Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Elife ; 132024 Jan 19.
Article in English | MEDLINE | ID: mdl-38240312

ABSTRACT

Out of the several hundred copies of rRNA genes arranged in the nucleolar organizing regions (NOR) of the five human acrocentric chromosomes, ~50% remain transcriptionally inactive. NOR-associated sequences and epigenetic modifications contribute to the differential expression of rRNAs. However, the mechanism(s) controlling the dosage of active versus inactive rRNA genes within each NOR in mammals is yet to be determined. We have discovered a family of ncRNAs, SNULs (Single NUcleolus Localized RNA), which form constrained sub-nucleolar territories on individual NORs and influence rRNA expression. Individual members of the SNULs monoallelically associate with specific NOR-containing chromosomes. SNULs share sequence similarity to pre-rRNA and localize in the sub-nucleolar compartment with pre-rRNA. Finally, SNULs control rRNA expression by influencing pre-rRNA sorting to the DFC compartment and pre-rRNA processing. Our study discovered a novel class of ncRNAs influencing rRNA expression by forming constrained nucleolar territories on individual NORs.


Subject(s)
Nucleolus Organizer Region , RNA Precursors , Humans , Animals , Nucleolus Organizer Region/genetics , Nucleolus Organizer Region/metabolism , RNA Precursors/genetics , RNA Precursors/metabolism , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , RNA, Ribosomal/genetics , RNA, Ribosomal/metabolism , Chromosomes, Human/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , Mammals/genetics
2.
Nat Commun ; 14(1): 7238, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37945559

ABSTRACT

The blood proteome holds great promise for precision medicine but poses substantial challenges due to the low abundance of most plasma proteins and the vast dynamic range of the plasma proteome. Here we address these challenges with NUcleic acid Linked Immuno-Sandwich Assay (NULISA™), which improves the sensitivity of traditional proximity ligation assays by ~10,000-fold to attomolar level, by suppressing assay background via a dual capture and release mechanism built into oligonucleotide-conjugated antibodies. Highly multiplexed quantification of both low- and high-abundance proteins spanning a wide dynamic range is achieved by attenuating signals from abundant targets with unconjugated antibodies and next-generation sequencing of barcoded reporter DNA. A 200-plex NULISA containing 124 cytokines and chemokines and other proteins demonstrates superior sensitivity to a proximity extension assay in detecting biologically important low-abundance biomarkers in patients with autoimmune diseases and COVID-19. Fully automated NULISA makes broad and in-depth proteomic analysis easily accessible for research and diagnostic applications.


Subject(s)
Proteome , Proteomics , Humans , Blood Proteins/genetics , Antibodies , Cytokines
3.
bioRxiv ; 2023 Jun 02.
Article in English | MEDLINE | ID: mdl-37090549

ABSTRACT

The blood proteome holds great promise for precision medicine but poses substantial challenges due to the low abundance of most plasma proteins and the vast dynamic range across the proteome. We report a novel proteomic technology - NUcleic acid Linked Immuno-Sandwich Assay (NULISA™) - that incorporates a dual capture and release mechanism to suppress the assay background and improves the sensitivity of the proximity ligation assay by over 10,000-fold to the attomolar level. It utilizes pairs of antibodies conjugated to DNA oligonucleotides that enable immunocomplex purification and generate reporter DNA containing target- and sample-specific barcodes for a next-generation sequencing-based, highly multiplexed readout. A 200-plex NULISA targeting 124 cytokines and chemokines and 80 other immune response-related proteins demonstrated superior sensitivity for detecting low-abundance proteins and high concordance with other immunoassays. The ultrahigh sensitivity allowed the detection of previously difficult-to-detect, but biologically important, low-abundance biomarkers in patients with autoimmune diseases and COVID-19. Fully automated NULISA addresses longstanding challenges in proteomic analysis of liquid biopsies and makes broad and in-depth proteomic analysis accessible to the general research community and future diagnostic applications.

4.
Nat Commun ; 14(1): 551, 2023 02 09.
Article in English | MEDLINE | ID: mdl-36759613

ABSTRACT

Regulation of RNA processing contributes profoundly to tissue development and physiology. Here, we report that serine-arginine-rich splicing factor 1 (SRSF1) is essential for hepatocyte function and survival. Although SRSF1 is mainly known for its many roles in mRNA metabolism, it is also crucial for maintaining genome stability. We show that acute liver damage in the setting of targeted SRSF1 deletion in mice is associated with the excessive formation of deleterious RNA-DNA hybrids (R-loops), which induce DNA damage. Combining hepatocyte-specific transcriptome, proteome, and RNA binding analyses, we demonstrate that widespread genotoxic stress following SRSF1 depletion results in global inhibition of mRNA transcription and protein synthesis, leading to impaired metabolism and trafficking of lipids. Lipid accumulation in SRSF1-deficient hepatocytes is followed by necroptotic cell death, inflammation, and fibrosis, resulting in NASH-like liver pathology. Importantly, SRSF1-depleted human liver cancer cells recapitulate this pathogenesis, illustrating a conserved and fundamental role for SRSF1 in preserving genome integrity and tissue homeostasis. Thus, our study uncovers how the accumulation of detrimental R-loops impedes hepatocellular gene expression, triggering metabolic derangements and liver damage.


Subject(s)
Non-alcoholic Fatty Liver Disease , Humans , Animals , Mice , RNA Splicing Factors/metabolism , Non-alcoholic Fatty Liver Disease/genetics , RNA/metabolism , Serine-Arginine Splicing Factors/genetics , Serine-Arginine Splicing Factors/metabolism , RNA, Messenger/metabolism , Alternative Splicing
5.
Proc Natl Acad Sci U S A ; 119(22): e2121406119, 2022 05 31.
Article in English | MEDLINE | ID: mdl-35622890

ABSTRACT

In eukaryotes, the origin recognition complex (ORC) is required for the initiation of DNA replication. The smallest subunit of ORC, Orc6, is essential for prereplication complex (pre-RC) assembly and cell viability in yeast and for cytokinesis in metazoans. However, unlike other ORC components, the role of human Orc6 in replication remains to be resolved. Here, we identify an unexpected role for hOrc6, which is to promote S-phase progression after pre-RC assembly and DNA damage response. Orc6 localizes at the replication fork and is an accessory factor of the mismatch repair (MMR) complex. In response to oxidative damage during S phase, often repaired by MMR, Orc6 facilitates MMR complex assembly and activity, without which the checkpoint signaling is abrogated. Mechanistically, Orc6 directly binds to MutSα and enhances the chromatin-association of MutLα, thus enabling efficient MMR. Based on this, we conclude that hOrc6 plays a fundamental role in genome surveillance during S phase.


Subject(s)
DNA Mismatch Repair , Origin Recognition Complex , S Phase , DNA-Binding Proteins/metabolism , Humans , MutL Proteins/metabolism , Origin Recognition Complex/genetics , Origin Recognition Complex/metabolism , Protein Binding
6.
Proc Natl Acad Sci U S A ; 119(9)2022 03 01.
Article in English | MEDLINE | ID: mdl-35217604

ABSTRACT

BEN domain-containing proteins are emerging rapidly as an important class of factors involved in modulating gene expression, yet the molecular basis of how they regulate chromatin function and transcription remains to be established. BEND3 is a quadruple BEN domain-containing protein that associates with heterochromatin and functions as a transcriptional repressor. We find that BEND3 is highly expressed in pluripotent cells, and the induction of differentiation results in the down-regulation of BEND3. The removal of BEND3 from pluripotent cells results in cells exhibiting upregulation of the differentiation-inducing gene expression signature. We find that BEND3 binds to the promoters of differentiation-associated factors and key cell cycle regulators, including CDKN1A, encoding the cell cycle inhibitor p21, and represses the expression of differentiation-associated genes by enhancing H3K27me3 decoration at these promoters. Our results support a model in which transcription repression mediated by BEND3 is essential for normal development and to prevent differentiation.


Subject(s)
Cell Differentiation/genetics , Pluripotent Stem Cells/cytology , Repressor Proteins/physiology , G-Quadruplexes , Gene Expression Regulation , Humans , Promoter Regions, Genetic
7.
Mamm Genome ; 33(2): 402-411, 2022 06.
Article in English | MEDLINE | ID: mdl-34436664

ABSTRACT

The nucleolus is the largest sub-nuclear domain, serving primarily as the place for ribosome biogenesis. A delicately regulated function of the nucleolus is vital to the cell not only for maintaining proper protein synthesis but is also tightly associated with responses to different types of cellular stresses. Recently, several long non-coding RNAs (lncRNAs) were found to be part of the regulatory network that modulate nucleolar functions. Several of these lncRNAs are encoded in the ribosomal DNA (rDNA) repeats or are transcribed from the genomic regions that are located near the nucleolus organizer regions (NORs). In this review, we first discuss the current understanding of the sequence of the NORs and variations between different NORs. We then focus on the NOR-derived lncRNAs in mammalian cells and their functions in rRNA transcription and the organization of nucleolar structure under different cellular conditions. The identification of these lncRNAs reveals great potential of the NORs in harboring novel genes involved in the regulation of nucleolar functions.


Subject(s)
Nucleolus Organizer Region , RNA, Long Noncoding , Animals , Cell Nucleolus/genetics , Cell Nucleolus/metabolism , DNA, Ribosomal/genetics , DNA, Ribosomal/metabolism , Mammals/genetics , Nucleolus Organizer Region/genetics , Nucleolus Organizer Region/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription, Genetic
8.
Cancer Res ; 81(6): 1540-1551, 2021 03 15.
Article in English | MEDLINE | ID: mdl-33472891

ABSTRACT

Innate immune defense mechanisms play a pivotal role in antitumor responses. Recent evidence suggests that antiviral innate immunity is regulated not only by exogenous non-self-RNA but also by host-derived pseudogene RNAs. A growing body of evidence also indicates a biological role for pseudogenes as gene expression regulators or immune modulators. Here, we report an important role for BRCA1P1, the pseudogene of the BRCA1 tumor-suppressor gene, in regulating innate immune defense mechanisms in breast cancer cells. BRCA1P1 expresses a long-noncoding RNA (lncRNA) in breast cancer cells through divergent transcription. Expression of lncRNA-BRCA1P1 is increased in breast tumors compared with normal breast tissues. Depletion of BRCA1P1 induces an antiviral defense-like program, including the expression of antiviral genes in breast cancer cells. Furthermore, BRCA1P1-deficient cancer cells mimic virus-infected cells by stimulating cytokines and inducing cell apoptosis. Accordingly, depletion of BRCA1P1 increases host innate immune responses and restricts virus replication. In converse, overexpression of BRCA1P1 reduces cytokine expression in breast cancer cells. Mechanistically, lncRNA-BRCA1P1 is localized in the nucleus, binds to the NF-κB subunit RelA, and negatively regulates antiviral gene expression. Finally, in a xenograft mouse model of breast cancer, depletion of BRCA1P1 stimulates cytokine expression and local immunity, and suppresses tumor growth. Our results suggest an important role for BRCA1P1 in innate immune defense mechanisms and antitumor responses. This mechanism of antiviral immunity regulated by a host-derived pseudogene RNA may guide the development of novel therapies targeting immune responses in breast cancer. SIGNIFICANCE: This study identifies a novel mechanism of innate immunity driven by a host pseudogene RNA that inhibits innate immune defense mechanisms and antitumor responses through regulation of antiviral gene expression.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Ductal, Breast/genetics , Pseudogenes/physiology , RNA, Long Noncoding/metabolism , Tumor Escape/genetics , Animals , Breast/pathology , Breast/surgery , Breast Neoplasms/immunology , Breast Neoplasms/pathology , Breast Neoplasms/surgery , Carcinoma, Ductal, Breast/immunology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Ductal, Breast/surgery , Cell Line, Tumor , Cell Nucleus/genetics , Cytokines/genetics , Female , Gene Expression Regulation, Neoplastic/immunology , Gene Knockout Techniques , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Humans , Immunity, Innate/genetics , Mastectomy , Mice , Primary Cell Culture , RNA, Long Noncoding/genetics , Respirovirus Infections/immunology , Respirovirus Infections/virology , Sendai virus/immunology , Transcription Factor RelA/genetics , Xenograft Model Antitumor Assays
9.
Elife ; 92020 10 28.
Article in English | MEDLINE | ID: mdl-33112233

ABSTRACT

Long noncoding RNAs (lncRNAs) are often associated with polysomes, indicating coding potential. However, only a handful of endogenous proteins encoded by putative lncRNAs have been identified and assigned a function. Here, we report the discovery of a putative gastrointestinal-tract-specific lncRNA (LINC00675) that is regulated by the pioneer transcription factor FOXA1 and encodes a conserved small protein of 79 amino acids which we termed FORCP (FOXA1-Regulated Conserved Small Protein). FORCP transcript is undetectable in most cell types but is abundant in well-differentiated colorectal cancer (CRC) cells where it functions to inhibit proliferation, clonogenicity, and tumorigenesis. The epitope-tagged and endogenous FORCP protein predominantly localizes to the endoplasmic reticulum (ER). In response to ER stress, FORCP depletion results in decreased apoptosis. Our findings on the initial characterization of FORCP demonstrate that FORCP is a novel, conserved small protein encoded by a mis-annotated lncRNA that regulates apoptosis and tumorigenicity in well-differentiated CRC cells.


Subject(s)
Apoptosis/genetics , Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , RNA, Long Noncoding/metabolism , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/pathology , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum Stress , Gastrointestinal Tract/metabolism , Genes, Reporter , Hepatocyte Nuclear Factor 3-alpha/genetics , Humans , Molecular Sequence Annotation , Organ Specificity , RNA, Long Noncoding/genetics
10.
Elife ; 92020 10 27.
Article in English | MEDLINE | ID: mdl-33108271

ABSTRACT

Cell cycle is a cellular process that is subject to stringent control. In contrast to the wealth of knowledge of proteins controlling the cell cycle, very little is known about the molecular role of lncRNAs (long noncoding RNAs) in cell-cycle progression. By performing genome-wide transcriptome analyses in cell-cycle-synchronized cells, we observed cell-cycle phase-specific induction of >2000 lncRNAs. Further, we demonstrate that an S-phase-upregulated lncRNA, SUNO1, facilitates cell-cycle progression by promoting YAP1-mediated gene expression. SUNO1 facilitates the cell-cycle-specific transcription of WTIP, a positive regulator of YAP1, by promoting the co-activator, DDX5-mediated stabilization of RNA polymerase II on chromatin. Finally, elevated SUNO1 levels are associated with poor cancer prognosis and tumorigenicity, implying its pro-survival role. Thus, we demonstrate the role of a S-phase up-regulated lncRNA in cell-cycle progression via modulating the expression of genes controlling cell proliferation.


Subject(s)
Cell Proliferation/genetics , Co-Repressor Proteins/genetics , Cytoskeletal Proteins/genetics , DEAD-box RNA Helicases/genetics , Gene Expression Regulation , RNA, Long Noncoding/genetics , Signal Transduction/physiology , Co-Repressor Proteins/metabolism , Cytoskeletal Proteins/metabolism , DEAD-box RNA Helicases/metabolism , HCT116 Cells , HeLa Cells , Humans , RNA, Long Noncoding/metabolism , S Phase , Up-Regulation
11.
RNA ; 26(11): 1603-1620, 2020 11.
Article in English | MEDLINE | ID: mdl-32675111

ABSTRACT

Cellular quiescence and cell cycle reentry regulate vital biological processes such as cellular development and tissue homeostasis and are controlled by precise regulation of gene expression. The roles of long noncoding RNAs (lncRNAs) during these processes remain to be elucidated. By performing genome-wide transcriptome analyses, we identify differential expression of several hundreds of lncRNAs, including a significant number of the less-characterized class of microRNA-host-gene (MIRHG) lncRNAs or lnc-MIRHGs, during cellular quiescence and cell cycle reentry in human diploid fibroblasts. We observe that MIR222HG lncRNA displays serum-stimulated RNA processing due to enhanced splicing of the host nascent pri-MIR222HG transcript. The pre-mRNA splicing factor SRSF1 negatively regulates the microprocessor-catalyzed cleavage of pri-miR-222, thereby increasing the cellular pool of the mature MIR222HG Association of SRSF1 to pri-MIR222HG, including to a mini-exon, which partially overlaps with the primary miR-222 precursor, promotes serum-stimulated splicing over microRNA processing of MIR222HG Further, we observe that the increased levels of spliced MIR222HG in serum-stimulated cells promote the cell cycle reentry post quiescence in a microRNA-independent manner. MIR222HG interacts with DNM3OS, another lncRNA whose expression is elevated upon serum-stimulation, and promotes cell cycle reentry. The double-stranded RNA binding protein ILF3/2 complex facilitates MIR222HG:DNM3OS RNP complex assembly, thereby promoting DNM3OS RNA stability. Our study identifies a novel mechanism whereby competition between the splicing and microprocessor machinery modulates the serum-induced RNA processing of MIR222HG, which dictates cell cycle reentry.


Subject(s)
Gene Expression Profiling/methods , Lung/cytology , RNA, Long Noncoding/genetics , Serine-Arginine Splicing Factors/metabolism , Serum/chemistry , Cell Cycle , Cell Line , Fibroblasts/chemistry , Fibroblasts/cytology , HEK293 Cells , Humans , Lung/chemistry , Nuclear Factor 45 Protein/metabolism , Nuclear Factor 90 Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA Splicing , Sequence Analysis, RNA , Single Molecule Imaging , Up-Regulation , Exome Sequencing
12.
RNA ; 26(2): 175-185, 2020 02.
Article in English | MEDLINE | ID: mdl-31690584

ABSTRACT

Triple-negative breast cancer (TNBC) is one of the most aggressive breast cancer (BC) subtypes with a poor prognosis and high recurrence rate. Recent studies have identified vital roles played by several lncRNAs (long noncoding RNAs) in BC pathobiology. Cell type-specific expression of lncRNAs and their potential role in regulating the expression of oncogenic and tumor suppressor genes have made them promising cancer drug targets. By performing a transcriptome screen in an isogenic TNBC/basal subtype BC progression cell line model, we recently reported ∼1800 lncRNAs that display aberrant expression during breast cancer progression. Mechanistic studies on one such nuclear-retained lncRNA, linc02095, reveal that it promotes breast cancer proliferation by facilitating the expression of oncogenic transcription factor, SOX9. Both linc02095 and SOX9 display coregulated expression in BC patients as well in basal subtype BC cell lines. Knockdown of linc02095 results in decreased BC cell proliferation, whereas its overexpression promotes cells proliferation. Linc02095-depleted cells display reduced expression of SOX9 concomitant with reduced RNA polymerase II occupancy at the SOX9 gene body as well as defective SOX9 mRNA export, implying that linc02095 positively regulates SOX9 transcription and mRNA export. Finally, we identify a positive feedback loop in BC cells that controls the expression of both linc02095 and SOX9 Thus, our results unearth tumor-promoting activities of a nuclear lncRNA linc02095 by facilitating the expression of key oncogenic transcription factor in BC.


Subject(s)
Breast Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , RNA, Long Noncoding/genetics , SOX9 Transcription Factor/genetics , Triple Negative Breast Neoplasms/genetics , Breast/pathology , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Transformation, Neoplastic , Female , Gene Expression Profiling , Humans , Transcriptome , Triple Negative Breast Neoplasms/pathology , Up-Regulation
13.
PLoS Genet ; 14(11): e1007802, 2018 11.
Article in English | MEDLINE | ID: mdl-30496290

ABSTRACT

The human genome encodes thousands of long noncoding RNA (lncRNA) genes; the function of majority of them is poorly understood. Aberrant expression of a significant number of lncRNAs is observed in various diseases, including cancer. To gain insights into the role of lncRNAs in breast cancer progression, we performed genome-wide transcriptome analyses in an isogenic, triple negative breast cancer (TNBC/basal-like) progression cell lines using a 3D cell culture model. We identified significantly altered expression of 1853 lncRNAs, including ~500 natural antisense transcript (NATs) lncRNAs. A significant number of breast cancer-deregulated NATs displayed co-regulated expression with oncogenic and tumor suppressor protein-coding genes in cis. Further studies on one such NAT, PDCD4-AS1 lncRNA reveal that it positively regulates the expression and activity of the tumor suppressor PDCD4 in mammary epithelial cells. Both PDCD4-AS1 and PDCD4 show reduced expression in TNBC cell lines and in patients, and depletion of PDCD4-AS1 compromised the cellular levels and activity of PDCD4. Further, tumorigenic properties of PDCD4-AS1-depleted TNBC cells were rescued by exogenous expression of PDCD4, implying that PDCD4-AS1 acts upstream of PDCD4. Mechanistically, PDCD4-AS1 stabilizes PDCD4 RNA by forming RNA duplex and controls the interaction between PDCD4 RNA and RNA decay promoting factors such as HuR. Our studies demonstrate crucial roles played by NAT lncRNAs in regulating post-transcriptional gene expression of key oncogenic or tumor suppressor genes, thereby contributing to TNBC progression.


Subject(s)
Apoptosis Regulatory Proteins/genetics , RNA Stability , RNA, Antisense/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , RNA-Binding Proteins/genetics , Triple Negative Breast Neoplasms/genetics , Apoptosis Regulatory Proteins/antagonists & inhibitors , Apoptosis Regulatory Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Protein Binding , RNA, Antisense/metabolism , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , RNA-Binding Proteins/antagonists & inhibitors , RNA-Binding Proteins/metabolism , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
14.
Nucleic Acids Res ; 46(19): 10405-10416, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30102375

ABSTRACT

Long non-coding RNAs (lncRNAs) regulate vital biological processes, including cell proliferation, differentiation and development. A subclass of lncRNAs is synthesized from microRNA (miRNA) host genes (MIRHGs) due to pre-miRNA processing, and are categorized as miRNA-host gene lncRNAs (lnc-miRHGs). Presently, the cellular function of most lnc-miRHGs is not well understood. We demonstrate a miRNA-independent role for a nuclear-enriched lnc-miRHG in cell cycle progression. MIR100HG produces spliced and stable lncRNAs that display elevated levels during the G1 phase of the cell cycle. Depletion of MIR100HG-encoded lncRNAs in human cells results in aberrant cell cycle progression without altering the levels of miRNA encoded within MIR100HG. Notably, MIR100HG interacts with HuR/ELAVL1 as well as with several HuR-target mRNAs. Further, MIR100HG-depleted cells show reduced interaction between HuR and three of its target mRNAs, indicating that MIR100HG facilitates interaction between HuR and target mRNAs. Our studies have unearthed novel roles played by a MIRHG-encoded lncRNA in regulating RNA binding protein activity, thereby underscoring the importance of determining the function of several hundreds of lnc-miRHGs that are present in human genome.


Subject(s)
Cell Cycle/genetics , ELAV-Like Protein 1/genetics , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Cell Differentiation/genetics , Cell Division/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Humans , RNA, Messenger/genetics , RNA-Binding Proteins/genetics
15.
Trends Genet ; 34(2): 142-157, 2018 02.
Article in English | MEDLINE | ID: mdl-29249332

ABSTRACT

A significant portion of the human genome encodes genes that transcribe long nonprotein-coding RNAs (lncRNAs). A large number of lncRNAs localize in the nucleus, either enriched on the chromatin or localized to specific subnuclear compartments. Nuclear lncRNAs participate in several biological processes, including chromatin organization, and transcriptional and post-transcriptional gene expression, and also act as structural scaffolds of nuclear domains. Here, we highlight recent studies demonstrating the role of lncRNAs in regulating gene expression and nuclear organization in mammalian cells. In addition, we update current knowledge about the involvement of the most-abundant and conserved lncRNA, metastasis-associated lung adenocarcinoma transcript 1 (MALAT1), in gene expression control.


Subject(s)
Cell Nucleus/genetics , Chromatin/chemistry , Neoplasms/genetics , RNA, Long Noncoding/genetics , Transcription, Genetic , Animals , Cell Nucleus/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/genetics , Chromosomal Proteins, Non-Histone/metabolism , Humans , Neoplasms/metabolism , Neoplasms/pathology , Polycomb-Group Proteins/genetics , Polycomb-Group Proteins/metabolism , RNA Processing, Post-Transcriptional , RNA, Long Noncoding/classification , RNA, Long Noncoding/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , X Chromosome Inactivation
16.
J Cell Sci ; 130(24): 4180-4192, 2017 Dec 15.
Article in English | MEDLINE | ID: mdl-29133588

ABSTRACT

Nuclear speckles are self-assembled organelles composed of RNAs and proteins. They are proposed to act as structural domains that control distinct steps in gene expression, including transcription, splicing and mRNA export. Earlier studies identified differential localization of a few components within the speckles. It was speculated that the spatial organization of speckle components might contribute directly to the order of operations that coordinate distinct processes. Here, by performing multi-color structured illumination microscopy, we characterized the multilayer organization of speckles at a higher resolution. We found that SON and SC35 (also known as SRSF2) localize to the central region of the speckle, whereas MALAT1 and small nuclear (sn)RNAs are enriched at the speckle periphery. Coarse-grained simulations indicate that the non-random organization arises due to the interplay between favorable sequence-encoded intermolecular interactions of speckle-resident proteins and RNAs. Finally, we observe positive correlation between the total amount of RNA present within a speckle and the speckle size. These results imply that speckle size may be regulated to accommodate RNA accumulation and processing. Accumulation of RNA from various actively transcribed speckle-associated genes could contribute to the observed speckle size variations within a single cell.


Subject(s)
Cell Nucleus/genetics , DNA-Binding Proteins/genetics , Minor Histocompatibility Antigens/genetics , Organelles/genetics , RNA, Long Noncoding/genetics , Serine-Arginine Splicing Factors/genetics , Cell Nucleus/ultrastructure , Gene Expression Regulation , HeLa Cells , Humans , Organelles/ultrastructure , Proteins/genetics , RNA/genetics , RNA, Small Nucleolar/genetics
17.
Psychiatry Clin Neurosci ; 68(6): 456-62, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24417771

ABSTRACT

AIMS: Progressive cognitive decline has been an important issue in the treatment and care of patients with schizophrenia. Tyrosine hydroxylase (TH) is the rate-limiting enzyme for the biosynthesis of catecholamine, including dopamine and noradrenaline. In this report, we examined a possible association of a genetic variant in the TH promoter region. METHODS: Association of a genetic variant in the TH promoter region, C-824T (rs10770141), with intellectual ability in 132 patients with schizophrenia and 282 healthy subjects was examined. The transcriptional activity of the plasmids harboring the TH promoter region with either C or T nucleotide at -824 was assayed using a luciferase gene as a reporter. RESULTS: We found significant effects of the genotype on the full-scale IQ, verbal IQ, and performance IQ, in patients with schizophrenia. IQ was lower in individuals with the C/C genotype than those with T carriers. The plasmid with the T allele at -824 showed higher transcriptional activity than that with the C allele in a transient transfection experiment using a luciferase gene as a reporter, implying that the T carriers may have higher TH activities and retain higher levels of catecholamines in the brain. CONCLUSIONS: The present data suggest that the biosynthesis of catecholamine by the action of TH should be deeply involved in decreased intellectual ability in patients with schizophrenia. This is the first report, as far as we know, showing a correlation between TH expression and IQ in humans.


Subject(s)
Intelligence Tests , Intelligence/genetics , Schizophrenia/genetics , Schizophrenic Psychology , Tyrosine 3-Monooxygenase/genetics , Adult , Alleles , Female , Genotype , Humans , Male , Middle Aged , Polymorphism, Genetic/genetics , Polymorphism, Single Nucleotide/genetics , Transcription, Genetic
18.
Biochem Biophys Res Commun ; 442(1-2): 72-8, 2013 Dec 06.
Article in English | MEDLINE | ID: mdl-24220333

ABSTRACT

GTP cyclohydrolase I (GCH) catalyzes the first and rate limiting step reaction for the de novo synthesis of 5,6,7,8-tetrahydrobiopterin (BH4). The expression of GCH is dramatically elevated by immune activation, while the mechanism remains to be elucidated. In this study, we investigated the transcription mechanism of the GCH gene using lipopolysaccharide (LPS) to stimulate mouse macrophage RAW264.7 cells. With luciferase assay, we found a highly conserved enhancer region spanning approximately 300 bp in intron 1 of GCH gene as a response element to LPS stimulation. The same enhancer region was also responsible for the induction of the GCH gene by IFN-γ and TNF-α in HUVECs. With electrophoresis mobility shift assay (EMSA) and site directed mutation analysis, we identified two key fragments containing C/EBP and Ets binding motifs within the enhancer. Furthermore, C/EBP-ß was involved in LPS activated GCH transcription through direct binding to the enhancer shown by supershift, chromatin immunoprecipitation, and RNA interference experiments. In conclusion, our findings uncovered a novel mechanism of GCH transcriptional regulation by immune activation.


Subject(s)
Enhancer Elements, Genetic/immunology , GTP Cyclohydrolase/genetics , Gene Expression Regulation, Enzymologic , Transcriptional Activation , Animals , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic/drug effects , Enhancer Elements, Genetic/genetics , Human Umbilical Vein Endothelial Cells/immunology , Humans , Interferon-gamma/pharmacology , Lipopolysaccharides/immunology , Macrophage Activation/genetics , Macrophages/immunology , Mice , Proto-Oncogene Proteins c-ets/metabolism , Tumor Necrosis Factor-alpha/pharmacology
19.
J Biol Chem ; 288(19): 13748-61, 2013 May 10.
Article in English | MEDLINE | ID: mdl-23546882

ABSTRACT

BACKGROUND: MicroRNA (miR) dysregulation is found in Alzheimer disease (AD). A disintegrin and metalloprotease 10 (ADAM10) prevents generation of amyloid ß (Aß) and decrease AD pathology. RESULTS: miR-144 suppresses ADAM10 expression and is up-regulated by activator protein-1. CONCLUSION: miR-144 is a negative regulator of ADAM10 and may be involved in AD pathogenesis. SIGNIFICANCE: The first work to demonstrate the function of miRNA-144 and its regulation in the pathogenesis of AD. Amyloid ß-peptide (Aß) accumulating in the brain of Alzheimer disease (AD) patients is believed to be the main pathophysiologcal cause of the disease. Proteolytic processing of the amyloid precursor protein by α-secretase ADAM10 (a disintegrin and metalloprotease 10) protects the brain from the production of the Aß. Meanwhile, dysregulation or aberrant expression of microRNAs (miRNAs) has been widely documented in AD patients. In this study, we demonstrated that overexpression of miR-144, which was previously reported to be increased in elderly primate brains and AD patients, significantly decreased activity of the luciferase reporter containing the ADAM10 3'-untranslated region (3'-UTR) and suppressed the ADAM10 protein level, whereas the miR-144 inhibitor led to an increase of the luciferase activity. The negative regulation caused by miR-144 was strictly dependent on the binding of the miRNA to its recognition element in the ADAM10 3'-UTR. Moreover, we also showed that activator protein-1 regulates the transcription of miR-144 and the up-regulation of miR-144 at least partially induces the suppression of the ADAM10 protein in the presence of Aß. In addition, we found that miR-451, a miRNA processed from a single gene locus with miR-144, is also involved in the regulation of ADAM10 expression. Taken together, our data therefore demonstrate miR-144/451 is a negative regulator of the ADAM10 protein and suggest a mechanistic role for miR-144/451 in AD pathogenesis.


Subject(s)
ADAM Proteins/metabolism , Alzheimer Disease/enzymology , Amyloid Precursor Protein Secretases/metabolism , Membrane Proteins/metabolism , MicroRNAs/physiology , RNA Interference , Transcription Factor AP-1/metabolism , 3' Untranslated Regions , ADAM Proteins/genetics , ADAM10 Protein , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/physiology , Base Sequence , Binding Sites , Enzyme Repression , Genes, Reporter , HeLa Cells , Humans , Membrane Proteins/genetics , MicroRNAs/metabolism , Promoter Regions, Genetic , Response Elements , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...