Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
EMBO J ; 40(3): e104569, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33300180

RESUMO

Post-transcriptional repression of gene expression by miRNAs occurs through transcript destabilization or translation inhibition. mRNA decay is known to account for most miRNA-dependent repression. However, because transcript decay occurs co-translationally, whether target translation is a requirement for miRNA-dependent transcript destabilization remains unknown. To decouple these two molecular processes, we used cytosolic long noncoding RNAs (lncRNAs) as models for endogenous transcripts that are not translated. We show that, despite interacting with the miRNA-loaded RNA-induced silencing complex, the steady-state abundance and decay rates of these transcripts are minimally affected by miRNA loss. To further validate the apparent requirement of translation for miRNA-dependent decay, we fused two lncRNA candidates to the 3'-end of a protein-coding gene reporter and found this results in their miRNA-dependent destabilization. Further analysis revealed that the few natural lncRNAs whose levels are regulated by miRNAs in mESCs tend to associate with translating ribosomes, and possibly represent misannotated micropeptides, further substantiating the necessity of target translation for miRNA-dependent transcript decay. In summary, our analyses suggest that translation is required for miRNA-dependent transcript destabilization, and demonstrate that the levels of coding and noncoding transcripts are differently affected by miRNAs.


Assuntos
MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Mensageiro/química , RNA Mensageiro/metabolismo , Animais , Fusão Gênica Artificial , Linhagem Celular , Regulação da Expressão Gênica , Genes Reporter , Sequenciamento de Nucleotídeos em Larga Escala , Camundongos , Células-Tronco Embrionárias Murinas/citologia , Células-Tronco Embrionárias Murinas/metabolismo , Biossíntese de Proteínas , Estabilidade de RNA , Ribossomos/metabolismo , Análise de Sequência de RNA
2.
Kidney Int ; 106(1): 85-97, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38431215

RESUMO

Despite the recent advances in our understanding of the role of lipids, metabolites, and related enzymes in mediating kidney injury, there is limited integrated multi-omics data identifying potential metabolic pathways driving impaired kidney function. The limited availability of kidney biopsies from living donors with acute kidney injury has remained a major constraint. Here, we validated the use of deceased transplant donor kidneys as a good model to study acute kidney injury in humans and characterized these kidneys using imaging and multi-omics approaches. We noted consistent changes in kidney injury and inflammatory markers in donors with reduced kidney function. Neighborhood and correlation analyses of imaging mass cytometry data showed that subsets of kidney cells (proximal tubular cells and fibroblasts) are associated with the expression profile of kidney immune cells, potentially linking these cells to kidney inflammation. Integrated transcriptomic and metabolomic analysis of human kidneys showed that kidney arachidonic acid metabolism and seven other metabolic pathways were upregulated following diminished kidney function. To validate the arachidonic acid pathway in impaired kidney function we demonstrated increased levels of cytosolic phospholipase A2 protein and related lipid mediators (prostaglandin E2) in the injured kidneys. Further, inhibition of cytosolic phospholipase A2 reduced injury and inflammation in human kidney proximal tubular epithelial cells in vitro. Thus, our study identified cell types and metabolic pathways that may be critical for controlling inflammation associated with impaired kidney function in humans.


Assuntos
Injúria Renal Aguda , Fenótipo , Humanos , Injúria Renal Aguda/metabolismo , Injúria Renal Aguda/patologia , Injúria Renal Aguda/etiologia , Masculino , Pessoa de Meia-Idade , Metabolômica/métodos , Feminino , Transplante de Rim/efeitos adversos , Adulto , Citometria por Imagem/métodos , Rim/patologia , Rim/metabolismo , Fosfolipases A2/metabolismo , Ácido Araquidônico/metabolismo , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/patologia , Transcriptoma , Dinoprostona/metabolismo , Dinoprostona/análise , Fibroblastos/metabolismo , Perfilação da Expressão Gênica , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Biópsia , Multiômica
3.
PLoS Genet ; 15(12): e1008501, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31881017

RESUMO

The MITF and SOX10 transcription factors regulate the expression of genes important for melanoma proliferation, invasion and metastasis. Despite growing evidence of the contribution of long noncoding RNAs (lncRNAs) in cancer, including melanoma, their functions within MITF-SOX10 transcriptional programmes remain poorly investigated. Here we identify 245 candidate melanoma associated lncRNAs whose loci are co-occupied by MITF-SOX10 and that are enriched at active enhancer-like regions. Our work suggests that one of these, Disrupted In Renal Carcinoma 3 (DIRC3), may be a clinically important MITF-SOX10 regulated tumour suppressor. DIRC3 depletion in human melanoma cells leads to increased anchorage-independent growth, a hallmark of malignant transformation, whilst melanoma patients classified by low DIRC3 expression have decreased survival. DIRC3 is a nuclear lncRNA that activates expression of its neighbouring IGFBP5 tumour suppressor through modulating chromatin structure and suppressing SOX10 binding to putative regulatory elements within the DIRC3 locus. In turn, DIRC3 dependent regulation of IGFBP5 impacts the expression of genes involved in cancer associated processes and is needed for DIRC3 control of anchorage-independent growth. Our work indicates that lncRNA components of MITF-SOX10 networks are an important new class of melanoma regulators and candidate therapeutic targets that can act not only as downstream mediators of MITF-SOX10 function but as feedback regulators of MITF-SOX10 activity.


Assuntos
Proteína 5 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Melanoma/genética , Fator de Transcrição Associado à Microftalmia/genética , RNA Longo não Codificante/genética , Fatores de Transcrição SOXE/genética , Linhagem Celular Tumoral , Núcleo Celular/genética , Proliferação de Células , Regulação para Baixo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Prognóstico , Análise de Sequência de RNA , Análise de Sobrevida
4.
Genome Res ; 25(5): 655-66, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25792609

RESUMO

Recently, a handful of intergenic long noncoding RNAs (lncRNAs) have been shown to compete with mRNAs for binding to miRNAs and to contribute to development and disease. Beyond these reports, little is yet known of the extent and functional consequences of miRNA-mediated regulation of mRNA levels by lncRNAs. To gain further insight into lncRNA-mRNA miRNA-mediated crosstalk, we reanalyzed transcriptome-wide changes induced by the targeted knockdown of over 100 lncRNA transcripts in mouse embryonic stem cells (mESCs). We predicted that, on average, almost one-fifth of the transcript level changes induced by lncRNAs are dependent on miRNAs that are highly abundant in mESCs. We validated these findings experimentally by temporally profiling transcriptome-wide changes in gene expression following the loss of miRNA biogenesis in mESCs. Following the depletion of miRNAs, we found that >50% of lncRNAs and their miRNA-dependent mRNA targets were up-regulated coordinately, consistent with their interaction being miRNA-mediated. These lncRNAs are preferentially located in the cytoplasm, and the response elements for miRNAs they share with their targets have been preserved in mammals by purifying selection. Lastly, miRNA-dependent mRNA targets of each lncRNA tended to share common biological functions. Post-transcriptional miRNA-mediated crosstalk between lncRNAs and mRNA, in mESCs, is thus surprisingly prevalent, conserved in mammals, and likely to contribute to critical developmental processes.


Assuntos
Células-Tronco Embrionárias/metabolismo , Regulação da Expressão Gênica no Desenvolvimento , MicroRNAs/genética , RNA Longo não Codificante/genética , RNA Mensageiro/genética , Animais , Células Cultivadas , Camundongos , Processamento Pós-Transcricional do RNA , Transcriptoma
5.
Bioessays ; 38(3): 295-301, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26835602

RESUMO

Recently, transcriptome-wide sequencing data have revealed the pervasiveness of intergenic long noncoding RNA (lncRNA) transcription. Subsets of lncRNAs have been demonstrated to crosstalk with and post-transcriptionally regulate mRNAs in a microRNA (miRNA)-dependent manner. Referred to as long noncoding competitive endogenous RNAs (lnceRNAs), these transcripts can contribute to diverse aspects of organismal and cellular biology, likely by providing a hitherto unrecognized layer of gene expression regulation. Here, we discuss the biological relevance of post-transcriptional regulation by lnceRNAs, provide insights on recent advances in the understanding of lnceRNA regulatory networks, and speculate on molecular factors that facilitate miRNA-dependent crosstalk between lnceRNAs and other transcripts.


Assuntos
MicroRNAs/fisiologia , Animais , Humanos , Interferência de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcrição Gênica
6.
Nat Commun ; 15(1): 5980, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39013948

RESUMO

Hyperpolarised magnetic resonance imaging (HP-13C-MRI) has shown promise as a clinical tool for detecting and characterising prostate cancer. Here we use a range of spatially resolved histological techniques to identify the biological mechanisms underpinning differential [1-13C]lactate labelling between benign and malignant prostate, as well as in tumours containing cribriform and non-cribriform Gleason pattern 4 disease. Here we show that elevated hyperpolarised [1-13C]lactate signal in prostate cancer compared to the benign prostate is primarily driven by increased tumour epithelial cell density and vascularity, rather than differences in epithelial lactate concentration between tumour and normal. We also demonstrate that some tumours of the cribriform subtype may lack [1-13C]lactate labelling, which is explained by lower epithelial lactate dehydrogenase expression, higher mitochondrial pyruvate carrier density, and increased lipid abundance compared to lactate-rich non-cribriform lesions. These findings highlight the potential of combining spatial metabolic imaging tools across scales to identify clinically significant metabolic phenotypes in prostate cancer.


Assuntos
Ácido Láctico , Imageamento por Ressonância Magnética , Fenótipo , Neoplasias da Próstata , Masculino , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/patologia , Humanos , Ácido Láctico/metabolismo , Imageamento por Ressonância Magnética/métodos , Próstata/diagnóstico por imagem , Próstata/metabolismo , Próstata/patologia , Isótopos de Carbono , Gradação de Tumores , Mitocôndrias/metabolismo , L-Lactato Desidrogenase/metabolismo
8.
Life Sci Alliance ; 3(4)2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32086317

RESUMO

Transcription is common at active mammalian enhancers sometimes giving rise to stable enhancer-associated long intergenic noncoding RNAs (elincRNAs). Expression of elincRNA is associated with changes in neighboring gene product abundance and local chromosomal topology, suggesting that transcription at these loci contributes to gene expression regulation in cis Despite the lack of evidence supporting sequence-dependent functions for most elincRNAs, splicing of these transcripts is unexpectedly common. Whether elincRNA splicing is a mere consequence of cognate enhancer activity or if it directly impacts enhancer function remains unresolved. Here, we investigate the association between elincRNA splicing and enhancer activity in mouse embryonic stem cells. We show that multi-exonic elincRNAs are enriched at conserved enhancers, and the efficient processing of elincRNAs is strongly associated with their cognate enhancer activity. This association is supported by their enrichment in enhancer-specific chromatin signatures; elevated binding of co-transcriptional regulators; increased local intra-chromosomal DNA contacts; and strengthened cis-regulation on target gene expression. Our results support the role of efficient RNA processing of enhancer-associated transcripts to cognate enhancer activity.


Assuntos
Splicing de RNA/genética , RNA Longo não Codificante/genética , Sequências Reguladoras de Ácido Nucleico/genética , Animais , Cromatina/genética , Cromossomos/metabolismo , Éxons/genética , Regulação da Expressão Gênica/genética , Camundongos , Células-Tronco Embrionárias Murinas/metabolismo , Regiões Promotoras Genéticas/genética , Splicing de RNA/fisiologia , RNA Longo não Codificante/metabolismo , Sequências Reguladoras de Ácido Nucleico/fisiologia
9.
Sci Rep ; 10(1): 8354, 2020 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-32415214

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

10.
Sci Rep ; 10(1): 6413, 2020 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-32286361

RESUMO

Long non-coding RNAs (lncRNAs) contribute to diverse cellular functions and the dysregulation of their expression or function can contribute to diseases, including diabetes. The contributions of lncRNAs to ß-cell development, function and survival has been extensively studied in vitro. However, very little is currently known on the in vivo roles of lncRNAs in the regulation of glucose and insulin homeostasis. Here we investigated the impact of loss-of-function in mice of the lncRNA A830019P07Rik, hereafter P07Rik, which was previously reported to be associated with reduced plasma insulin levels. Compared with wild-type littermates, male and female P07Rik mutant mice did not show any defect in glycaemia and plasma insulin levels in both fed and fasted state. Furthermore, P07Rik mutant mice displayed similar glucose and insulin levels in response to an intra-peritoneal glucose tolerance test. Ex vivo, islets from mutant P07Rik released similar amount of insulin in response to increased glucose concentration as wildtype littermates. In contrast with previous reports, our characterization of P07Rik mouse mutants revealed that loss of function of this lncRNA does not affect glucose and insulin homeostasis in mice.


Assuntos
Secreção de Insulina/genética , Insulina/metabolismo , RNA Longo não Codificante/metabolismo , Animais , Glicemia/metabolismo , Peso Corporal , Sequência Conservada/genética , Diabetes Mellitus Experimental/sangue , Diabetes Mellitus Experimental/genética , Regulação para Baixo/genética , Jejum/sangue , Comportamento Alimentar , Feminino , Homeostase , Insulina/sangue , Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos Obesos , RNA Longo não Codificante/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
11.
Sci Adv ; 6(27): eaaz4012, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32656337

RESUMO

Expanded CAG/CTG repeats underlie 13 neurological disorders, including myotonic dystrophy type 1 (DM1) and Huntington's disease (HD). Upon expansion, disease loci acquire heterochromatic characteristics, which may provoke changes to chromatin conformation and thereby affect both gene expression and repeat instability. Here, we tested this hypothesis by performing 4C sequencing at the DMPK and HTT loci from DM1 and HD-derived cells. We find that allele sizes ranging from 15 to 1700 repeats displayed similar chromatin interaction profiles. This was true for both loci and for alleles with different DNA methylation levels and CTCF binding. Moreover, the ectopic insertion of an expanded CAG repeat tract did not change the conformation of the surrounding chromatin. We conclude that CAG/CTG repeat expansions are not enough to alter chromatin conformation in cis. Therefore, it is unlikely that changes in chromatin interactions drive repeat instability or changes in gene expression in these disorders.

12.
Elife ; 82019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-31045494

RESUMO

To generate energy efficiently, the cell is uniquely challenged to co-ordinate the abundance of electron transport chain protein subunits expressed from both nuclear and mitochondrial genomes. How an effective stoichiometry of this many constituent subunits is co-ordinated post-transcriptionally remains poorly understood. Here we show that Cerox1, an unusually abundant cytoplasmic long noncoding RNA (lncRNA), modulates the levels of mitochondrial complex I subunit transcripts in a manner that requires binding to microRNA-488-3p. Increased abundance of Cerox1 cooperatively elevates complex I subunit protein abundance and enzymatic activity, decreases reactive oxygen species production, and protects against the complex I inhibitor rotenone. Cerox1 function is conserved across placental mammals: human and mouse orthologues effectively modulate complex I enzymatic activity in mouse and human cells, respectively. Cerox1 is the first lncRNA demonstrated, to our knowledge, to regulate mitochondrial oxidative phosphorylation and, with miR-488-3p, represent novel targets for the modulation of complex I activity.


Assuntos
Complexo I de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica , Mitocôndrias/enzimologia , RNA Longo não Codificante/metabolismo , Animais , Linhagem Celular , Complexo I de Transporte de Elétrons/genética , Perfilação da Expressão Gênica , Humanos , Camundongos , MicroRNAs/metabolismo
13.
Adv Genet ; 85: 149-99, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24880735

RESUMO

Endogenously expressed transcripts that are posttranscriptionally regulated by the same microRNAs (miRNAs) will, in principle, compete for the binding of their shared small noncoding RNA regulators and modulate each other's abundance. Recently, the levels of some coding as well as noncoding transcripts have indeed been found to be regulated in this way. Transcripts that engage in such regulatory interactions are referred to as competitive endogenous RNAs (ceRNAs). This novel layer of posttranscriptional regulation has been shown to contribute to diverse aspects of organismal and cellular biology, despite the number of functionally characterized ceRNAs being as yet relatively low. Importantly, increasing evidence suggests that the dysregulation of some ceRNA interactions is associated with disease etiology, most preeminently with cancer. Here we review how posttranscriptional regulation by miRNAs contributes to the cross-talk between transcripts and review examples of known ceRNAs by highlighting the features underlying their interactions and what might be their biological relevance.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/metabolismo , Precursores de RNA/genética , Animais , Humanos , RNA Longo não Codificante/genética
15.
Nat Struct Mol Biol ; 21(11): 955-961, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25306109

RESUMO

What causes the tissue-specific pathology of diseases resulting from mutations in housekeeping genes? Specifically, in spinocerebellar ataxia type 7 (SCA7), a neurodegenerative disorder caused by a CAG-repeat expansion in ATXN7 (which encodes an essential component of the mammalian transcription coactivation complex, STAGA), the factors underlying the characteristic progressive cerebellar and retinal degeneration in patients were unknown. We found that STAGA is required for the transcription initiation of miR-124, which in turn mediates the post-transcriptional cross-talk between lnc-SCA7, a conserved long noncoding RNA, and ATXN7 mRNA. In SCA7, mutations in ATXN7 disrupt these regulatory interactions and result in a neuron-specific increase in ATXN7 expression. Strikingly, in mice this increase is most prominent in the SCA7 disease-relevant tissues, namely the retina and cerebellum. Our results illustrate how noncoding RNA-mediated feedback regulation of a ubiquitously expressed housekeeping gene may contribute to specific neurodegeneration.


Assuntos
Cerebelo/metabolismo , MicroRNAs/genética , Proteínas do Tecido Nervoso/genética , RNA Longo não Codificante/genética , Retina/metabolismo , Ataxias Espinocerebelares/genética , Animais , Ataxina-7 , Linhagem Celular Tumoral , Cerebelo/patologia , Retroalimentação Fisiológica , Feminino , Fibroblastos/metabolismo , Fibroblastos/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Masculino , Camundongos , MicroRNAs/metabolismo , Mutação , Proteínas do Tecido Nervoso/metabolismo , Neurônios/metabolismo , Neurônios/patologia , RNA Longo não Codificante/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Retina/patologia , Transdução de Sinais , Ataxias Espinocerebelares/metabolismo , Ataxias Espinocerebelares/patologia , Iniciação da Transcrição Genética
17.
Mol Diagn Ther ; 11(3): 155-60, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17570737

RESUMO

BACKGROUND: Depression is thought to involve, in part, dysregulation of serotonergic neurotransmission. In depressed individuals, the number of serotonin receptors, including the 5-hydroxytryptamine (serotonin)-1A (5-HT(1A)) autoreceptors, are increased. Clinical improvement with selective serotonin reuptake inhibitors (SSRIs) is not usually observed until several weeks after treatment initiation. This delay may be due to the time it takes for the autoreceptors to downregulate. Roughly one-third of patients with depression do not respond to an initial trial of antidepressant medication treatment, possibly as a result of structural variations in the 5-HT(1A) receptor. AIMS: This study was designed to determine the allelic frequency of seven 5-HT(1A) receptor polymorphisms in a depressed versus a nondepressed population, and in SSRI responders versus nonresponders. All the polymorphisms studied are single nucleotide polymorphisms (SNPs) in the HTR1A gene, which encodes 5-HT(1A). Seven prevalent SNPs were included in the analysis. RESULTS: The study showed no relationship between any of the HTR1A polymorphisms and SSRI responders versus nonresponders. CONCLUSION: While the study has several limitations, the results are consistent with a growing body of literature that suggests that the pharmacogenetics of depression (an inherently complex disorder) may turn out to be multifactorial, and may include the HTR1A gene in concert with other serotonin-related genes.


Assuntos
Antidepressivos/uso terapêutico , Depressão/tratamento farmacológico , Polimorfismo de Nucleotídeo Único , Receptor 5-HT1A de Serotonina/genética , Inibidores Seletivos de Recaptação de Serotonina/uso terapêutico , Adulto , Idoso , Idoso de 80 Anos ou mais , Depressão/genética , Resistência a Medicamentos , Humanos , Pessoa de Meia-Idade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA