Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Cell Rep ; 39(12): 110988, 2022 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-35732123

RESUMO

MacroH2A histone variants have a function in gene regulation that is poorly understood at the molecular level. We report that macroH2A1.2 and macroH2A2 modulate the transcriptional ground state of cancer cells and how they respond to inflammatory cytokines. Removal of macroH2A1.2 and macroH2A2 in hepatoblastoma cells affects the contact frequency of promoters and distal enhancers coinciding with changes in enhancer activity or preceding them in response to the cytokine tumor necrosis factor alpha. Although macroH2As regulate genes in both directions, they globally facilitate the nuclear factor κB (NF-κB)-mediated response. In contrast, macroH2As suppress the response to the pro-inflammatory cytokine interferon gamma. MacroH2A2 has a stronger contribution to gene repression than macroH2A1.2. Taken together, our results suggest that macroH2As have a role in regulating the response of cancer cells to inflammatory signals on the level of chromatin structure. This is likely relevant for the interaction of cancer cells with immune cells of their microenvironment.


Assuntos
Citocinas , Regulação da Expressão Gênica , NF-kappa B , Regiões Promotoras Genéticas/genética
2.
Proc Natl Acad Sci U S A ; 118(46)2021 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-34750251

RESUMO

One of the central problems of vertebrate evolution is understanding the relationship among the distal portions of fins and limbs. Lacking comparable morphological markers of these regions in fish and tetrapods, these relationships have remained uncertain for the past century and a half. Here we show that Gli3 functions in controlling the proliferative expansion of distal progenitors are shared among dorsal and paired fins as well as tetrapod limbs. Mutant knockout gli3 fins in medaka (Oryzias latipes) form multiple radials and rays, in a pattern reminiscent of the polydactyly observed in Gli3-null mutant mice. In limbs, Gli3 controls both anterior-posterior patterning and cell proliferation, two processes that can be genetically uncoupled. In situ hybridization, quantification of proliferation markers, and analysis of regulatory regions reveal that in paired and dorsal fins, gli3 plays a main role in controlling proliferation but not in patterning. Moreover, gli3 down-regulation in shh mutant fins rescues fin loss in a manner similar to how Gli3 deficiency restores digits in the limbs of Shh mutant mouse embryos. We hypothesize that the Gli3/Shh gene pathway preceded the origin of paired appendages and was originally involved in modulating cell proliferation. Accordingly, the distal regions of dorsal fins, paired fins, and limbs retain a deep regulatory and functional homology that predates the origin of paired appendages.


Assuntos
Nadadeiras de Animais/crescimento & desenvolvimento , Redes Reguladoras de Genes/genética , Proteínas do Tecido Nervoso/genética , Oryzias/genética , Proteína Gli3 com Dedos de Zinco/genética , Animais , Evolução Biológica , Padronização Corporal/genética , Proliferação de Células/genética , Extremidades/crescimento & desenvolvimento , Proteínas de Peixes/genética , Regulação da Expressão Gênica no Desenvolvimento/genética , Camundongos
3.
Science ; 372(6545): 984-989, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-34045355

RESUMO

We investigated genome folding across the eukaryotic tree of life. We find two types of three-dimensional (3D) genome architectures at the chromosome scale. Each type appears and disappears repeatedly during eukaryotic evolution. The type of genome architecture that an organism exhibits correlates with the absence of condensin II subunits. Moreover, condensin II depletion converts the architecture of the human genome to a state resembling that seen in organisms such as fungi or mosquitoes. In this state, centromeres cluster together at nucleoli, and heterochromatin domains merge. We propose a physical model in which lengthwise compaction of chromosomes by condensin II during mitosis determines chromosome-scale genome architecture, with effects that are retained during the subsequent interphase. This mechanism likely has been conserved since the last common ancestor of all eukaryotes.


Assuntos
Adenosina Trifosfatases/genética , Adenosina Trifosfatases/fisiologia , Evolução Biológica , Cromossomos/ultraestrutura , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/fisiologia , Eucariotos/genética , Genoma , Complexos Multiproteicos/genética , Complexos Multiproteicos/fisiologia , Adenosina Trifosfatases/química , Algoritmos , Animais , Nucléolo Celular/ultraestrutura , Núcleo Celular/ultraestrutura , Centrômero/ultraestrutura , Cromossomos/química , Cromossomos Humanos/química , Cromossomos Humanos/ultraestrutura , Proteínas de Ligação a DNA/química , Genoma Humano , Genômica , Heterocromatina/ultraestrutura , Humanos , Interfase , Mitose , Modelos Biológicos , Complexos Multiproteicos/química , Telômero/ultraestrutura
4.
Mol Biol Evol ; 37(10): 2857-2864, 2020 10 01.
Artigo em Inglês | MEDLINE | ID: mdl-32421818

RESUMO

We investigated how the two rounds of whole-genome duplication that occurred at the base of the vertebrate lineage have impacted ancient microsyntenic associations involving developmental regulators (known as genomic regulatory blocks, GRBs). We showed that the majority of GRBs identified in the last common ancestor of chordates have been maintained as a single copy in humans. We found evidence that dismantling of the duplicated GRB copies occurred early in vertebrate evolution often through the differential retention of the regulatory gene but loss of the bystander gene's exonic sequences. Despite the large evolutionary scale, the presence of duplicated highly conserved noncoding regions provided unambiguous proof for this scenario for multiple ancient GRBs. Remarkably, the dismantling of ancient GRB duplicates has contributed to the creation of large gene deserts associated with regulatory genes in vertebrates, providing a potentially widespread mechanism for the origin of these enigmatic genomic traits.


Assuntos
Evolução Molecular , Regulação da Expressão Gênica no Desenvolvimento , Genes Reguladores , Poliploidia , Vertebrados/genética , Animais , Duplicação Cromossômica , Genoma Humano , Humanos , Elementos Reguladores de Transcrição
5.
Development ; 146(13)2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31142542

RESUMO

Yap1/Taz are well-known Hippo effectors triggering complex transcriptional programs controlling growth, survival and cancer progression. Here, we describe yap1b, a new Yap1/Taz family member with a unique transcriptional activation domain that cannot be phosphorylated by Src/Yes kinases. We show that yap1b evolved specifically in euteleosts (i.e. including medaka but not zebrafish) by duplication and adaptation of yap1. Using DamID-seq, we generated maps of chromatin occupancy for Yap1, Taz (Wwtr1) and Yap1b in gastrulating zebrafish and medaka embryos. Our comparative analyses uncover the genetic programs controlled by Yap family proteins during early embryogenesis, and show largely overlapping targets for Yap1 and Yap1b. CRISPR/Cas9-induced mutation of yap1b in medaka does not result in an overt phenotype during embryogenesis or adulthood. However, yap1b mutation strongly enhances the embryonic malformations observed in yap1 mutants. Thus yap1-/-; yap1b-/- double mutants display more severe body flattening, eye misshaping and increased apoptosis than yap1-/- single mutants, thus revealing overlapping gene functions. Our results indicate that, despite its divergent transactivation domain, Yap1b cooperates with Yap1 to regulate cell survival and tissue morphogenesis during early development.


Assuntos
Perda do Embrião/genética , Regulação da Expressão Gênica no Desenvolvimento , Morfogênese/genética , Transativadores/fisiologia , Proteínas de Peixe-Zebra/fisiologia , Animais , Animais Geneticamente Modificados , Perda do Embrião/veterinária , Embrião não Mamífero , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Mutação , Oryzias/embriologia , Oryzias/genética , Domínios Proteicos/genética , Isoformas de Proteínas/química , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiologia , Transativadores/química , Transativadores/genética , Fatores de Transcrição/química , Fatores de Transcrição/genética , Fatores de Transcrição/fisiologia , Proteínas com Motivo de Ligação a PDZ com Coativador Transcricional , Proteínas de Sinalização YAP , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
6.
PLoS Comput Biol ; 14(3): e1006030, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29522512

RESUMO

The use of 3C-based methods has revealed the importance of the 3D organization of the chromatin for key aspects of genome biology. However, the different caveats of the variants of 3C techniques have limited their scope and the range of scientific fields that could benefit from these approaches. To address these limitations, we present 4Cin, a method to generate 3D models and derive virtual Hi-C (vHi-C) heat maps of genomic loci based on 4C-seq or any kind of 4C-seq-like data, such as those derived from NG Capture-C. 3D genome organization is determined by integrative consideration of the spatial distances derived from as few as four 4C-seq experiments. The 3D models obtained from 4C-seq data, together with their associated vHi-C maps, allow the inference of all chromosomal contacts within a given genomic region, facilitating the identification of Topological Associating Domains (TAD) boundaries. Thus, 4Cin offers a much cheaper, accessible and versatile alternative to other available techniques while providing a comprehensive 3D topological profiling. By studying TAD modifications in genomic structural variants associated to disease phenotypes and performing cross-species evolutionary comparisons of 3D chromatin structures in a quantitative manner, we demonstrate the broad potential and novel range of applications of our method.


Assuntos
Mapeamento Cromossômico/métodos , Biologia Computacional/métodos , Imageamento Tridimensional/métodos , Cromatina/fisiologia , Cromossomos , Simulação por Computador , Genoma , Genômica/métodos , Conformação de Ácido Nucleico , Análise de Sequência de DNA/métodos , Software
7.
Development ; 142(17): 3009-20, 2015 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-26253404

RESUMO

Microphthalmos is a rare congenital anomaly characterized by reduced eye size and visual deficits of variable degree. Sporadic and hereditary microphthalmos have been associated with heterozygous mutations in genes fundamental for eye development. Yet, many cases are idiopathic or await the identification of molecular causes. Here we show that haploinsufficiency of Meis1, which encodes a transcription factor with evolutionarily conserved expression in the embryonic trunk, brain and sensory organs, including the eye, causes microphthalmic traits and visual impairment in adult mice. By combining analysis of Meis1 loss-of-function and conditional Meis1 functional rescue with ChIP-seq and RNA-seq approaches we show that, in contrast to its preferential association with Hox-Pbx BSs in the trunk, Meis1 binds to Hox/Pbx-independent sites during optic cup development. In the eye primordium, Meis1 coordinates, in a dose-dependent manner, retinal proliferation and differentiation by regulating genes responsible for human microphthalmia and components of the Notch signaling pathway. In addition, Meis1 is required for eye patterning by controlling a set of eye territory-specific transcription factors, so that in Meis1(-/-) embryos boundaries among the different eye territories are shifted or blurred. We propose that Meis1 is at the core of a genetic network implicated in eye patterning/microphthalmia, and represents an additional candidate for syndromic cases of these ocular malformations.


Assuntos
Olho/embriologia , Olho/metabolismo , Redes Reguladoras de Genes , Proteínas de Homeodomínio/metabolismo , Microftalmia/embriologia , Microftalmia/genética , Proteínas de Neoplasias/metabolismo , Envelhecimento/patologia , Animais , Apoptose/genética , Sequência de Bases , Sítios de Ligação , Vasos Sanguíneos/metabolismo , Vasos Sanguíneos/patologia , Imunoprecipitação da Cromatina , Embrião de Mamíferos/metabolismo , Embrião de Mamíferos/patologia , Elementos Facilitadores Genéticos/genética , Haploinsuficiência/genética , Hematopoese/genética , Proteínas de Homeodomínio/genética , Humanos , Camundongos , Dados de Sequência Molecular , Proteína Meis1 , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Neurogênese/genética , Ligação Proteica , Receptores Notch/metabolismo , Transdução de Sinais/genética
8.
Biochim Biophys Acta ; 1849(8): 955-65, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26079690

RESUMO

The three-dimensional architecture of genomes provides new insights about genome organization and function, but many aspects remain unsolved at the local genomic scale. Here we investigate the regulation of two erythroid-specific loci, a folate receptor gene (FOLR1) and the ß-globin gene cluster, which are separated by 16kb of constitutive heterochromatin. We found that in early erythroid differentiation the FOLR1 gene presents a permissive chromatin configuration that allows its expression. Once the transition to the next differentiation state occurs, the heterochromatin spreads into the FOLR1 domain, concomitant with the dissociation of CTCF from a novel binding site, thereby resulting in irreversible silencing of the FOLR1 gene. We demonstrate that the sequences surrounding the CTCF-binding site possess classical insulator properties in vitro and in vivo. In contrast, the chicken cHS4 ß-globin insulator present on the other side of the heterochromatic segment is in a constitutive open chromatin configuration, with CTCF constantly bound from the early stages of erythroid differentiation. Therefore, this study demonstrates that the 16kb of constitutive heterochromatin contributes to silencing of the FOLR1 gene during erythroid differentiation.


Assuntos
Receptor 1 de Folato/genética , Loci Gênicos , Elementos Isolantes/fisiologia , Globinas beta/genética , Animais , Diferenciação Celular/genética , Linhagem Celular Transformada , Embrião de Galinha , Galinhas , Cromatina/genética , Cromatina/metabolismo , Eritropoese/genética , Receptor 1 de Folato/metabolismo , Regulação da Expressão Gênica , Heterocromatina/genética , Heterocromatina/metabolismo
9.
Nat Cell Biol ; 17(5): 615-626, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25915126

RESUMO

The genomic regulatory programmes that underlie human organogenesis are poorly understood. Pancreas development, in particular, has pivotal implications for pancreatic regeneration, cancer and diabetes. We have now characterized the regulatory landscape of embryonic multipotent progenitor cells that give rise to all pancreatic epithelial lineages. Using human embryonic pancreas and embryonic-stem-cell-derived progenitors we identify stage-specific transcripts and associated enhancers, many of which are co-occupied by transcription factors that are essential for pancreas development. We further show that TEAD1, a Hippo signalling effector, is an integral component of the transcription factor combinatorial code of pancreatic progenitor enhancers. TEAD and its coactivator YAP activate key pancreatic signalling mediators and transcription factors, and regulate the expansion of pancreatic progenitors. This work therefore uncovers a central role for TEAD and YAP as signal-responsive regulators of multipotent pancreatic progenitors, and provides a resource for the study of embryonic development of the human pancreas.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células-Tronco Embrionárias/metabolismo , Células-Tronco Multipotentes/metabolismo , Proteínas Nucleares/metabolismo , Pâncreas/metabolismo , Fosfoproteínas/metabolismo , Transdução de Sinais , Fatores de Transcrição/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Animais Geneticamente Modificados , Diferenciação Celular , Linhagem da Célula , Proliferação de Células , Células Cultivadas , Biologia Computacional , Proteínas de Ligação a DNA/genética , Bases de Dados Genéticas , Regulação da Expressão Gênica no Desenvolvimento , Redes Reguladoras de Genes , Humanos , Camundongos Endogâmicos C57BL , Proteínas Nucleares/genética , Organogênese , Pâncreas/embriologia , Fenótipo , Fosfoproteínas/genética , RNA Mensageiro/metabolismo , Fatores de Transcrição de Domínio TEA , Fatores de Tempo , Fatores de Transcrição/genética , Proteínas de Sinalização YAP , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
10.
Dev Dyn ; 244(5): 693-702, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25728805

RESUMO

BACKGROUND: SPARC/osteonectin is an evolutionarily conserved matricellular protein that modulates cell-matrix interaction and cell function. In all vertebrates, SPARC is dynamically expressed during embryogenesis. However, the precise function of SPARC and the regulatory elements required for its expression in particular during early embryogenesis are largely unknown. RESULTS: The present study was undertaken to explore the molecular mechanisms that regulate sparc gene expression by in vivo functional characterization of the sparc promoter and identification of possible putative regulatory elements that govern basal promoter activity. We report here transient expression analyses of eGFP expression from transgenic zebrafish containing a Sparc-iTol2-eGFP-BAC and/or 7.25 kb-sparc-Tol2-eGFP constructs. eGFP expression was specifically found in the notochord, otic vesicle, fin fold, intermediate cell mass, and olfactory placode of BAC and Tol2 transposon vectors injected embryos. Deletion analysis revealed that promoter activity resides in the unique 5'-untranslated intronic region. Computer-based analysis revealed a putative CpG island immediately proximal to the translation start site within the intron sequence. Global inhibition of methylation with 5-Aza-2-deoxycytidine promoted sparc expression in association with decreasing CpG methylation. CONCLUSIONS: Taken together, these data identify a contributory role for DNA methylation in regulating sparc expression in zebrafish embryogenesis.


Assuntos
Metilação de DNA/fisiologia , Embrião não Mamífero/embriologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Osteonectina/biossíntese , Regiões Promotoras Genéticas/fisiologia , Peixe-Zebra/embriologia , Animais , Animais Geneticamente Modificados , Embrião não Mamífero/citologia , Osteonectina/genética , Peixe-Zebra/genética
11.
Dev Neurobiol ; 75(1): 80-92, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25044975

RESUMO

The development of organs composed by repeated functional units is, in many cases, accomplished by the transition of cells from a progenitor to a differentiation domain, triggering a reiterated developmental program. Yet, how these discrete fields are formed during development is still a largely unresolved question. The posterior lateral line (pLL), a sensory organ present in fish and amphibians, develops from a primordium that migrates along the flanks of the animal periodically depositing neuromasts, the pLL functional units. In zebrafish (Danio rerio), the developmental program of the pLL is triggered by the transit of progenitor cells from a Wnt to a Fgf signaling domain. It has been proposed that these two fields are defined by the antagonistic activity of these two signaling pathways, but how they are formed and maintained is still an open question in the development of the pLL. In this work, we show that sox21a, an HMG -box transcription factor, is expressed within the Fgf domain. We demonstrate that, while the Fgf signaling pathway do not control sox21a, knockdown of sox21a causes impairment of Fgf signaling, expansion of the Wnt signaling domain and disruption of neuromast development. These results suggest that sox21a is a key player in the pLL primordium patterning, fine-tuning the border of the Fgf and Wnt signaling domains.


Assuntos
Padronização Corporal/fisiologia , Fatores de Crescimento de Fibroblastos/metabolismo , Sistema da Linha Lateral/embriologia , Fatores de Transcrição SOXB2/fisiologia , Transdução de Sinais/fisiologia , Proteínas de Peixe-Zebra/metabolismo , Animais , Embrião não Mamífero , Via de Sinalização Wnt/fisiologia , Peixe-Zebra , Proteínas de Peixe-Zebra/fisiologia
12.
Cell Rep ; 8(4): 983-90, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25131200

RESUMO

A rare germline duplication upstream of the bone morphogenetic protein antagonist GREM1 causes a Mendelian-dominant predisposition to colorectal cancer (CRC). The underlying disease mechanism is strong, ectopic GREM1 overexpression in the intestinal epithelium. Here, we confirm that a common GREM1 polymorphism, rs16969681, is also associated with CRC susceptibility, conferring ∼20% differential risk in the general population. We hypothesized the underlying cause to be moderate differences in GREM1 expression. We showed that rs16969681 lies in a region of active chromatin with allele- and tissue-specific enhancer activity. The CRC high-risk allele was associated with stronger gene expression, and higher Grem1 mRNA levels increased the intestinal tumor burden in Apc(Min) mice. The intestine-specific transcription factor CDX2 and Wnt effector TCF7L2 bound near rs16969681, with significantly higher affinity for the risk allele, and CDX2 overexpression in CDX2/GREM1-negative cells caused re-expression of GREM1. rs16969681 influences CRC risk through effects on Wnt-driven GREM1 expression in colorectal tumors.


Assuntos
Neoplasias do Colo/genética , Proteínas de Homeodomínio/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/genética , Proteína 2 Semelhante ao Fator 7 de Transcrição/metabolismo , Animais , Sequência de Bases , Fator de Transcrição CDX2 , Linhagem Celular Tumoral , Elementos Facilitadores Genéticos , Regulação Neoplásica da Expressão Gênica , Estudos de Associação Genética , Predisposição Genética para Doença , Humanos , Camundongos Transgênicos , Especificidade de Órgãos , Polimorfismo de Nucleotídeo Único , Risco
13.
Genome Res ; 24(4): 592-603, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24642863

RESUMO

Genome-wide association studies (GWAS) identified the MEIS1 locus for Restless Legs Syndrome (RLS), but causal single nucleotide polymorphisms (SNPs) and their functional relevance remain unknown. This locus contains a large number of highly conserved noncoding regions (HCNRs) potentially functioning as cis-regulatory modules. We analyzed these HCNRs for allele-dependent enhancer activity in zebrafish and mice and found that the risk allele of the lead SNP rs12469063 reduces enhancer activity in the Meis1 expression domain of the murine embryonic ganglionic eminences (GE). CREB1 binds this enhancer and rs12469063 affects its binding in vitro. In addition, MEIS1 target genes suggest a role in the specification of neuronal progenitors in the GE, and heterozygous Meis1-deficient mice exhibit hyperactivity, resembling the RLS phenotype. Thus, in vivo and in vitro analysis of a common SNP with small effect size showed allele-dependent function in the prospective basal ganglia representing the first neurodevelopmental region implicated in RLS.


Assuntos
Elementos Facilitadores Genéticos , Proteínas de Homeodomínio/genética , Proteínas de Neoplasias/genética , Síndrome das Pernas Inquietas/genética , Telencéfalo/crescimento & desenvolvimento , Alelos , Animais , Gânglios da Base/metabolismo , Gânglios da Base/patologia , Modelos Animais de Doenças , Estudo de Associação Genômica Ampla , Íntrons , Camundongos , Proteína Meis1 , Polimorfismo de Nucleotídeo Único , Telencéfalo/patologia
14.
J Biol Chem ; 288(51): 36285-301, 2013 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-24214975

RESUMO

The homeodomain transcription factor Prrxl1/DRG11 has emerged as a crucial molecule in the establishment of the pain circuitry, in particular spinal cord targeting of dorsal root ganglia (DRG) axons and differentiation of nociceptive glutamatergic spinal cord neurons. Despite Prrxl1 importance in the establishment of the DRG-spinal nociceptive circuit, the molecular mechanisms that regulate its expression along development remain largely unknown. Here, we show that Prrxl1 transcription is regulated by three alternative promoters (named P1, P2, and P3), which control the expression of three distinct Prrxl1 5'-UTR variants, named 5'-UTR-A, 5'-UTR-B, and 5'-UTR-C. These 5'-UTR sequences confer distinct mRNA stability and translation efficiency to the Prrxl1 transcript. The most conserved promoter (P3) contains a TATA-box and displays in vivo enhancer activity in a pattern that overlaps with the zebrafish Prrxl1 homologue, drgx. Regulatory modules present in this sequence were identified and characterized, including a binding site for Phox2b. Concomitantly, we demonstrate that zebrafish Phox2b is required for the expression of drgx in the facial, glossopharyngeal, and vagal cranial ganglia.


Assuntos
Regiões 5' não Traduzidas , Proteínas de Homeodomínio/genética , Proteínas do Tecido Nervoso/genética , Estabilidade de RNA , RNA Mensageiro/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica , Animais , Sequência de Bases , Sítios de Ligação , Regulação da Expressão Gênica no Desenvolvimento , Células HEK293 , Células HeLa , Proteínas de Homeodomínio/metabolismo , Humanos , Camundongos , Dados de Sequência Molecular , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Células PC12 , Biossíntese de Proteínas , RNA Mensageiro/genética , Ratos , TATA Box , Fatores de Transcrição/metabolismo , Peixe-Zebra
15.
J Med Genet ; 49(7): 442-50, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22791839

RESUMO

BACKGROUND: SHOX, located in the pseudoautosomal region 1 (PAR1) of the sexual chromosomes, encodes a transcription factor implicated in human growth. Defects in SHOX or its enhancers have been observed in ∼60% of Leri-Weill dyschondrosteosis (LWD) patients, a skeletal dysplasia characterised by short stature and/or the characteristic Madelung deformity, and in 2-5% of idiopathic short stature (ISS). To identify the molecular defect in the remaining genetically undiagnosed LWD and ISS patients, this study screened previously unanalysed PAR1 regions in 124 LWD and 576 ISS probands. METHODS: PAR1 screening was undertaken by multiplex ligation dependent probe amplification (MLPA). Copy number alterations were subsequently confirmed and delimited by locus-specific custom-designed MLPA, array comparative genomic hybridisation (CGH) and breakpoint junction PCR/sequencing. RESULTS: A recurrent PAR1 deletion downstream of SHOX spanning 47543 bp with identical breakpoints was identified in 19 LWD (15.3%) and 11 ISS (1.9%) probands, from 30 unrelated families. Eight evolutionarily conserved regions (ECRs 1-8) identified within the deleted sequence were evaluated for SHOX regulatory activity by means of chromosome conformation capture (3C) in chicken embryo limbs and luciferase reporter assays in human U2OS osteosarcoma cells. The 3C assay indicated potential SHOX regulatory activity by ECR1, which was subsequently confirmed to act as a SHOX enhancer, operating in an orientation and position independent manner, in human U2OS cells. CONCLUSIONS: This study has identified the first recurrent PAR1 deletion in LWD and ISS, which results in the loss of a previously uncharacterised SHOX enhancer. The loss of this enhancer may decrease SHOX transcription, resulting in LWD or ISS due to SHOX haploinsufficiency.


Assuntos
Elementos Facilitadores Genéticos , Transtornos do Crescimento/genética , Proteínas de Homeodomínio/genética , Osteocondrodisplasias/genética , Deleção de Sequência/genética , Cromossomos Sexuais/genética , Animais , Embrião de Galinha , Criança , Estudos de Coortes , Extremidades/patologia , Feminino , Dosagem de Genes , Frequência do Gene , Haploinsuficiência , Proteínas de Homeodomínio/metabolismo , Humanos , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Linhagem , Cromossomos Sexuais/metabolismo , Proteína de Homoeobox de Baixa Estatura , Fatores de Transcrição/genética
16.
PLoS One ; 7(3): e33617, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22448256

RESUMO

Meis1, a conserved transcription factor of the TALE-homeodomain class, is expressed in a wide variety of tissues during development. Its complex expression pattern is likely to be controlled by an equally complex regulatory landscape. Here we have scanned the Meis1 locus for regulatory elements and found 13 non-coding regions, highly conserved between humans and teleost fishes, that have enhancer activity in stable transgenic zebrafish lines. All these regions are syntenic in most vertebrates. The composite expression of all these enhancer elements recapitulate most of Meis1 expression during early embryogenesis, indicating they comprise a basic set of regulatory elements of the Meis1 gene. Using bioinformatic tools, we identify a number of potential binding sites for transcription factors that are compatible with the regulation of these enhancers. Specifically, HHc2:066650, which is expressed in the developing retina and optic tectum, harbors several predicted Pax6 sites. Biochemical, functional and transgenic assays indicate that pax6 genes directly regulate HHc2:066650 activity.


Assuntos
Proteínas do Olho/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Proteínas de Neoplasias/genética , Fatores de Transcrição Box Pareados/metabolismo , Elementos Reguladores de Transcrição , Proteínas Repressoras/metabolismo , Peixe-Zebra/genética , Animais , Animais Geneticamente Modificados , Sítios de Ligação , Imunoprecipitação da Cromatina , Biologia Computacional , Olho/embriologia , Olho/metabolismo , Proteínas do Olho/genética , Humanos , Hibridização In Situ , Proteína Meis1 , Especificidade de Órgãos , Fator de Transcrição PAX6 , Fatores de Transcrição Box Pareados/genética , Proteínas Repressoras/genética , Peixe-Zebra/crescimento & desenvolvimento , Peixe-Zebra/metabolismo
17.
Development ; 139(2): 301-11, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22129829

RESUMO

Vertebrate odd-skipped related genes (Osr) have an essential function during the formation of the intermediate mesoderm (IM) and the kidney structures derived from it. Here, we show that these genes are also crucial for limb bud formation in the adjacent lateral plate mesoderm (LPM). Reduction of zebrafish Osr function impairs fin development by the failure of tbx5a maintenance in the developing pectoral fin bud. Osr morphant embryos show reduced wnt2b expression, and increasing Wnt signaling in Osr morphant embryos partially rescues tbx5a expression. Thus, Osr genes control limb bud development in a non-cell-autonomous manner, probably through the activation of Wnt2b. Finally, we demonstrate that Osr genes are downstream targets of retinoic acid (RA) signaling. Therefore, Osr genes act as a relay within the genetic cascade of fin bud formation: by controlling the expression of the signaling molecule Wnt2ba in the IM they play an essential function transmitting the RA signaling originated in the somites to the LPM.


Assuntos
Nadadeiras de Animais/embriologia , Botões de Extremidades/embriologia , Mesoderma/embriologia , Transdução de Sinais/fisiologia , Fatores de Transcrição/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/embriologia , Animais , Primers do DNA/genética , Dimetil Sulfóxido , Membro Anterior/embriologia , Hibridização In Situ , Pronefro/embriologia , Reação em Cadeia da Polimerase em Tempo Real , Proteínas com Domínio T/metabolismo , Tretinoína/metabolismo , Proteína Wnt2/metabolismo , p-Aminoazobenzeno/análogos & derivados
18.
J Biotechnol ; 157(3): 413-6, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22178780

RESUMO

Microbial host-pathogen interactions have been traditionally well studied at genetic and physiological levels, but cell-resolution analyses have been particularly scarce. This has been especially remarkable for intracellular parasites for two major reasons: first, the inherent loss of bacteria traceability once infects its hosts; second and more important, the limited availability of genetic tools that allow a tight regulated expression of bacterial virulence genes once inside the host tissues. Here we present novel data supporting the use of zebrafish embryos to monitor Salmonella enterica serovar Thyphimurium infection. Intravenous infection of Salmonella can be easily monitored using in vivo fluorescence that allows the visualization of free-swimming bacteria through the circulatory system. Moreover, we have engineered Salmonella to voluntarily activate heterologous gene expression at any point during infection once inside the zebrafish macrophages using a salicylate-based expression system. This approach allows real-time cell-resolution in vivo monitoring of the infection. All together, this approach paves the road to cell-based resolution experiments that would be harder to mimic in other vertebrate infection models.


Assuntos
Proteínas de Bactérias/metabolismo , Regulação Bacteriana da Expressão Gênica/fisiologia , Interações Hospedeiro-Patógeno/fisiologia , Infecções por Salmonella/diagnóstico , Infecções por Salmonella/fisiopatologia , Salmonella typhimurium/genética , Peixe-Zebra/embriologia , Animais , Sistemas Computacionais , Vermelho Congo , Embrião não Mamífero/microbiologia , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Engenharia Genética/métodos , Macrófagos/metabolismo , Microscopia de Fluorescência , Salicilatos/farmacologia , Infecções por Salmonella/microbiologia
19.
Dev Biol ; 350(2): 382-92, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21146514

RESUMO

The development of the Drosophila melanogaster wing depends on its subdivision into anterior and posterior compartments, which constitute two independent cell lineages since their origin in the embryonic ectoderm. The anterior-posterior compartment boundary is the place where signaling by the Hedgehog pathway takes place, and this requires pathway activation in anterior cells by ligand expressed exclusively in posterior cells. Several mechanisms ensure the confinement of hedgehog expression to posterior cells, including repression by Cubitus interruptus, the co-repressor Groucho and Master of thick veins. In this work we identified Kismet, a chromodomain-containing protein of the SNF2-like family of ATPases, as a novel component of the hedgehog transcriptional repression mechanism in anterior compartment cells. In kismet mutants, hedgehog is ectopically expressed in a domain of anterior cells close to the anterior-posterior compartment boundary, causing inappropriate activation of the pathway and changes in the development of the central region of the wing. The contribution of Kismet to the silencing of hedgehog expression is limited to anterior cells with low levels of the repressor form of Cubitus interruptus. We also show that knockdown of CHD8, the kismet homolog in Xenopus tropicalis, is also associated with ectopic sonic hedgehog expression and up-regulation of one of its target genes in the eye, Pax2, indicating the evolutionary conservation of Kismet/CHD8 function in negatively controlling hedgehog expression.


Assuntos
DNA Helicases/fisiologia , Proteínas de Drosophila/genética , Proteínas de Drosophila/fisiologia , Drosophila melanogaster/crescimento & desenvolvimento , Proteínas Hedgehog/genética , Proteínas de Homeodomínio/fisiologia , Animais , Proteínas de Ligação a DNA/fisiologia , Receptores ErbB/fisiologia , Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Fator de Transcrição PAX2/genética , Fenótipo , Transdução de Sinais , Fatores de Transcrição/fisiologia , Asas de Animais/embriologia , Xenopus/metabolismo
20.
PLoS Genet ; 6(9): e1001126, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20862326

RESUMO

Common genetic variation at human 8q23.3 is significantly associated with colorectal cancer (CRC) risk. To elucidate the basis of this association we compared the frequency of common variants at 8q23.3 in 1,964 CRC cases and 2,081 healthy controls. Reporter gene studies showed that the single nucleotide polymorphism rs16888589 acts as an allele-specific transcriptional repressor. Chromosome conformation capture (3C) analysis demonstrated that the genomic region harboring rs16888589 interacts with the promoter of gene for eukaryotic translation initiation factor 3, subunit H (EIF3H). We show that increased expression of EIF3H gene increases CRC growth and invasiveness thereby providing a biological mechanism for the 8q23.3 association. These data provide evidence for a functional basis for the non-coding risk variant rs16888589 at 8q23.3 and provides novel insight into the etiological basis of CRC.


Assuntos
Alelos , Cromossomos Humanos Par 8/genética , Neoplasias Colorretais/genética , Fator de Iniciação 3 em Eucariotos/genética , Predisposição Genética para Doença , Variação Genética , Sequências Reguladoras de Ácido Nucleico/genética , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/patologia , Ensaio de Desvio de Mobilidade Eletroforética , Feminino , Regulação Neoplásica da Expressão Gênica , Genes Reporter/genética , Loci Gênicos/genética , Humanos , Desequilíbrio de Ligação/genética , Masculino , Pessoa de Meia-Idade , Polimorfismo de Nucleotídeo Único/genética , Ligação Proteica , Fatores de Risco
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA