Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Commun Biol ; 6(1): 145, 2023 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-36737664

RESUMO

Cancer cells experience confinement as they navigate the tumour microenvironment during metastasis. Recent studies have revealed that the nucleus can function as a 'ruler' for measuring physical confinement via membrane tension, allowing for compression-sensitive changes in migration. Cell nuclei contain many nuclear bodies that form when their components phase separate and condense within permissive local regions within the nucleus. However, how sub-nuclear organisation and phase separation changes with cell confinement and compression is largely unknown. Here we focus on paraspeckles, stress-responsive subnuclear bodies that form by phase separation around the long non-coding RNA NEAT1. As cells entered moderate confinement, a significant increase in paraspeckle number and size was observed compared to unconfined cells. Paraspeckle polarization bias towards the leading edge was also observed in confinement, correlating with regions of euchromatin. Increasing paraspeckle abundance resulted in increases in confined migration likelihood, speed, and directionality, as well as an enhancement of paraspeckle polarization towards the leading edge. This polarization of paraspeckle condensates may play a key role in regulating confined migration and invasion in cancer cells, and illustrates the utility of microchannel-based assays for identifying phenomena not observed on 2D or 3D bulk substrates.


Assuntos
Paraspeckles , RNA Longo não Codificante , Núcleo Celular/genética , RNA Longo não Codificante/genética
2.
EMBO Rep ; 24(2): e54977, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36416237

RESUMO

High-risk neuroblastoma patients have poor survival rates and require better therapeutic options. High expression of a multifunctional DNA and RNA-binding protein, NONO, in neuroblastoma is associated with poor patient outcome; however, there is little understanding of the mechanism of NONO-dependent oncogenic gene regulatory activity in neuroblastoma. Here, we used cell imaging, biochemical and genome-wide molecular analysis to reveal complex NONO-dependent regulation of gene expression. NONO forms RNA- and DNA-tethered condensates throughout the nucleus and undergoes phase separation in vitro, modulated by nucleic acid binding. CLIP analyses show that NONO mainly binds to the 5' end of pre-mRNAs and modulates pre-mRNA processing, dependent on its RNA-binding activity. NONO regulates super-enhancer-associated genes, including HAND2 and GATA2. Abrogating NONO RNA binding, or phase separation activity, results in decreased expression of HAND2 and GATA2. Thus, future development of agents that target RNA-binding activity of NONO may have therapeutic potential in this cancer context.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Proteínas de Ligação a DNA , Neuroblastoma , Humanos , DNA/metabolismo , Proteínas de Ligação a DNA/genética , Fator de Transcrição GATA2/genética , Fator de Transcrição GATA2/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Processamento Pós-Transcricional do RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Fatores de Transcrição/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo
3.
Biochimie ; 190: 124-131, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34329720

RESUMO

Paraspeckles are RNA-protein structures within the nucleus of mammalian cells, capable of orchestrating various biochemical processes. An overexpression of the architectural component of paraspeckles, a long non-coding RNA called NEAT1 (Nuclear Enriched Abundant Transcript 1), has been linked to a variety of cancers and is often associated with poor patient prognosis. Thus, there is an accumulating interest in the role of paraspeckles in carcinogenesis, however there is a limited understanding of how NEAT1 expression is regulated. Here, we demonstrate that both nuclear G-quadruplex (G4) and paraspeckle formation are significantly increased in a human breast cancer cell line compared to non-tumorigenic breast cells. Moreover, we identified and characterized G4-forming sequences within the NEAT1 promoter and demonstrate stabilization of G4 DNA with a G4-stabilizing small molecule results in a significant alteration in both paraspeckle formation and NEAT1 expression. This G4-mediated alteration of NEAT1 at both the transcriptional and post-transcriptional levels was evident in U2OS osteosarcoma cells, MCF-7 breast adenocarcinoma and MDA-MB-231 triple negative breast cancer cells.


Assuntos
Quadruplex G , Neoplasias/genética , Neoplasias/metabolismo , Paraspeckles/genética , Paraspeckles/metabolismo , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Humanos , RNA Longo não Codificante/química , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo
4.
Clin Transl Med ; 11(4): e381, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33931969

RESUMO

BACKGROUND: Caveolae proteins play diverse roles in cancer development and progression. In prostate cancer, non-caveolar caveolin-1 (CAV1) promotes metastasis, while CAVIN1 attenuates CAV1-induced metastasis. Here, we unveil a novel mechanism linking CAV1 to selective loading of exosomes with metastasis-promoting microRNAs. RESULTS: We identify hnRNPK as a CAV1-regulated microRNA binding protein. In the absence of CAVIN1, non-caveolar CAV1 drives localisation of hnRPNK to multi-vesicular bodies (MVBs), recruiting AsUGnA motif-containing miRNAs and causing their release within exosomes. This process is dependent on the lipid environment of membranes as shown by cholesterol depletion using methyl-ß-cyclodextrin or by treatment with n-3 polyunsaturated fatty acids. Consistent with a role in bone metastasis, knockdown of hnRNPK in prostate cancer PC3 cells abolished the ability of PC3 extracellular vesicles (EV) to induce osteoclastogenesis, and biofluid EV hnRNPK is elevated in metastatic prostate and colorectal cancer. CONCLUSIONS: Taken together, these results support a novel pan-cancer mechanism for CAV1-driven exosomal release of hnRNPK and associated miRNA in metastasis, which is modulated by the membrane lipid environment.


Assuntos
Caveolina 1/metabolismo , Neoplasias Colorretais/metabolismo , Exossomos/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo K/metabolismo , MicroRNAs/metabolismo , Neoplasias da Próstata/metabolismo , Membrana Celular/metabolismo , Células HEK293 , Humanos , Masculino , RNA Neoplásico/metabolismo
5.
Bioessays ; 43(5): e2000245, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33748979

RESUMO

Paraspeckles are nuclear condensates, or membranelees organelles, that are built on the long noncoding RNA, NEAT1, and have been linked to many diseases. Although originally described as constitutive structures, here, in reviewing this field, we develop the hypothesis that cells increase paraspeckle abundance as part of a general stress response, to aid pro-survival pathways. Paraspeckles increase in many scenarios: when cells transform from one state to another, become infected with viruses and bacteria, begin to degenerate, under inflammation, in aging, and in cancer. Cells increase paraspeckles by increasing transcription of NEAT1 and adjusting its RNA processing. These increases in NEAT1 are driven by numerous stress-sensing signaling pathways, including signaling to mitochondria and stress granules, revealing crosstalk between the cytoplasm and nucleoplasm in the stress response. Thus, paraspeckles are an important piece of the puzzle in cellular homeostasis, and could be considered RNA-scaffolded nuclear equivalents of dynamic stress-induced structures that form in the cytoplasm. We speculate that, in general, cells rely on phase-separated paraspeckles to transiently tweak gene regulation in times of cellular flux.


Assuntos
Núcleo Celular , RNA Longo não Codificante , Regulação da Expressão Gênica , RNA Longo não Codificante/genética
6.
Cell Mol Life Sci ; 78(5): 2213-2230, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32914209

RESUMO

Many long non-coding RNAs (lncRNA) are highly dysregulated in cancer and are emerging as therapeutic targets. One example is NEAT1, which consists of two overlapping lncRNA isoforms, NEAT1_1 (3.7 kb) and NEAT1_2 (23 kb), that are functionally distinct. The longer NEAT1_2 is responsible for scaffolding gene-regulatory nuclear bodies termed paraspeckles, whereas NEAT1_1 is involved in paraspeckle-independent function. The NEAT1 isoform ratio is dependent on the efficient cleavage and polyadenylation of NEAT1_1 at the expense of NEAT1_2. Here, we developed a targeted antisense oligonucleotide (ASO) approach to sterically block NEAT1_1 polyadenylation processing, achieving upregulation of NEAT1_2 and abundant paraspeckles. We have applied these ASOs to cells of the heterogeneous infant cancer, neuroblastoma, as we found higher NEAT1_1:NEAT1_2 ratio and lack of paraspeckles in high-risk neuroblastoma cells. These ASOs decrease NEAT1_1 levels, increase NEAT1_2/paraspeckles and concomitantly reduce cell viability in high-risk neuroblastoma specifically. In contrast, overexpression of NEAT1_1 has the opposite effect, increasing cell proliferation. Transcriptomic analyses of high-risk neuroblastoma cells with altered NEAT1 ratios and increased paraspeckle abundance after ASO treatment showed an upregulation of differentiation pathways, as opposed to the usual aggressive neuroblastic phenotype. Thus, we have developed potential anti-cancer ASO drugs that can transiently increase growth-inhibiting NEAT1_2 RNA at the expense of growth-promoting NEAT1_1 RNA. These ASOs, unlike others that degrade lncRNAs, provide insights into the importance of altering lncRNA polyadenylation events to suppress tumorigenesis as a strategy to combat cancer.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neuroblastoma/genética , Oligonucleotídeos Antissenso/genética , Poli A/genética , RNA Longo não Codificante/genética , Linhagem Celular Tumoral , Estudos de Coortes , Perfilação da Expressão Gênica/métodos , Humanos , Estimativa de Kaplan-Meier , Neuroblastoma/metabolismo , Neuroblastoma/patologia , Poli A/metabolismo , Isoformas de RNA/genética , Isoformas de RNA/metabolismo
7.
Mol Biol Cell ; 31(16): 1654-1662, 2020 07 21.
Artigo em Inglês | MEDLINE | ID: mdl-32293985

RESUMO

Cancer progression is influenced by changes in the tumor microenvironment, such as the stiffening of the extracellular matrix. Yet our understanding of how cancer cells sense and convert mechanical stimuli into biochemical signals and physiological responses is still limited. The long noncoding RNA nuclear paraspeckle assembly transcript 1 (NEAT1), which forms the backbone of subnuclear "paraspeckle" bodies, has been identified as a key genetic regulator in numerous cancers. Here, we investigated whether paraspeckles, as defined by NEAT1 localization, are mechanosensitive. Using tunable polyacrylamide hydrogels of extreme stiffnesses, we measured paraspeckle parameters in several cancer cell lines and observed an increase in paraspeckles in cells cultured on soft (3 kPa) hydrogels compared with stiffer (40 kPa) hydrogels. This response to soft substrate is erased when cells are first conditioned on stiff substrate, and then transferred onto soft hydrogels, suggestive of mechanomemory upstream of paraspeckle regulation. We also examined some well-characterized mechanosensitive markers, but found that lamin A expression, as well as YAP and MRTF-A nuclear translocation did not show consistent trends between stiffnesses, despite all cell types having increased migration, nuclear, and cell area on stiffer hydrogels. We thus propose that paraspeckles may prove of use as mechanosensors in cancer mechanobiology.


Assuntos
Matriz Extracelular/metabolismo , Neoplasias/metabolismo , RNA Longo não Codificante/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Fenômenos Biomecânicos , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Núcleo Celular/metabolismo , Forma Celular , Feminino , Humanos , Lamina Tipo A/metabolismo , Mecanotransdução Celular , Miosina Tipo II/metabolismo , Metástase Neoplásica , Neoplasias/patologia , RNA Longo não Codificante/genética , Transativadores/metabolismo , Fatores de Transcrição/metabolismo , Proteínas de Sinalização YAP
8.
Acta Neuropathol Commun ; 7(1): 7, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30642400

RESUMO

Mutations in the FUS gene cause amyotrophic lateral sclerosis (ALS-FUS). Mutant FUS is known to confer cytoplasmic gain of function but its effects in the nucleus are less understood. FUS is an essential component of paraspeckles, subnuclear bodies assembled on a lncRNA NEAT1. Paraspeckles may play a protective role specifically in degenerating spinal motor neurons. However it is still unknown how endogenous levels of mutant FUS would affect NEAT1/paraspeckles. Using novel cell lines with the FUS gene modified by CRISPR/Cas9 and human patient fibroblasts, we found that endogenous levels of mutant FUS cause accumulation of NEAT1 isoforms and paraspeckles. However, despite only mild cytoplasmic mislocalisation of FUS, paraspeckle integrity is compromised in these cells, as confirmed by reduced interaction of mutant FUS with core paraspeckle proteins NONO and SFPQ and increased NEAT1 extractability. This results in NEAT1 localisation outside paraspeckles, especially prominent under conditions of paraspeckle-inducing stress. Consistently, paraspeckle-dependent microRNA production, a readout for functionality of paraspeckles, is impaired in cells expressing mutant FUS. In line with the cellular data, we observed paraspeckle hyper-assembly in spinal neurons of ALS-FUS patients. Therefore, despite largely preserving its nuclear localisation, mutant FUS leads to loss (dysfunctional paraspeckles) and gain (excess of free NEAT1) of function in the nucleus. Perturbed fine structure and functionality of paraspeckles accompanied by accumulation of non-paraspeckle NEAT1 may contribute to the disease severity in ALS-FUS.


Assuntos
Esclerose Lateral Amiotrófica/metabolismo , Esclerose Lateral Amiotrófica/patologia , Núcleo Celular/metabolismo , Corpos de Inclusão Intranuclear/metabolismo , RNA Longo não Codificante/metabolismo , Proteína FUS de Ligação a RNA/genética , Proteína FUS de Ligação a RNA/metabolismo , Esclerose Lateral Amiotrófica/genética , Sistemas CRISPR-Cas , Linhagem Celular , Linhagem Celular Tumoral , Humanos , Mutação com Perda de Função , Isoformas de Proteínas/metabolismo
9.
Cell ; 175(2): 488-501.e22, 2018 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-30270045

RESUMO

Detection of viruses by innate immune sensors induces protective antiviral immunity. The viral DNA sensor cyclic GMP-AMP synthase (cGAS) is necessary for detection of HIV by human dendritic cells and macrophages. However, synthesis of HIV DNA during infection is not sufficient for immune activation. The capsid protein, which associates with viral DNA, has a pivotal role in enabling cGAS-mediated immune activation. We now find that NONO is an essential sensor of the HIV capsid in the nucleus. NONO protein directly binds capsid with higher affinity for weakly pathogenic HIV-2 than highly pathogenic HIV-1. Upon infection, NONO is essential for cGAS activation by HIV and cGAS association with HIV DNA in the nucleus. NONO recognizes a conserved region in HIV capsid with limited tolerance for escape mutations. Detection of nuclear viral capsid by NONO to promote DNA sensing by cGAS reveals an innate strategy to achieve distinction of viruses from self in the nucleus.


Assuntos
Proteínas do Capsídeo/imunologia , Proteínas Associadas à Matriz Nuclear/imunologia , Proteínas Associadas à Matriz Nuclear/fisiologia , Fatores de Transcrição de Octâmero/imunologia , Fatores de Transcrição de Octâmero/fisiologia , Proteínas de Ligação a RNA/imunologia , Proteínas de Ligação a RNA/fisiologia , Capsídeo/metabolismo , Proteínas do Capsídeo/metabolismo , Proteínas do Capsídeo/fisiologia , Núcleo Celular/metabolismo , DNA Viral/genética , DNA Viral/imunologia , Proteínas de Ligação a DNA , Células Dendríticas/imunologia , Infecções por HIV/imunologia , HIV-1/genética , HIV-1/imunologia , HIV-2/genética , HIV-2/imunologia , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata/imunologia , Macrófagos/imunologia , Proteínas de Membrana/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Nucleotidiltransferases/metabolismo , Nucleotidiltransferases/fisiologia , Proteínas de Ligação a RNA/metabolismo , Transdução de Sinais/imunologia
10.
J Biol Chem ; 293(17): 6593-6602, 2018 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-29530979

RESUMO

Members of the Drosophila behavior human splicing (DBHS) protein family are nuclear proteins implicated in many layers of nuclear functions, including RNA biogenesis as well as DNA repair. Definitive of the DBHS protein family, the conserved DBHS domain provides a dimerization platform that is critical for the structural integrity and function of these proteins. The three human DBHS proteins, splicing factor proline- and glutamine-rich (SFPQ), paraspeckle component 1 (PSPC1), and non-POU domain-containing octamer-binding protein (NONO), form either homo- or heterodimers; however, the relative affinity and mechanistic details of preferential heterodimerization are yet to be deciphered. Here we report the crystal structure of a SFPQ/PSPC1 heterodimer to 2.3-Å resolution and analyzed the subtle structural differences between the SFPQ/PSPC1 heterodimer and the previously characterized SFPQ homodimer. Analytical ultracentrifugation to estimate the dimerization equilibrium of the SFPQ-containing dimers revealed that the SFPQ-containing dimers dissociate at low micromolar concentrations and that the heterodimers have higher affinities than the homodimer. Moreover, we observed that the apparent dissociation constant for the SFPQ/PSPC1 heterodimer was over 6-fold lower than that of the SFPQ/NONO heterodimer. We propose that these differences in dimerization affinity may represent a potential mechanism by which PSPC1 at a lower relative cellular abundance can outcompete NONO to heterodimerize with SFPQ.


Assuntos
Proteínas Nucleares/química , Fator de Processamento Associado a PTB/química , Multimerização Proteica , Proteínas de Ligação a RNA/química , Cristalografia por Raios X , Humanos , Proteínas Nucleares/metabolismo , Fator de Processamento Associado a PTB/metabolismo , Estrutura Quaternária de Proteína , Proteínas de Ligação a RNA/metabolismo
11.
Curr Opin Cell Biol ; 46: 94-101, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28577509

RESUMO

The cell nucleus contains a number of different dynamic bodies that are variously composed of proteins and generally, but not always, specific RNA molecules. Recent studies have revealed new understanding about nuclear body formation and function in different aspects of nuclear metabolism. Here, we focus on findings describing the role of nuclear bodies in the biogenesis of specific ribonucleoprotein complexes, processing of key mRNAs, and subnuclear sequestration of protein factors. We highlight how nuclear bodies are involved in stress responses, innate immunity and tumorigenesis. We further review organization of nuclear bodies and principles that govern their assembly, highlighting the pivotal role of scaffolding noncoding RNAs, and liquid-liquid phase separation, which are transforming our picture of nuclear body formation.


Assuntos
Corpos de Inclusão Intranuclear/química , Corpos de Inclusão Intranuclear/metabolismo , Animais , Núcleo Celular/metabolismo , Humanos , Corpos de Inclusão Intranuclear/patologia , Proteínas Nucleares/metabolismo , RNA não Traduzido/metabolismo
12.
Neuron ; 94(2): 322-336.e5, 2017 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-28392072

RESUMO

Recent progress revealed the complexity of RNA processing and its association to human disorders. Here, we unveil a new facet of this complexity. Complete loss of function of the ubiquitous splicing factor SFPQ affects zebrafish motoneuron differentiation cell autonomously. In addition to its nuclear localization, the protein unexpectedly localizes to motor axons. The cytosolic version of SFPQ abolishes motor axonal defects, rescuing key transcripts, and restores motility in the paralyzed sfpq null mutants, indicating a non-nuclear processing role in motor axons. Novel variants affecting the conserved coiled-coil domain, so far exclusively found in fALS exomes, specifically affect the ability of SFPQ to localize in axons. They broadly rescue morphology and motility in the zebrafish mutant, but alter motor axon morphology, demonstrating functional requirement for axonal SFPQ. Altogether, we uncover the axonal function of the splicing factor SFPQ in motor development and highlight the importance of the coiled-coil domain in this process. VIDEO ABSTRACT.


Assuntos
Axônios/metabolismo , Neurônios Motores/metabolismo , Fator de Processamento Associado a PTB/metabolismo , Splicing de RNA/genética , Esclerose Lateral Amiotrófica/metabolismo , Animais , Proteínas de Ligação a DNA/metabolismo , Humanos , Camundongos , Córtex Motor/crescimento & desenvolvimento , Fator de Processamento Associado a PTB/genética , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/metabolismo , Peixe-Zebra
13.
RNA ; 23(6): 872-881, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28325845

RESUMO

Large numbers of long noncoding RNAs have been discovered in recent years, but only a few have been characterized. NEAT1 (nuclear paraspeckle assembly transcript 1) is a mammalian long noncoding RNA that is important for the reproductive physiology of mice, cancer development, and the formation of subnuclear bodies termed paraspeckles. The two major isoforms of NEAT1 (3.7 kb NEAT1_1 and 23 kb NEAT1_2 in human) are generated from a common promoter and are produced through the use of alternative transcription termination sites. This gene structure has made the functional relationship between the two isoforms difficult to dissect. Here we used CRISPR-Cas9 genome editing to create several different cell lines: total NEAT1 knockout cells, cells that only express the short form NEAT1_1, and cells with twofold more NEAT1_2. Using these reagents, we obtained evidence that NEAT1_1 is not a major component of paraspeckles. In addition, our data suggest NEAT1_1 localizes in numerous nonparaspeckle foci we termed "microspeckles," which may carry paraspeckle-independent functions. This study highlights the complexity of lncRNA and showcases how genome editing tools are useful in dissecting the structural and functional roles of overlapping transcripts.


Assuntos
Edição de Genes , RNA Longo não Codificante/genética , Sistemas CRISPR-Cas , Linhagem Celular , Núcleo Celular/metabolismo , Repetições Palindrômicas Curtas Agrupadas e Regularmente Espaçadas , Técnicas de Inativação de Genes , Humanos , Isoformas de RNA , Fatores de Processamento de RNA/metabolismo , Transporte de RNA , RNA Guia de Cinetoplastídeos
14.
Hum Mol Genet ; 26(11): 2042-2052, 2017 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-28334947

RESUMO

De novo and inherited mutations of X-chromosome cell adhesion molecule protocadherin 19 (PCDH19) cause frequent, highly variable epilepsy, autism, cognitive decline and behavioural problems syndrome. Intriguingly, hemizygous null males are not affected while heterozygous females are, contradicting established X-chromosome inheritance. The disease mechanism is not known. Cellular mosaicism is the likely driver. We have identified p54nrb/NONO, a multifunctional nuclear paraspeckle protein with known roles in nuclear hormone receptor gene regulation, as a PCDH19 protein interacting partner. Using breast cancer cells we show that PCDH19-NONO complex is a positive co-regulator of ERα-mediated gene expression. Expression of mutant PCDH19 affects at least a subset of known ERα-regulated genes. These data are consistent with our findings that genes regulated by nuclear hormone receptors and those involved in the metabolism of neurosteroids in particular are dysregulated in PCDH19-epilepsy girls and affected mosaic males. Overall we define and characterize a novel mechanism of gene regulation driven by PCDH19, which is mediated by paraspeckle constituent NONO and is ERα-dependent. This PCDH19-NONO-ERα axis is of relevance not only to PCDH19-epilepsy and its comorbidities but likely also to ERα and generally nuclear hormone receptor-associated cancers.


Assuntos
Caderinas/metabolismo , Proteínas Associadas à Matriz Nuclear/metabolismo , Fatores de Transcrição de Octâmero/metabolismo , Proteínas de Ligação a RNA/metabolismo , Neoplasias da Mama/metabolismo , Caderinas/genética , Linhagem Celular Tumoral , Proteínas de Ligação a DNA , Epilepsia/genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Humanos , Deficiência Intelectual/genética , Mutação , Proteínas Associadas à Matriz Nuclear/genética , Fatores de Transcrição de Octâmero/genética , Linhagem , Protocaderinas , Proteínas de Ligação a RNA/genética
15.
Nucleic Acids Res ; 44(9): 3989-4004, 2016 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-27084935

RESUMO

Nuclear proteins are often given a concise title that captures their function, such as 'transcription factor,' 'polymerase' or 'nuclear-receptor.' However, for members of the Drosophila behavior/human splicing (DBHS) protein family, no such clean-cut title exists. DBHS proteins are frequently identified engaging in almost every step of gene regulation, including but not limited to, transcriptional regulation, RNA processing and transport, and DNA repair. Herein, we present a coherent picture of DBHS proteins, integrating recent structural insights on dimerization, nucleic acid binding modalities and oligomerization propensity with biological function. The emerging paradigm describes a family of dynamic proteins mediating a wide range of protein-protein and protein-nucleic acid interactions, on the whole acting as a multipurpose molecular scaffold. Overall, significant steps toward appreciating the role of DBHS proteins have been made, but we are only beginning to understand the complexity and broader importance of this family in cellular biology.


Assuntos
Regulação da Expressão Gênica/genética , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas Nucleares/metabolismo , Fatores de Transcrição de Octâmero/metabolismo , Fator de Processamento Associado a PTB/metabolismo , Processamento Pós-Transcricional do RNA/genética , Fatores de Processamento de RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Animais , Caenorhabditis elegans , Reparo do DNA/genética , Proteínas de Ligação a DNA , Drosophila melanogaster , Humanos , Proteínas Associadas à Matriz Nuclear/ultraestrutura , Proteínas Nucleares/ultraestrutura , Fatores de Transcrição de Octâmero/ultraestrutura , Fator de Processamento Associado a PTB/ultraestrutura , Mapas de Interação de Proteínas , Estrutura Secundária de Proteína , Proteínas de Ligação a RNA/ultraestrutura , Transcrição Gênica/genética , Ativação Transcricional/genética
16.
Nucleic Acids Res ; 43(7): 3826-40, 2015 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-25765647

RESUMO

SFPQ, (a.k.a. PSF), is a human tumor suppressor protein that regulates many important functions in the cell nucleus including coordination of long non-coding RNA molecules into nuclear bodies. Here we describe the first crystal structures of Splicing Factor Proline and Glutamine Rich (SFPQ), revealing structural similarity to the related PSPC1/NONO heterodimer and a strikingly extended structure (over 265 Å long) formed by an unusual anti-parallel coiled-coil that results in an infinite linear polymer of SFPQ dimers within the crystals. Small-angle X-ray scattering and transmission electron microscopy experiments show that polymerization is reversible in solution and can be templated by DNA. We demonstrate that the ability to polymerize is essential for the cellular functions of SFPQ: disruptive mutation of the coiled-coil interaction motif results in SFPQ mislocalization, reduced formation of nuclear bodies, abrogated molecular interactions and deficient transcriptional regulation. The coiled-coil interaction motif thus provides a molecular explanation for the functional aggregation of SFPQ that directs its role in regulating many aspects of cellular nucleic acid metabolism.


Assuntos
Regulação da Expressão Gênica/fisiologia , Polímeros/química , Proteínas de Ligação a RNA/química , Western Blotting , Cristalografia por Raios X , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Microscopia Eletrônica de Transmissão , Fator de Processamento Associado a PTB , Conformação Proteica , Proteínas de Ligação a RNA/fisiologia
17.
Nat Commun ; 5: 5383, 2014 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-25415230

RESUMO

The androgen receptor (AR) plays a central role in establishing an oncogenic cascade that drives prostate cancer progression. Some prostate cancers escape androgen dependence and are often associated with an aggressive phenotype. The oestrogen receptor alpha (ERα) is expressed in prostate cancers, independent of AR status. However, the role of ERα remains elusive. Using a combination of chromatin immunoprecipitation (ChIP) and RNA-sequencing data, we identified an ERα-specific non-coding transcriptome signature. Among putatively ERα-regulated intergenic long non-coding RNAs (lncRNAs), we identified nuclear enriched abundant transcript 1 (NEAT1) as the most significantly overexpressed lncRNA in prostate cancer. Analysis of two large clinical cohorts also revealed that NEAT1 expression is associated with prostate cancer progression. Prostate cancer cells expressing high levels of NEAT1 were recalcitrant to androgen or AR antagonists. Finally, we provide evidence that NEAT1 drives oncogenic growth by altering the epigenetic landscape of target gene promoters to favour transcription.


Assuntos
Adenocarcinoma/genética , Epigênese Genética , Receptor alfa de Estrogênio/genética , Neoplasias da Próstata/genética , RNA Longo não Codificante/genética , Adenocarcinoma/metabolismo , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina , Progressão da Doença , Receptor alfa de Estrogênio/metabolismo , Humanos , Masculino , Neoplasias da Próstata/metabolismo , RNA Longo não Codificante/metabolismo , Análise de Sequência de RNA
18.
Mol Biol Cell ; 25(1): 169-83, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24173718

RESUMO

Paraspeckles are subnuclear structures formed around nuclear paraspeckle assembly transcript 1 (NEAT1)/MENε/ß long noncoding RNA (lncRNA). Here we show that paraspeckles become dramatically enlarged after proteasome inhibition. This enlargement is mainly caused by NEAT1 transcriptional up-regulation rather than accumulation of undegraded paraspeckle proteins. Of interest, however, using immuno-electron microscopy, we find that key paraspeckle proteins become effectively depleted from the nucleoplasm by 50% when paraspeckle assembly is enhanced, suggesting a sequestration mechanism. We also perform microarrays from NEAT1-knockdown cells and find that NEAT1 represses transcription of several genes, including the RNA-specific adenosine deaminase B2 (ADARB2) gene. In contrast, the NEAT1-binding paraspeckle protein splicing factor proline/glutamine-rich (SFPQ) is required for ADARB2 transcription. This leads us to hypothesize that ADARB2 expression is controlled by NEAT1-dependent sequestration of SFPQ. Accordingly, we find that ADARB2 expression is strongly reduced upon enhanced SFPQ sequestration by proteasome inhibition, with concomitant reduction in SFPQ binding to the ADARB2 promoter. Finally, NEAT1(-/-) fibroblasts are more sensitive to proteasome inhibition, which triggers cell death, suggesting that paraspeckles/NEAT1 attenuates the cell death pathway. These data further confirm that paraspeckles are stress-responsive nuclear bodies and provide a model in which induced NEAT1 controls target gene transcription by protein sequestration into paraspeckles.


Assuntos
Núcleo Celular/metabolismo , Regulação da Expressão Gênica , RNA Longo não Codificante/genética , Transcrição Gênica , Adenosina Desaminase/genética , Adenosina Desaminase/metabolismo , Núcleo Celular/ultraestrutura , Proteínas de Ligação a DNA , Células HeLa , Humanos , Leupeptinas/farmacologia , Proteínas Associadas à Matriz Nuclear/metabolismo , Fatores de Transcrição de Octâmero/metabolismo , Regiões Promotoras Genéticas , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Transporte Proteico , Proteólise , RNA Longo não Codificante/metabolismo , Proteínas de Ligação a RNA/metabolismo
19.
Proc Natl Acad Sci U S A ; 109(13): 4846-50, 2012 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-22416126

RESUMO

Proteins of the Drosophila behavior/human splicing (DBHS) family include mammalian SFPQ (PSF), NONO (p54nrb), PSPC1, and invertebrate NONA and Hrp65. DBHS proteins are predominately nuclear, and are involved in transcriptional and posttranscriptional gene regulatory functions as well as DNA repair. DBHS proteins influence a wide gamut of biological processes, including the regulation of circadian rhythm, carcinogenesis, and progression of cancer. Additionally, mammalian DBHS proteins associate with the architectural long noncoding RNA NEAT1 (Menε/ß) to form paraspeckles, subnuclear bodies that alter gene expression via the nuclear retention of RNA. Here we describe the crystal structure of the heterodimer of the multidomain conserved region of the DBHS proteins, PSPC1 and NONO. These proteins form an extensively intertwined dimer, consistent with the observation that the different DBHS proteins are typically copurified from mammalian cells, and suggesting that they act as obligate heterodimers. The PSPC1/NONO heterodimer has a right-handed antiparallel coiled-coil that positions two of four RNA recognition motif domains in an unprecedented arrangement on either side of a 20-Å channel. This configuration is supported by a protein:protein interaction involving the NONA/paraspeckle domain, which is characteristic of the DBHS family. By examining various mutants and truncations in cell culture, we find that DBHS proteins require an additional antiparallel coiled-coil emanating from either end of the dimer for paraspeckle subnuclear body formation. These results suggest that paraspeckles may potentially form through self-association of DBHS dimers into higher-order structures.


Assuntos
Espaço Intranuclear/metabolismo , Proteínas Associadas à Matriz Nuclear/química , Proteínas Associadas à Matriz Nuclear/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Fatores de Transcrição de Octâmero/química , Fatores de Transcrição de Octâmero/metabolismo , Multimerização Proteica , Proteínas de Ligação a RNA/química , Proteínas de Ligação a RNA/metabolismo , Sequência de Aminoácidos , Sequência Conservada/genética , Proteínas de Ligação a DNA , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Domínios e Motivos de Interação entre Proteínas , Estrutura Terciária de Proteína , Relação Estrutura-Atividade
20.
Genome Res ; 22(5): 885-98, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22406755

RESUMO

Transcriptomic analyses have identified tens of thousands of intergenic, intronic, and cis-antisense long noncoding RNAs (lncRNAs) that are expressed from mammalian genomes. Despite progress in functional characterization, little is known about the post-transcriptional regulation of lncRNAs and their half-lives. Although many are easily detectable by a variety of techniques, it has been assumed that lncRNAs are generally unstable, but this has not been examined genome-wide. Utilizing a custom noncoding RNA array, we determined the half-lives of ∼800 lncRNAs and ∼12,000 mRNAs in the mouse Neuro-2a cell line. We find only a minority of lncRNAs are unstable. LncRNA half-lives vary over a wide range, comparable to, although on average less than, that of mRNAs, suggestive of complex metabolism and widespread functionality. Combining half-lives with comprehensive lncRNA annotations identified hundreds of unstable (half-life < 2 h) intergenic, cis-antisense, and intronic lncRNAs, as well as lncRNAs showing extreme stability (half-life > 16 h). Analysis of lncRNA features revealed that intergenic and cis-antisense RNAs are more stable than those derived from introns, as are spliced lncRNAs compared to unspliced (single exon) transcripts. Subcellular localization of lncRNAs indicated widespread trafficking to different cellular locations, with nuclear-localized lncRNAs more likely to be unstable. Surprisingly, one of the least stable lncRNAs is the well-characterized paraspeckle RNA Neat1, suggesting Neat1 instability contributes to the dynamic nature of this subnuclear domain. We have created an online interactive resource (http://stability.matticklab.com) that allows easy navigation of lncRNA and mRNA stability profiles and provides a comprehensive annotation of ~7200 mouse lncRNAs.


Assuntos
Genoma , Camundongos/genética , Estabilidade de RNA , RNA não Traduzido/metabolismo , Análise de Variância , Animais , Linhagem Celular Tumoral , Análise por Conglomerados , Expressão Gênica , Meia-Vida , Humanos , Anotação de Sequência Molecular , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA não Traduzido/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA