologia , Pálpebras/anormalidades , Pálpebras/metabolismo , Pálpebras/fisiopatologia , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Mutação , Células-Tronco/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
18.
Am J Hum Genet ; 90(1): 69-75, 2012 Jan 13.
Artigo em Inglês | MEDLINE | ID: mdl-22197488

RESUMO

Pterygium syndromes are complex congenital disorders that encompass several distinct clinical conditions characterized by multiple skin webs affecting the flexural surfaces often accompanied by craniofacial anomalies. In severe forms, such as in the autosomal-recessive Bartsocas-Papas syndrome, early lethality is common, complicating the identification of causative mutations. Using exome sequencing in a consanguineous family, we identified the homozygous mutation c.1127C>A in exon 7 of RIPK4 that resulted in the introduction of the nonsense mutation p.Ser376X into the encoded ankyrin repeat-containing kinase, a protein that is essential for keratinocyte differentiation. Subsequently, we identified a second mutation in exon 2 of RIPK4 (c.242T>A) that resulted in the missense variant p.Ile81Asn in the kinase domain of the protein. We have further demonstrated that RIPK4 is a direct transcriptional target of the protein p63, a master regulator of stratified epithelial development, which acts as a nodal point in the cascade of molecular events that prevent pterygium syndromes.


Assuntos
Fenda Labial/genética , Fissura Palatina/genética , Exoma , Proteínas Serina-Treonina Quinases/genética , Pterígio/congênito , Sequência de Aminoácidos , Animais , Sequência de Bases , Criança , Fenda Labial/diagnóstico , Fissura Palatina/diagnóstico , Consanguinidade , Anormalidades Craniofaciais/genética , Éxons , Genes Recessivos , Loci Gênicos , Humanos , Queratinócitos/metabolismo , Masculino , Camundongos , Dados de Sequência Molecular , Mutação , Fosfoproteínas/metabolismo , Pterígio/diagnóstico , Pterígio/genética , Índice de Gravidade de Doença , Anormalidades da Pele , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas Supressoras de Tumor/metabolismo
19.
Dev Cell ; 21(4): 627-41, 2011 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-21982646

RESUMO

Morphogenesis of mammalian facial processes requires coordination of cellular proliferation, migration, and apoptosis to develop intricate features. Cleft lip and/or palate (CL/P), the most frequent human craniofacial birth defect, can be caused by perturbation of any of these programs. Mutations of WNT, P63, and IRF6 yield CL/P in humans and mice; however, how these genes are regulated remains elusive. We generated mouse lines lacking Pbx genes in cephalic ectoderm and demonstrated that they exhibit fully penetrant CL/P and perturbed Wnt signaling. We also characterized a midfacial regulatory element that Pbx proteins bind to control the expression of Wnt9b-Wnt3, which in turn regulates p63. Altogether, we establish a Pbx-dependent Wnt-p63-Irf6 regulatory module in midfacial ectoderm that is conserved within mammals. Dysregulation of this network leads to localized suppression of midfacial apoptosis and CL/P. Ectopic Wnt ectodermal expression in Pbx mutants rescues the clefting, opening avenues for tissue repair.


Assuntos
Apoptose , Células Epiteliais/metabolismo , Face/embriologia , Proteínas de Homeodomínio/fisiologia , Fatores Reguladores de Interferon/metabolismo , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Fatores de Transcrição/fisiologia , Proteínas Wnt/metabolismo , Proteína Wnt3/metabolismo , Animais , Sequência de Bases , Western Blotting , Proliferação de Células , Imunoprecipitação da Cromatina , Fenda Labial/embriologia , Fenda Labial/metabolismo , Fissura Palatina/embriologia , Fissura Palatina/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Técnicas Imunoenzimáticas , Fatores Reguladores de Interferon/genética , Luciferases/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos , Dados de Sequência Molecular , Morfogênese/fisiologia , Fenótipo , Fosfoproteínas/genética , Fator de Transcrição 1 de Leucemia de Células Pré-B , RNA Mensageiro/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Homologia de Sequência do Ácido Nucleico , Transativadores/genética , Transfecção , Proteínas Wnt/genética , Proteína Wnt3/genética
20.
Nat Genet ; 43(1): 20-2, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21131976

RESUMO

We identified a deletion of a gene encoding a subunit of RNA polymerases I and III, POLR1D, in an individual with Treacher Collins syndrome (TCS). Subsequently, we detected 20 additional heterozygous mutations of POLR1D in 252 individuals with TCS. Furthermore, we discovered mutations in both alleles of POLR1C in three individuals with TCS. These findings identify two additional genes involved in TCS, confirm the genetic heterogeneity of TCS and support the hypothesis that TCS is a ribosomopathy.


Assuntos
Disostose Mandibulofacial/genética , Mutação , RNA Polimerase III/genética , RNA Polimerase I/genética , Adulto , Alelos , Criança , Pré-Escolar , Feminino , Heterogeneidade Genética , Humanos , Masculino , Disostose Mandibulofacial/enzimologia , Fenótipo , Subunidades Proteicas/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...