Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
Am J Physiol Gastrointest Liver Physiol ; 325(2): G147-G157, 2023 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-37129245

RESUMO

Although midnolin has been studied for over 20 years, its biological roles in vivo remain largely unknown, especially due to the lack of a functional animal model. Indeed, given our recent discovery that the knockdown of midnolin suppresses liver cancer cell tumorigenicity and that this antitumorigenic effect is associated with modulation of lipid metabolism, we hypothesized that knockout of midnolin in vivo could potentially protect from nonalcoholic fatty liver disease (NAFLD) which has become the most common cause of chronic liver disease in the Western world. Accordingly, in the present study, we have developed and now report on the first functional global midnolin knockout mouse model. Although the overwhelming majority of global homozygous midnolin knockout mice demonstrated embryonic lethality, heterozygous knockout mice were observed to be similar to wild-type mice in their viability and were used to determine the effect of reduced midnolin expression on NAFLD. We found that global heterozygous midnolin knockout attenuated the severity of NAFLD in mice fed a Western-style diet, high in fat, cholesterol, and fructose, and this attenuation in disease was associated with significantly reduced levels of large lipid droplets, hepatic free cholesterol, and serum LDL, with significantly differential gene expression involved in cholesterol/lipid metabolism. Collectively, our results support a role for midnolin in regulating cholesterol/lipid metabolism in the liver. Thus, midnolin may represent a novel therapeutic target for NAFLD. Finally, our observation that midnolin was essential for survival underscores the broad importance of this gene beyond its role in liver biology.NEW & NOTEWORTHY We have developed and now report on the first functional global midnolin knockout mouse model. We found that global heterozygous midnolin knockout attenuated the severity of nonalcoholic fatty liver disease (NAFLD) in mice fed a Western-style diet, high in fat, cholesterol, and fructose, and this attenuation in disease was associated with significantly reduced levels of large lipid droplets, hepatic free cholesterol, and serum LDL, with significantly differential gene expression involved in cholesterol/lipid metabolism.


Assuntos
Hepatopatia Gordurosa não Alcoólica , Camundongos , Animais , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Frutose/metabolismo , Dieta Hiperlipídica/métodos , Fígado/metabolismo , Colesterol/metabolismo , Camundongos Knockout , Modelos Animais de Doenças , Camundongos Endogâmicos C57BL
2.
J Clin Invest ; 133(3)2023 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-36512424

RESUMO

Impaired angiogenesis in diabetes is a key process contributing to ischemic diseases such as peripheral arterial disease. Epigenetic mechanisms, including those mediated by long noncoding RNAs (lncRNAs), are crucial links connecting diabetes and the related chronic tissue ischemia. Here we identify the lncRNA that enhances endothelial nitric oxide synthase (eNOS) expression (LEENE) as a regulator of angiogenesis and ischemic response. LEENE expression was decreased in diabetic conditions in cultured endothelial cells (ECs), mouse hind limb muscles, and human arteries. Inhibition of LEENE in human microvascular ECs reduced their angiogenic capacity with a dysregulated angiogenic gene program. Diabetic mice deficient in Leene demonstrated impaired angiogenesis and perfusion following hind limb ischemia. Importantly, overexpression of human LEENE rescued the impaired ischemic response in Leene-knockout mice at tissue functional and single-cell transcriptomic levels. Mechanistically, LEENE RNA promoted transcription of proangiogenic genes in ECs, such as KDR (encoding VEGFR2) and NOS3 (encoding eNOS), potentially by interacting with LEO1, a key component of the RNA polymerase II-associated factor complex and MYC, a crucial transcription factor for angiogenesis. Taken together, our findings demonstrate an essential role for LEENE in the regulation of angiogenesis and tissue perfusion. Functional enhancement of LEENE to restore angiogenesis for tissue repair and regeneration may represent a potential strategy to tackle ischemic vascular diseases.


Assuntos
Diabetes Mellitus Experimental , RNA Longo não Codificante , Humanos , Camundongos , Animais , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , Células Endoteliais/metabolismo , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/metabolismo , Músculo Esquelético/metabolismo , Neovascularização Fisiológica/genética , Isquemia/genética , Isquemia/metabolismo , Camundongos Knockout , Membro Posterior , Camundongos Endogâmicos C57BL
3.
BMC Nephrol ; 23(1): 227, 2022 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-35761198

RESUMO

Nephrolithiasis is not common in children, but the incidence is gradually increased in these years. Urinary tract malformations, urinary infection, dietary habits, geographic region and genetic factor are involved in the etiology of nephrolithiasis. For the affected child, it is especially important to elucidate the etiology, which may provide an accurate diagnosis, a personalized therapy and effective follow-up strategy. Here to seek the etiology of a ten-year-old boy incidentally found with nephrolithiasis, next generation sequencing (NGS) including a panel with 248 genes involved in hereditary kidney diseases was performed for the boy and identified two mutations of KCNJ1, c.89G > A (p.C30Y) and c.65G > T (p.R22M), and the later was a novel missense mutation originated from his father. The child was confirmed with type II Bartter syndrome (BS) caused by KCNJ1 mutations. Our study suggests that BS may be difficult to get diagnosed at an early stage based on clinical manifestations or biochemical laboratory tests, and NGS is an efficient way to determine the etiology and provide further treatment and guide fertility counseling for the affected family.


Assuntos
Síndrome de Bartter , Cálculos Renais , Canais de Potássio Corretores do Fluxo de Internalização , Criança , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Mutação , Canais de Potássio Corretores do Fluxo de Internalização/genética
4.
Endocrinology ; 163(1)2022 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-34902009

RESUMO

Concordant transcriptional regulation can generate multiple gene products that collaborate to achieve a common goal. Here we report a case of concordant transcriptional regulation that instead drives a single protein to be produced in the same cell type from divergent promoters. This gene product-the RHOX5 homeobox transcription factor-is translated from 2 different mRNAs with different 5' untranslated regions (UTRs) transcribed from alternative promoters. Despite the fact that these 2 promoters-the proximal promoter (Pp) and the distal promoter (Pd)-exhibit different patterns of tissue-specific activity, share no obvious sequence identity, and depend on distinct transcription factors for expression, they exhibit a remarkably similar expression pattern in the testes. In particular, both depend on androgen signaling for expression in the testes, where they are specifically expressed in Sertoli cells and have a similar stage-specific expression pattern during the seminiferous epithelial cycle. We report evidence for 3 mechanisms that collaborate to drive concordant Pp/Pd expression. First, both promoters have an intrinsic ability to respond to androgen receptor and androgen. Second, the Pp acts as an enhancer to promote androgen-dependent transcription from the Pd. Third, Pd transcription is positively autoregulated by the RHOX5 protein, which is first produced developmentally from the Pp. Together, our data support a model in which the Rhox5 homeobox gene evolved multiple mechanisms to activate both of its promoters in Sertoli cells to produce Rhox5 in an androgen-dependent manner during different phases of spermatogenesis.


Assuntos
Androgênios/metabolismo , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Regiões Promotoras Genéticas , Células de Sertoli/metabolismo , Fatores de Transcrição/genética , Regiões 5' não Traduzidas , Animais , Metilação de DNA , Genes Homeobox , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plasmídeos/metabolismo , Isoformas de Proteínas , Receptores Androgênicos/metabolismo , Túbulos Seminíferos/metabolismo , Espermatogênese , Testículo/metabolismo , Fatores de Transcrição/metabolismo
5.
Cancers (Basel) ; 13(23)2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34884992

RESUMO

The integration of cellular status with metabolism is critically important and the coupling of energy production and cellular function is highly evolutionarily conserved. This has been demonstrated in stem cell biology, organismal, cellular and tissue differentiation and in immune cell biology. However, a molecular mechanism delineating how cells coordinate and couple metabolism with transcription as they navigate quiescence, growth, proliferation, differentiation and migration remains in its infancy. The extreme N-termini of the Kat3 coactivator family members, CBP and p300, by far the least homologous regions with only 66% identity, interact with members of the nuclear receptor family, interferon activated Stat1 and transcriptionally competent ß-catenin, a critical component of the Wnt signaling pathway. We now wish to report based on multiomic and functional investigations, utilizing p300 knockdown, N-terminal p300 edited and p300 S89A edited cell lines and p300 S89A knockin mice, that the N-termini of the Kat3 coactivators provide a highly evolutionarily conserved hub to integrate multiple signaling cascades to coordinate cellular metabolism with the regulation of cellular status and function.

6.
Haematologica ; 106(9): 2427-2438, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-32732362

RESUMO

Extra-nodal natural killer/T-cell lymphoma, nasal type (ENKTCL) is a highly aggressive lymphoma, where the tumor suppressor gene (TSG) PRDM1 is frequently lost/inactivated. We employed two different CRISPR/Cas9 approaches to generate PRDM1-/- primary NK cells to study its role in NK-cell homeostasis. PRDM1-/- NK cells showed a marked increase in cloning efficiency, higher proliferation rate and less apoptosis compared with their wild type counterparts. Gene expression profiling demonstrated a marked enrichment in pathways associated with proliferation, cell cycle, MYC, MYB and TCR/NK signaling in PRDM1-/- NK cells, but pathways associated with normal cellular functions including cytotoxic functions were down-regulated, suggesting that the loss of PRDM1 shifted NK cells toward proliferation and survival rather than the performance of its normal functions. We were also able to further modify a PRDM1 deleted clone to introduce heterozygous deletions of common TSG in ENKTCL such as TP53, DDX3X, or PTPN6. We have established an in vitro model to elucidate the major pathways through which PRDM1 mediates its homeostatic control of NK-cells. This approach can be applied to the study of other relevant genetic lesions and oncogenic collaborations in lymphoma pathogenesis.


Assuntos
Carcinogênese , Regulação Neoplásica da Expressão Gênica , Células Matadoras Naturais , Linfoma Extranodal de Células T-NK/genética , Fator 1 de Ligação ao Domínio I Regulador Positivo , Linhagem Celular Tumoral , Perfilação da Expressão Gênica , Humanos , Fator 1 de Ligação ao Domínio I Regulador Positivo/genética
7.
Nat Commun ; 9(1): 292, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29348663

RESUMO

The optimal expression of endothelial nitric oxide synthase (eNOS), the hallmark of endothelial homeostasis, is vital to vascular function. Dynamically regulated by various stimuli, eNOS expression is modulated at transcriptional, post-transcriptional, and post-translational levels. However, epigenetic modulations of eNOS, particularly through long non-coding RNAs (lncRNAs) and chromatin remodeling, remain to be explored. Here we identify an enhancer-associated lncRNA that enhances eNOS expression (LEENE). Combining RNA-sequencing and chromatin conformation capture methods, we demonstrate that LEENE is co-regulated with eNOS and that its enhancer resides in proximity to eNOS promoter in endothelial cells (ECs). Gain- and Loss-of-function of LEENE differentially regulate eNOS expression and EC function. Mechanistically, LEENE facilitates the recruitment of RNA Pol II to the eNOS promoter to enhance eNOS nascent RNA transcription. Our findings unravel a new layer in eNOS regulation and provide novel insights into cardiovascular regulation involving endothelial function.


Assuntos
Células Endoteliais/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação Enzimológica da Expressão Gênica , Óxido Nítrico Sintase Tipo III/genética , RNA Longo não Codificante/genética , Animais , Células Cultivadas , Perfilação da Expressão Gênica , Humanos , Masculino , Camundongos Endogâmicos C57BL , Óxido Nítrico Sintase Tipo III/metabolismo , Regiões Promotoras Genéticas/genética , RNA Polimerase II/metabolismo , Transcrição Gênica
8.
Stem Cell Reports ; 6(6): 844-857, 2016 06 14.
Artigo em Inglês | MEDLINE | ID: mdl-27304915

RESUMO

Nonsense-mediated RNA decay (NMD) is a highly conserved pathway that selectively degrades specific subsets of RNA transcripts. Here, we provide evidence that NMD regulates early human developmental cell fate. We found that NMD factors tend to be expressed at higher levels in human pluripotent cells than in differentiated cells, raising the possibility that NMD must be downregulated to permit differentiation. Loss- and gain-of-function experiments in human embryonic stem cells (hESCs) demonstrated that, indeed, NMD downregulation is essential for efficient generation of definitive endoderm. RNA-seq analysis identified NMD target transcripts induced when NMD is suppressed in hESCs, including many encoding signaling components. This led us to test the role of TGF-ß and BMP signaling, which we found NMD acts through to influence definitive endoderm versus mesoderm fate. Our results suggest that selective RNA decay is critical for specifying the developmental fate of specific human embryonic cell lineages.


Assuntos
Linhagem da Célula/genética , Endoderma/metabolismo , Células-Tronco Embrionárias Humanas/metabolismo , Degradação do RNAm Mediada por Códon sem Sentido , Células-Tronco Pluripotentes/metabolismo , RNA Mensageiro/genética , Proteína Morfogenética Óssea 4/genética , Proteína Morfogenética Óssea 4/metabolismo , Diferenciação Celular , Linhagem Celular , Ectoderma/citologia , Ectoderma/metabolismo , Endoderma/citologia , Perfilação da Expressão Gênica , Células-Tronco Embrionárias Humanas/citologia , Humanos , Mesoderma/citologia , Mesoderma/metabolismo , Células-Tronco Pluripotentes/citologia , RNA Helicases , RNA Mensageiro/metabolismo , Análise de Sequência de RNA , Transdução de Sinais , Transativadores , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
9.
Cell ; 165(2): 382-95, 2016 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-27040500

RESUMO

Gene duplication is a major evolutionary force driving adaptation and speciation, as it allows for the acquisition of new functions and can augment or diversify existing functions. Here, we report a gene duplication event that yielded another outcome--the generation of antagonistic functions. One product of this duplication event--UPF3B--is critical for the nonsense-mediated RNA decay (NMD) pathway, while its autosomal counterpart--UPF3A--encodes an enigmatic protein previously shown to have trace NMD activity. Using loss-of-function approaches in vitro and in vivo, we discovered that UPF3A acts primarily as a potent NMD inhibitor that stabilizes hundreds of transcripts. Evidence suggests that UPF3A acquired repressor activity through simple impairment of a critical domain, a rapid mechanism that may have been widely used in evolution. Mice conditionally lacking UPF3A exhibit "hyper" NMD and display defects in embryogenesis and gametogenesis. Our results support a model in which UPF3A serves as a molecular rheostat that directs developmental events.


Assuntos
Desenvolvimento Embrionário , Genes Duplicados , Degradação do RNAm Mediada por Códon sem Sentido , Proteínas de Ligação a RNA/metabolismo , Animais , Linhagem Celular Tumoral , Evolução Molecular , Gametogênese , Células HeLa , Humanos , Camundongos
10.
Dev Biol ; 409(1): 26-38, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26548531

RESUMO

MicroRNAs (miRNAs) are known to play diverse roles in the regulation of vertebrate development. To investigate miRNA-target mRNA relationships in embryonic development, we have carried out small-RNA sequencing to identify miRNAs expressed in the early gastrula of Xenopus laevis. We identify a total of 180 miRNAs, and we have identified the locations of the miRNA precursor sequences in the X. laevis genome. Of these miRNAs, 141 represent miRs previously identified in Xenopus tropicalis. Alignment to human miRNAs led to the identification of 39 miRNAs that have not previously been described for Xenopus. We have also used a biochemical approach to isolate mRNAs that are associated with the RNA-Induced Silencing Complex (RISC) in early gastrulae and thus candidate targets of miRNA-dependent regulation. Interrogation of this RISC-associated mRNA pool by RT-PCR indicates that a number of genes essential for early patterning and specification may be under regulation by miRNAs. Smad1 transcripts are associated with the RISC; target prediction algorithms identify a single miRNA-binding site for miR-26, which is common to the 3'UTRs of Smad1a and Smad1b. Disruption of the interaction between miR-26 and the Smad1 3'UTR via a Target Protector Morpholino Oligonucleotide (TPMO) leads to a 2-fold increase in Smad1 protein accumulation, moderate increases in the expression of BMP4/Smad1 target genes, and a reduction in organizer gene expression, as well as a partially ventralized phenotype in approximately 25% of embryos. Overexpression of miR-26 resulted in moderately decreased expression of Smad1-dependent genes and an expansion of the region expressing the Organizer gene not1. Our findings indicate that interactions between miR-26 and the Smad1 3'UTR modulate Smad1 function in the establishment of axial patterning; they also establish a foundation for the functional analysis of miRNAs and their regulatory interactions during gastrulation.


Assuntos
Gástrula/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/metabolismo , Proteína Smad1/genética , Proteínas de Xenopus/genética , Xenopus/embriologia , Xenopus/genética , Regiões 3' não Traduzidas/genética , Animais , Proteínas Argonautas/genética , Proteínas Argonautas/metabolismo , Sequência de Bases , Embrião não Mamífero/metabolismo , Desenvolvimento Embrionário/genética , Gástrula/embriologia , Imunoprecipitação , MicroRNAs/genética , Dados de Sequência Molecular , Fenótipo , Ligação Proteica/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reprodutibilidade dos Testes , Proteína Smad1/metabolismo , Proteínas de Xenopus/metabolismo
12.
EMBO Rep ; 16(5): 599-609, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25807986

RESUMO

Endoplasmic reticulum (ER) stress induces the unfolded protein response (UPR), an essential adaptive intracellular pathway that relieves the stress. Although the UPR is an evolutionarily conserved and beneficial pathway, its chronic activation contributes to the pathogenesis of a wide variety of human disorders. The fidelity of UPR activation must thus be tightly regulated to prevent inappropriate signaling. The nonsense-mediated RNA decay (NMD) pathway has long been known to function in RNA quality control, rapidly degrading aberrant mRNAs, and has been suggested to regulate subsets of normal mRNAs. Here, we report that the NMD pathway regulates the UPR. NMD increases the threshold for triggering the UPR in vitro and in vivo, thereby preventing UPR activation in response to normally innocuous levels of ER stress. NMD also promotes the timely termination of the UPR. We demonstrate that NMD directly targets the mRNAs encoding several UPR components, including the highly conserved UPR sensor, IRE1α, whose NMD-dependent degradation partly underpins this process. Our work not only sheds light on UPR regulation, but demonstrates the physiological relevance of NMD's ability to regulate normal mRNAs.


Assuntos
Degradação do RNAm Mediada por Códon sem Sentido , Resposta a Proteínas não Dobradas/genética , Animais , Linhagem Celular , Proteínas de Ligação a DNA/genética , Estresse do Retículo Endoplasmático/genética , Endorribonucleases/metabolismo , Expressão Gênica , Regulação da Expressão Gênica , Ordem dos Genes , Vetores Genéticos/genética , Camundongos , Camundongos Knockout , Proteínas Serina-Treonina Quinases/metabolismo , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição de Fator Regulador X , Fatores de Transcrição/genética , Transcrição Gênica
13.
Differentiation ; 83(4): 210-9, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22387344

RESUMO

FGFs and BMPs act in concert to regulate a wide range of processes in vertebrate development. In most cases, FGFs and BMPs have opposing effects, and specific developmental outcomes arise out of a balance between the two growth factors. We and others have previously demonstrated that signaling pathways activated by FGFs and BMPs interact via inhibitory crosstalk. Here we demonstrate a role for the BMP effector TGF-ß Activated Kinase 1 (TAK1) in the maintenance of Smad1 activity in Xenopus embryos, via the inhibition of erk MAPK. Up- or downregulation of TAK1 levels produces an inverse alteration in the amount of activated erk MAPK. The inhibition of erk MAPK by TAK1 is mediated by p38 and a corresponding decrease in phosphorylation of MEK. TAK1 morphant embryos show a decrease in the nuclear accumulation of Smad1. Conversely, reduction of erk MAPK activity via overexpression of MAP Kinase Phosphatase1 (MKP1) leads to an increase in nuclear Smad1. Both TAK1 morphant ectoderm and ectoderm treated with FGF show a decrease in the expression of several Smad1-inducible genes. Neural-specific gene expression is inhibited in isolated ectoderm coexpressing noggin and TAK1, suggesting that TAK1 is sufficient to inhibit neural specification. Introduction of TAK1 morpholino oligonucleotide expands the expression of organizer genes, disrupts formation of the boundary between organizer and non-organizer mesoderm, and increases the spatial range of MAPK activation in response to localized FGF. Our results indicate that inhibitory interactions between FGF and BMP4 effector pathways increase the robustness of BMP signaling via a feed-forward mechanism.


Assuntos
Proteína Morfogenética Óssea 4/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/antagonistas & inibidores , Fatores de Crescimento de Fibroblastos/fisiologia , MAP Quinase Quinase Quinases/fisiologia , Transdução de Sinais , Proteína Smad1/metabolismo , Proteínas de Xenopus/metabolismo , Animais , Regulação para Baixo , Imuno-Histoquímica , Hibridização In Situ , Reação em Cadeia da Polimerase em Tempo Real , Xenopus/embriologia
14.
Mol Cell ; 43(6): 950-61, 2011 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-21925383

RESUMO

Nonsense-mediated mRNA decay (NMD) is a conserved RNA decay pathway that degrades aberrant mRNAs and directly regulates many normal mRNAs. This dual role for NMD raises the possibility that its magnitude is buffered to prevent the potentially catastrophic alterations in gene expression that would otherwise occur if NMD were perturbed by environmental or genetic insults. In support of this, here we report the existence of a negative feedback regulatory network that directly acts on seven NMD factors. Feedback regulation is conferred by different branches of the NMD pathway in a cell type-specific and developmentally regulated manner. We identify feedback-regulated NMD factors that are rate limiting for NMD and demonstrate that reversal of feedback regulation in response to NMD perturbation is crucial for maintaining NMD. Together, our results suggest the existence of an intricate feedback network that maintains both RNA surveillance and the homeostasis of normal gene expression in mammalian cells.


Assuntos
Estabilidade de RNA , RNA Mensageiro/metabolismo , Fator 3 Ativador da Transcrição/metabolismo , Western Blotting , Retroalimentação Fisiológica , Regulação da Expressão Gênica , Células HeLa , Homeostase , Humanos , RNA Helicases , Interferência de RNA , Transativadores/antagonistas & inibidores
15.
Mol Cell Biol ; 31(6): 1275-87, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21245380

RESUMO

Histone H1 is an abundant and essential component of chromatin whose precise role in regulating gene expression is poorly understood. Here, we report that a major target of H1-mediated regulation in embryonic stem (ES) cells is the X-linked Rhox homeobox gene cluster. To address the underlying mechanism, we examined the founding member of the Rhox gene cluster-Rhox5-and found that its distal promoter (Pd) loses H1, undergoes demethylation, and is transcriptionally activated in response to loss of H1 genes in ES cells. Demethylation of the Pd is required for its transcriptional induction and we identified a single cytosine in the Pd that, when methylated, is sufficient to inhibit Pd transcription. Methylation of this single cytosine prevents the Pd from binding GA-binding protein (GABP), a transcription factor essential for Pd transcription. Thus, H1 silences Rhox5 transcription by promoting methylation of one of its promoters, a mechanism likely to extend to other H1-regulated Rhox genes, based on analysis of ES cells lacking DNA methyltransferases. The Rhox cluster genes targeted for H1-mediated transcriptional repression are also subject to another DNA methylation-regulated process: Xp imprinting. Remarkably, we found that only H1-regulated Rhox genes are imprinted, not those immune to H1-mediated repression. Together, our results indicate that the Rhox gene cluster is a major target of H1-mediated transcriptional repression in ES cells and that H1 is a candidate to have a role in Xp imprinting.


Assuntos
Metilação de DNA , Células-Tronco Embrionárias/metabolismo , Impressão Genômica , Histonas/metabolismo , Proteínas de Homeodomínio/genética , Fatores de Transcrição/genética , Animais , Células Cultivadas , Citosina/metabolismo , Metilases de Modificação do DNA/metabolismo , Fator de Transcrição de Proteínas de Ligação GA/metabolismo , Deleção de Genes , Genes Homeobox , Histonas/genética , Proteínas de Homeodomínio/metabolismo , Camundongos , Família Multigênica , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo , Transcrição Gênica
16.
Dev Biol ; 339(2): 494-506, 2010 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-20060393

RESUMO

The retinal homeobox (Rx) gene product is essential for eye development. However little is known about its molecular function. It has been demonstrated that Rx binds to photoreceptor conserved element (PCE-1), a highly conserved element found in the promoter region of photoreceptor-specific genes such as rhodopsin and red cone opsin. We verify that Rx is co-expressed with rhodopsin and red cone opsin in maturing photoreceptors and demonstrate that Rx binds to the rhodopsin and red cone opsin promoters in vivo. We also find that Rx can cooperate with the Xenopus analogs of Crx and Nrl, otx5b and XLMaf (respectively), to activate a Xenopus opsin promoter-dependent reporter. Finally, we demonstrate that reduction of Rx expression in tadpoles results in decreases in expression of several PCE-1 containing photoreceptor genes, abnormal photoreceptor morphology, and impaired vision. Our data suggests that Rx, in combination with other transcription factors, is necessary for normal photoreceptor gene expression, maintenance, and function. This establishes a direct role for Rx in regulation of genes expressed in a differentiated cell type.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , Proteínas de Homeodomínio/metabolismo , Células Fotorreceptoras/metabolismo , Rodopsina/genética , Opsinas de Bastonetes/genética , Proteínas de Xenopus/genética , Animais , Embrião não Mamífero/metabolismo , Proteínas do Olho/genética , Proteínas do Olho/metabolismo , Proteínas de Homeodomínio/genética , Retinaldeído , Rodopsina/metabolismo , Opsinas de Bastonetes/metabolismo , Proteínas de Xenopus/metabolismo , Xenopus laevis
17.
Dev Dyn ; 235(7): 1895-907, 2006 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16680723

RESUMO

The novel gene ashwin was isolated in a differential display screen for genes activated or up-regulated early in neural specification. ashwin is expressed maternally and zygotically, and it is up-regulated in the neural ectoderm after the midgastrula stage. It is expressed in the neural plate and later in the embryonic brain, eyes, and spinal cord. Overexpression of ashwin in whole embryos leads to anterior truncations and other defects. However, a second Organizer does not form, and the secondary axial structures may result from splitting of the Organizer, rather than axis duplication. Morpholino oligonucleotide-mediated reduction in ashwin expression leads to lethality or abnormalities in gastrulation, as well as significant apoptosis in midgastrula embryos. Apoptosis is also observed in midgastrula embryos overexpressing ashwin. Coexpression of ashwin with the bone morphogenetic protein-4 antagonist noggin has a synergistic effect on neural-specific gene expression in isolated animal cap ectoderm. Ashwin has no previously characterized domains, although two nuclear localization signals can be identified. Orthologues have been identified in the human, mouse, chicken, and pufferfish genomes. Our results suggest that ashwin regulates cell survival and anteroposterior patterning.


Assuntos
Padronização Corporal/fisiologia , Sistema Nervoso/embriologia , Proteínas Nucleares/biossíntese , Proteínas de Xenopus/biossíntese , Sequência de Aminoácidos , Animais , Apoptose , Proteínas de Transporte/metabolismo , Sobrevivência Celular , Embrião não Mamífero/anormalidades , Embrião não Mamífero/metabolismo , Gástrula/fisiologia , Dados de Sequência Molecular , Sistema Nervoso/metabolismo , Organizadores Embrionários/embriologia , Homologia de Sequência de Aminoácidos , Regulação para Cima , Xenopus laevis
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA