Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
J Biol Chem ; 299(8): 105071, 2023 08.
Article in English | MEDLINE | ID: mdl-37474102

ABSTRACT

Paraspeckles (PS) are nuclear structures scaffolded by the long noncoding RNA NEAT1 and protein components such as NONO and SFPQ. We previously found that the upregulation of RNA N6-methyl-adenosine (m6A) demethylase ALKBH5 facilitates hypoxia-induced paraspeckle assembly through erasing m6A marks on NEAT1, thus stabilizing it. However, it remains unclear how these processes are spatiotemporally coordinated. Here we discover that ALKBH5 specifically binds to proteins in PS and forms phase-separated droplets that are incorporated into PS through its C-terminal intrinsically disordered region (cIDR). Upon exposure to hypoxia, rapid ALKBH5 condensation in PS induces m6A demethylation of NEAT1, which further facilitates PS formation before the upregulation of ALKBH5 expression. In cells expressing ALKBH5 lacking cIDR, PS fail to be formed in response to hypoxia, accompanied with insufficient m6A demethylation of NEAT1 and its destabilization. We also demonstrate that ALKBH5-cIDR is indispensable for hypoxia-induced effects such as cancer cell invasion. Therefore, our study has identified the role of ALKBH5 in phase separation as the molecular basis of the positive feedback loop for PS formation between ALKBH5 incorporation into PS and NEAT1 stabilization.


Subject(s)
AlkB Homolog 5, RNA Demethylase , Paraspeckles , RNA, Long Noncoding , Humans , AlkB Homolog 5, RNA Demethylase/genetics , AlkB Homolog 5, RNA Demethylase/metabolism , Hypoxia , Paraspeckles/metabolism , RNA, Long Noncoding/genetics , Transcriptional Activation , Up-Regulation
2.
Br J Cancer ; 126(4): 551-561, 2022 03.
Article in English | MEDLINE | ID: mdl-34671127

ABSTRACT

NEAT1 is a highly abundant nuclear architectural long non-coding RNA. There are two overlapping NEAT1 isoforms, NEAT1_1 and NEAT1_2, of which the latter is an essential scaffold for the assembly of a class of nuclear ribonucleoprotein bodies called paraspeckles. Paraspeckle formation is elevated by a wide variety of cellular stressors and in certain developmental processes, either through transcriptional upregulation of the NEAT1 gene or through a switch from NEAT1_1 to NEAT1_2 isoform production. In such conditions, paraspeckles modulate cellular processes by sequestering proteins or RNA molecules. NEAT1 is abnormally expressed in many cancers and a growing body of evidence suggests that, in many cases, high NEAT1 levels are associated with therapy resistance and poor clinical outcome. Here we review the current knowledge of NEAT1 expression and functions in breast cancer, highlighting its established role in postnatal mammary gland development. We will discuss possible isoform-specific roles of NEAT1_1 and NEAT1_2 in different breast cancer subtypes, which critically needs to be considered when studying NEAT1 and breast cancer.


Subject(s)
Alternative Splicing , Breast Neoplasms/genetics , RNA, Long Noncoding/genetics , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm , Female , Gene Expression Regulation, Neoplastic , Humans , Paraspeckles/metabolism , Up-Regulation
3.
Cancer Res ; 81(23): 5876-5888, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34670781

ABSTRACT

The dynamic changes of RNA N6-methyl-adenosine (m6A) during cancer progression contribute to quick adaption to microenvironmental changes. Here, we profiled the cancer cell m6A dynamics in the hypoxic tumor niche and its pathological consequences in glioblastoma multiforme (GBM). The m6A demethylase ALKBH5 was induced in GBM models under hypoxic conditions and was associated with a hypoxic gene signature in GBM patient samples. Depletion or inactivation of ALKBH5 in GBM cells significantly suppressed hypoxia-induced tumor-associated macrophage (TAM) recruitment and immunosuppression in allograft tumors. Expression and secretion of CXCL8/IL8 were significantly suppressed in ALKBH5-deficient tumors. However, ALKBH5 did not regulate CXCL8 m6A directly. Instead, hypoxia-induced ALKBH5 erased m6A deposition from the lncRNA NEAT1, stabilizing the transcript and facilitating NEAT1-mediated paraspeckle assembly, which led to relocation of the transcriptional repressor SFPQ from the CXCL8 promoter to paraspeckles and, ultimately, upregulation of CXCL8/IL8 expression. Accordingly, ectopic expression of CXCL8 in ALKBH5-deficient GBM cells partially restored TAM recruitment and tumor progression. Together, this study links hypoxia-induced epitranscriptomic changes to the emergence of an immunosuppressive microenvironment facilitating tumor evasion. SIGNIFICANCE: Hypoxia induces tumor immune microenvironment remodeling through an ALKBH5-mediated epigenetic and epitranscriptomic mechanism, providing potential immunotherapeutic strategies for treating glioblastoma.


Subject(s)
AlkB Homolog 5, RNA Demethylase/metabolism , DNA Methylation , Gene Expression Regulation, Neoplastic , Glioblastoma/pathology , Interleukin-8/metabolism , Paraspeckles/pathology , Tumor Microenvironment , AlkB Homolog 5, RNA Demethylase/genetics , Animals , Apoptosis , Cell Proliferation , Glioblastoma/immunology , Glioblastoma/metabolism , Humans , Immunosuppressive Agents , Interleukin-8/genetics , Male , Mice , Mice, Inbred C57BL , Paraspeckles/immunology , Paraspeckles/metabolism , Tumor Cells, Cultured , Tumor-Associated Macrophages/immunology , Xenograft Model Antitumor Assays
4.
Cell Rep ; 36(8): 109576, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433054

ABSTRACT

Paraspeckles are membraneless organelles formed through liquid-liquid phase separation and consist of multiple proteins and RNAs, including NONO, SFPQ, and NEAT1. The role of paraspeckles and the component NONO in hematopoiesis remains unknown. In this study, we show histone modifier ASXL1 is involved in paraspeckle formation. ASXL1 forms phase-separated droplets, upregulates NEAT1 expression, and increases NONO-NEAT1 interactions through the C-terminal intrinsically disordered region (IDR). In contrast, a pathogenic ASXL mutant (ASXL1-MT) lacking IDR does not support the interaction of paraspeckle components. Furthermore, paraspeckles are disrupted and Nono localization is abnormal in the cytoplasm of hematopoietic stem and progenitor cells (HSPCs) derived from ASXL1-MT knockin mice. Nono depletion and the forced expression of cytoplasmic NONO impair the repopulating potential of HSPCs, as does ASXL1-MT. Our study indicates a link between ASXL1 and paraspeckle components in the maintenance of normal hematopoiesis.


Subject(s)
DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/metabolism , Paraspeckles/metabolism , RNA-Binding Proteins/metabolism , Repressor Proteins/metabolism , Animals , DNA-Binding Proteins/genetics , Female , HL-60 Cells , HeLa Cells , Hematopoiesis , Humans , Mice , Mice, Transgenic , Paraspeckles/genetics , RNA-Binding Proteins/genetics , Repressor Proteins/genetics , THP-1 Cells
5.
Biochimie ; 190: 124-131, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34329720

ABSTRACT

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.


Subject(s)
G-Quadruplexes , Neoplasms/genetics , Neoplasms/metabolism , Paraspeckles/genetics , Paraspeckles/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Humans , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
6.
Cell Mol Gastroenterol Hepatol ; 12(2): 465-487, 2021.
Article in English | MEDLINE | ID: mdl-33667716

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is the most common type of hepatic malignancies, with poor prognosis and low survival rate. Paraspeckles, which are unique subnuclear structures, are recently found to be involved in the development of various tumors, including HCC, and are related to induction in chemoresistance of HCC. This study aimed to investigate the possibility of paraspeckle in HCC cells participating in immune escape and its underlying mechanism in vitro and in vivo. METHODS: Expression of NEAT1_2, the framework of paraspeckle, in HCC cells and tissues was detected by qRT-PCR and RNA-FISH. mRNAs interacted with NEAT1_2 were pull-downed and sequenced in C-terminal S1-aptamer-tagged NEAT1_2 endogenously expressed HCC cells constructed using CRISPR-CAS9 knock-in technology. The effects of paraspeckle on HCC sensitivity to T-cell-mediated cytolysis were detected by T-cell mediated tumor cell killing assay. The roles of NEAT1_2 or NONO on IFNGR1 expression and IFN-γ signaling by applying gene function loss analysis in HCC cells were detected by qRT-PCR, RNA immunoprecipitation, Western blotting, and ELISA. The role of paraspeckle during adoptive T-cell transfer therapy for HCC in vivo was performed with a subcutaneous xenograft mouse. RESULTS: Paraspeckle in HCC cells is negatively related to T-cell-mediated cytolysis. Destruction of paraspeckle in HCC cells by knockdown of NEAT1_2 or NONO significantly improved the sensibility of resistant HCC cells to T-cell killing effects. Furthermore, IFNGR1 mRNA, which is sequestered by NEAT1_2 and NONO, is abundant in paraspeckle of T-cell killing-resistant HCC cells. Incapable IFN-γ-IFNGR1 signaling accounts for paraspeckle mediated-adoptive T-cell therapy resistance. Moreover, NEAT1_2 expression negatively correlates with IFNGR1 expression in clinical HCC tissues. CONCLUSIONS: Paraspeckle in HCC cells helps tumor cells escape from immunosurveillance through sequestering IFNGR1 mRNA to inhibiting IFN-γ-IFNGR1 signaling, thereby avoiding T-cell killing effects. Collectively, our results hint that NEAT1_2 highly expressed HCC patient is more resistant to T-cell therapy in clinic, and NEAT1_2 may be potential target for HCC immunotherapy.


Subject(s)
Carcinoma, Hepatocellular/immunology , Immune Evasion , Liver Neoplasms/immunology , Paraspeckles/metabolism , Receptors, Interferon/genetics , Adoptive Transfer , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Death , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Humans , Interferon-gamma/metabolism , Liver Neoplasms/pathology , Liver Neoplasms/therapy , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Receptors, Interferon/metabolism , T-Lymphocytes/immunology , Interferon gamma Receptor
7.
RNA Biol ; 18(11): 2016-2027, 2021 11.
Article in English | MEDLINE | ID: mdl-33573434

ABSTRACT

Paraspeckles are nuclear ribonucleic complex formed of a long non-coding RNA, nuclear-enriched abundant transcript one (Neat1) and associated RNA-binding proteins (RBP) whose cellular known functions are to sequester in the nucleus both proteins and RNAs. However, how RNAs are bound to paraspeckles is largely unknown. It is highly likely that binding of RNAs may occur via interactions with RBPs and accordingly, two structures present in the 3'UTR of some RNAs have been shown to allow their association to paraspeckles via protein binding. However, Neat1 could also be involved in the targeting of RNAs through direct RNA-RNA interactions. Using an RNA pull-down procedure adapted to select only RNAs engaged in direct RNA-RNA interactions and followed by RNA-seq we showed that in a rat pituitary cell line, GH4C1 cells, 1791 RNAs were associated with paraspeckles by direct interaction with Neat1. Neat1 was actually found able to bind more than 30% of the total transcripts targeted by the paraspeckles, we have identified in this cell line in a previous study. Furthermore, given the biological processes in which direct RNAs targets of Neat1 were involved as determined by gene ontology analysis, it was proposed that Neat1 played a major role in paraspeckle functions such as circadian rhythms, mRNA processing, RNA splicing and regulation of cell cycle. Finally, we provided evidence that direct RNA targets of Neat1 were preferentially bound to the 5' end of Neat1 demonstrating that they are located in the shell region of paraspeckles.


Subject(s)
Cell Nucleus/metabolism , Paraspeckles/metabolism , Pituitary Gland/metabolism , RNA Processing, Post-Transcriptional , RNA, Long Noncoding/genetics , RNA-Binding Proteins/metabolism , RNA/metabolism , Animals , Cell Nucleus/genetics , Cells, Cultured , Paraspeckles/genetics , RNA/genetics , RNA-Binding Proteins/genetics , Rats
8.
RNA Biol ; 18(11): 1807-1817, 2021 11.
Article in English | MEDLINE | ID: mdl-33404286

ABSTRACT

Non-coding RNAs (ncRNAs) are powerful regulators of gene expression but medium-sized (50-300 nts in length) ncRNAs (msRNAs) are barely picked-up precisely by RNA-sequencing. Here we describe msRNA-sequencing (msRNAseq), a modified protocol that associated with a computational analyses pipeline identified about ~1800 msRNA loci, including over 300 putatively novel msRNAs, in human and murine cells. We focused on the identification and initial characterization of three POLIII-derived transcripts. The validation of these uncharacterized msRNAs identified an ncRNA in antisense orientation from the POLR3E locus transcribed by POLIII. This msRNA, termed POLAR (POLR3E Antisense RNA), has a strikingly short half-life, localizes to paraspeckles (PSPs) and associates with PSP-associated proteins indicating that msRNAseq identifies functional msRNAs. Thus, our analyses will pave the way for analysing the roles of msRNAs in cells, development and diseases.


Subject(s)
Paraspeckles/metabolism , RNA Polymerase III/metabolism , RNA, Antisense/genetics , RNA, Messenger/genetics , RNA, Untranslated/genetics , Sequence Analysis, RNA/methods , Humans , Paraspeckles/genetics , RNA Polymerase III/genetics , RNA, Messenger/analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...