Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Mol Cancer ; 22(1): 204, 2023 12 13.
Article in English | MEDLINE | ID: mdl-38093367

ABSTRACT

Lung squamous cell carcinoma (LUSC) is associated with high mortality and limited targeted therapies. USP13 is one of the most amplified genes in LUSC, yet its role in lung cancer is largely unknown. Here, we established a novel mouse model of LUSC by overexpressing USP13 on KrasG12D/+; Trp53flox/flox background (KPU). KPU-driven lung squamous tumors faithfully recapitulate key pathohistological, molecular features, and cellular pathways of human LUSC. We found that USP13 altered lineage-determining factors such as NKX2-1 and SOX2 in club cells of the airway and reinforced the fate of club cells to squamous carcinoma development. We showed a strong molecular association between USP13 and c-MYC, leading to the upregulation of squamous programs in murine and human lung cancer cells. Collectively, our data demonstrate that USP13 is a molecular driver of lineage plasticity in club cells and provide mechanistic insight that may have potential implications for the treatment of LUSC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Lung Neoplasms , Animals , Humans , Mice , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/pathology , Cell Lineage , Lung/metabolism , Lung Neoplasms/pathology , Ubiquitin-Specific Proteases
2.
Front Mol Biosci ; 9: 912727, 2022.
Article in English | MEDLINE | ID: mdl-35874614

ABSTRACT

DEAD-Box Helicase 3 X-Linked (DDX3X) is essential for RNA metabolism and participates in various cellular processes involving RNA. DDX3X has been implicated in cancer growth and metastasis. DDX3X is involved in antiviral responses for viral RNAs and contributes to pro- or anti-microbial responses. A better understanding of how human cells regulate innate immune response against the viral "non-self" double-stranded RNAs (dsRNAs) and endogenous viral-like "self" dsRNAs is critical to understanding innate immune sensing, anti-microbial immunity, inflammation, immune cell homeostasis, and developing novel therapeutics for infectious, immune-mediated diseases, and cancer. DDX3X has known for activating the viral dsRNA-sensing pathway and innate immunity. However, accumulating research reveals a more complex role of DDX3X in regulating dsRNA-mediated signaling in cells. Here, we discuss the role of DDX3X in viral dsRNA- or endogenous dsRNA-mediated immune signaling pathways.

3.
Oncogene ; 41(13): 1974-1985, 2022 03.
Article in English | MEDLINE | ID: mdl-35173307

ABSTRACT

Epithelial ovarian cancer is the most lethal gynecologic malignancy and one of the most common causes of cancer mortality among women worldwide. Ubiquitin-Specific Peptidase 13 (USP13) gene copy is strongly amplified in human epithelial ovarian cancer, and high USP13 expression is correlated with poor survival outcomes. Yet, its pathological contribution to ovarian tumorigenesis remains unknown. We crossed a conditional Usp13 overexpressing knock-in mouse with a conditional knockout of Trp53 and Pten mouse and generated a novel ovarian cancer genetically engineered mouse model (GEMM), which closely recapitulates the genetic changes driving ovarian cancer in humans. Overexpression of USP13 with deletion of Trp53 and Pten in murine ovarian surface epithelium accelerated ovarian tumorigenesis and led to decreased survival in mice. Notably, USP13 greatly enhanced peritoneal metastasis of ovarian tumors with frequent development of hemorrhagic ascites. The primary and metastatic tumors exhibited morphology and clinical behavior similar to human high-grade serous ovarian cancer. Co-inhibition of USP13 and AKT significantly decreased the viability of the primary murine ovarian cancer cells isolated from the GEMM. USP13 also increased the tumorigenic and metastatic abilities of primary murine ovarian cancer cells in a syngeneic mouse study. These findings suggest a critical role of USP13 in ovarian cancer development and reveal USP13 as a potential therapeutic target for ovarian cancer.


Subject(s)
Ovarian Neoplasms , Ubiquitin-Specific Proteases , Animals , Carcinogenesis/genetics , Carcinoma, Ovarian Epithelial , Disease Models, Animal , Female , Humans , Mice , Ovarian Neoplasms/pathology , Ubiquitin-Specific Proteases/genetics
4.
Cancers (Basel) ; 15(1)2022 Dec 29.
Article in English | MEDLINE | ID: mdl-36612196

ABSTRACT

Ubiquitin-specific Peptidase 13 (USP13) is a deubiquitinating enzyme that regulates the stability or function of its substrate. USP13 is highly amplified in human ovarian cancer, and elevated expression of USP13 promotes tumorigenesis and metastasis of ovarian cancer. However, there is little known about USP13 post-translational modifications and their role in ovarian cancer. Here, we found that USP13 is phosphorylated at Thr122 in ovarian cancer cells. Phosphorylated Thr122 (pT122) on endogenous USP13 was observed in most human ovarian cancer cells, and the abundance of this phosphorylation was correlated to the total level of USP13. We further demonstrated that Casein kinase 2 (CK2) directly interacts with and phosphorylates USP13 at Thr122, which promotes the stability of USP13 protein. Finally, we showed that Threonine 122 is important for cell proliferation of ovarian cancer cells. Our findings may reveal a novel regulatory mechanism for USP13, which may lead to novel therapeutic targeting of USP13 in ovarian cancer.

6.
Cancer Res ; 81(13): 3607-3620, 2021 07 01.
Article in English | MEDLINE | ID: mdl-33941613

ABSTRACT

Induction of nucleic acid sensing-mediated type I interferon (IFN) has emerged as a novel approach to activate the immune system against cancer. Here we show that the depletion of DEAD-box RNA helicase 3X (DDX3X) triggers a tumor-intrinsic type I IFN response in breast cancer cells. Depletion or inhibition of DDX3X activity led to aberrant cytoplasmic accumulation of cellular endogenous double-stranded RNAs (dsRNA), which triggered type I IFN production through the melanoma differentiation-associated gene 5 (MDA5)-mediated dsRNA-sensing pathway. Furthermore, DDX3X interacted with dsRNA-editing ADAR1 and dual depletion of DDX3X and ADAR1 synergistically activated the cytosolic dsRNA pathway in breast cancer cells. Loss of DDX3X in mouse mammary tumors enhanced antitumor activity by increasing the tumor-intrinsic type I IFN response, antigen presentation, and tumor infiltration of cytotoxic T and dendritic cells. These findings may lead to the development of a novel therapeutic approach for breast cancer by targeting DDX3X in combination with immune-checkpoint blockade. SIGNIFICANCE: This study elucidates the novel role of DDX3X in regulating endogenous cellular dsRNA homeostasis and type I IFN signaling in breast cancer. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/81/13/3607/F1.large.jpg.


Subject(s)
Breast Neoplasms/immunology , DEAD-box RNA Helicases/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Immunity, Innate/immunology , Interferon Type I/immunology , Interferon-Induced Helicase, IFIH1/metabolism , RNA, Double-Stranded/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Breast Neoplasms/prevention & control , Cell Proliferation , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Humans , Interferon Type I/metabolism , Interferon-Induced Helicase, IFIH1/genetics , Mice , Mice, Inbred BALB C , Mice, Nude , Prognosis , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
J Biol Chem ; 296: 100194, 2021.
Article in English | MEDLINE | ID: mdl-33334891

ABSTRACT

Cohesin is a multiprotein ring complex that regulates 3D genome organization, sister chromatid cohesion, gene expression, and DNA repair. Cohesin is known to be ubiquitinated, although the mechanism, regulation, and effects of cohesin ubiquitination remain poorly defined. We previously used gene editing to introduce a dual epitope tag into the endogenous allele of each of 11 known components of cohesin in human HCT116 cells. Here we report that mass spectrometry analysis of dual-affinity purifications identified the USP13 deubiquitinase as a novel cohesin-interacting protein. Subsequent immunoprecipitation/Western blots confirmed the endogenous interaction in HCT116, 293T, HeLa, and RPE-hTERT cells; demonstrated that the interaction occurs specifically in the soluble nuclear fraction (not in the chromatin); requires the ubiquitin-binding domains (UBA1/2) of USP13; and occurs preferentially during DNA replication. Reciprocal dual-affinity purification of endogenous USP13 followed by mass spectrometry demonstrated that cohesin is its primary interactor in the nucleus. Ectopic expression and CRISPR knockout of USP13 showed that USP13 is paradoxically required for both deubiquitination and ubiquitination of cohesin subunits in human cells. USP13 was dispensable for sister chromatid cohesion in HCT116 and HeLa cells, whereas it was required for the dissociation of cohesin from chromatin as cells transit through mitosis. Together these results identify USP13 as a new cohesin-interacting protein that regulates the ubiquitination of cohesin and its cell cycle regulated interaction with chromatin.


Subject(s)
Cell Cycle Proteins/metabolism , Chromatin/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Ubiquitin-Specific Proteases/metabolism , Ubiquitin/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Chromatin/genetics , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/genetics , Chromosome Segregation , DNA Repair , DNA Replication , HCT116 Cells , HeLa Cells , Humans , Protein Interaction Domains and Motifs , Ubiquitin-Specific Proteases/chemistry , Ubiquitin-Specific Proteases/genetics , Ubiquitination , Cohesins
8.
Nat Commun ; 9(1): 4718, 2018 11 09.
Article in English | MEDLINE | ID: mdl-30413718

ABSTRACT

Chromosome 17q23 amplification occurs in ~11% of human breast cancers. Enriched in HER2+ breast cancers, the 17q23 amplification is significantly correlated with poor clinical outcomes. In addition to the previously identified oncogene WIP1, we uncover an oncogenic microRNA gene, MIR21, in a majority of the WIP1-containing 17q23 amplicons. The 17q23 amplification results in aberrant expression of WIP1 and miR-21, which not only promotes breast tumorigenesis, but also leads to resistance to anti-HER2 therapies. Inhibiting WIP1 and miR-21 selectively inhibits the proliferation, survival and tumorigenic potential of the HER2+ breast cancer cells harboring 17q23 amplification. To overcome the resistance of trastuzumab-based therapies in vivo, we develop pH-sensitive nanoparticles for specific co-delivery of the WIP1 and miR-21 inhibitors into HER2+ breast tumors, leading to a profound reduction of tumor growth. These results demonstrate the great potential of the combined treatment of WIP1 and miR-21 inhibitors for the trastuzumab-resistant HER2+ breast cancers.


Subject(s)
Breast Neoplasms/genetics , Chromosomes, Human, Pair 17/genetics , Drug Resistance, Neoplasm/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DEAD-box RNA Helicases/metabolism , Drug Delivery Systems , Drug Resistance, Neoplasm/drug effects , Female , Gene Amplification/drug effects , Humans , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Nanoparticles/chemistry , Protein Phosphatase 2C/genetics , Protein Phosphatase 2C/metabolism , Receptor, ErbB-2/metabolism , Trastuzumab/pharmacology , Trastuzumab/therapeutic use
9.
Mol Cell Oncol ; 5(5): e1285384, 2018.
Article in English | MEDLINE | ID: mdl-30263935

ABSTRACT

Our recent studies determined molecular interactions between genes in the ubiquitin-proteasome pathways and cancer cell metabolism. Ubiquitin-specific peptidase 13 (USP13) specifically deubiquitinates and thus upregulates ATP citrate lyase and oxoglutarate dehydrogenase that drive ovarian cancer metabolism. These findings may lead to the development of USP13 inhibitors and new-targeted therapies in ovarian cancers.

10.
Nat Commun ; 7: 13525, 2016 11 28.
Article in English | MEDLINE | ID: mdl-27892457

ABSTRACT

Dysregulated energetic metabolism has been recently identified as a hallmark of cancer. Although mutations in metabolic enzymes hardwire metabolism to tumourigenesis, they are relatively infrequent in ovarian cancer. More often, cancer metabolism is re-engineered by altered abundance and activity of the metabolic enzymes. Here we identify ubiquitin-specific peptidase 13 (USP13) as a master regulator that drives ovarian cancer metabolism. USP13 specifically deubiquitinates and thus upregulates ATP citrate lyase and oxoglutarate dehydrogenase, two key enzymes that determine mitochondrial respiration, glutaminolysis and fatty acid synthesis. The USP13 gene is co-amplified with PIK3CA in 29.3% of high-grade serous ovarian cancers and its overexpression is significantly associated with poor clinical outcome. Inhibiting USP13 remarkably suppresses ovarian tumour progression and sensitizes tumour cells to the treatment of PI3K/AKT inhibitor. Our results reveal an important metabolism-centric role of USP13, which may lead to potential therapeutics targeting USP13 in ovarian cancers.


Subject(s)
Endopeptidases/genetics , Gene Amplification , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Carcinogenesis/drug effects , Carcinogenesis/genetics , Carcinogenesis/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Endopeptidases/metabolism , Female , Gene Knockdown Techniques , Genome, Human , Glutamine/metabolism , HEK293 Cells , Humans , Lipids/biosynthesis , Mitochondria/drug effects , Mitochondria/metabolism , Neoplasm Metastasis , Ovarian Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Protein Stability/drug effects , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Proto-Oncogene Proteins c-akt/metabolism , Substrate Specificity/drug effects , Ubiquitin-Specific Proteases , Ubiquitination/drug effects
11.
Cell Metab ; 24(5): 685-700, 2016 11 08.
Article in English | MEDLINE | ID: mdl-27829138

ABSTRACT

Reactive stromal cells are an integral part of tumor microenvironment (TME) and interact with cancer cells to regulate their growth. Although targeting stromal cells could be a viable therapy to regulate the communication between TME and cancer cells, identification of stromal targets that make cancer cells vulnerable has remained challenging and elusive. Here, we identify a previously unrecognized mechanism whereby metabolism of reactive stromal cells is reprogrammed through an upregulated glutamine anabolic pathway. This dysfunctional stromal metabolism confers atypical metabolic flexibility and adaptive mechanisms in stromal cells, allowing them to harness carbon and nitrogen from noncanonical sources to synthesize glutamine in nutrient-deprived conditions existing in TME. Using an orthotopic mouse model for ovarian carcinoma, we find that co-targeting glutamine synthetase in stroma and glutaminase in cancer cells reduces tumor weight, nodules, and metastasis. We present a synthetic lethal approach to target tumor stroma and cancer cells simultaneously for desirable therapeutic outcomes.


Subject(s)
Glutamate-Ammonia Ligase/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Tumor Microenvironment , Amino Acids/metabolism , Animals , Aspartate Aminotransferases/metabolism , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Carbon/metabolism , Cell Line, Tumor , Cell Proliferation , Citric Acid Cycle , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial Cells/pathology , Humans , Metabolome , Mice, Nude , Nitrogen/metabolism , Nucleotides/metabolism , Stromal Cells/enzymology , Up-Regulation
12.
Bioessays ; 37(12): 1277-86, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26445307

ABSTRACT

Cancer drugs are broadly classified into two categories: cytotoxic chemotherapies and targeted therapies that specifically modulate the activity of one or more proteins involved in cancer. Major advances have been achieved in targeted cancer therapies in the past few decades, which is ascribed to the increasing understanding of molecular mechanisms for cancer initiation and progression. Consequently, monoclonal antibodies and small molecules have been developed to interfere with a specific molecular oncogenic target. Targeting gain-of-function mutations, in general, has been productive. However, it has been a major challenge to use standard pharmacologic approaches to target loss-of-function mutations of tumor suppressor genes. Novel approaches, including synthetic lethality and collateral vulnerability screens, are now being developed to target gene defects in p53, PTEN, and BRCA1/2. Here, we review and summarize the recent findings in cancer genomics, drug development, and molecular cancer biology, which show promise in targeting tumor suppressors in cancer therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Genes, Tumor Suppressor/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Drug Discovery/methods , Genomics/methods , Humans , Mutation/drug effects
13.
Biol Reprod ; 93(3): 70, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26246218

ABSTRACT

The mammalian epididymis is a highly convoluted tubule that connects the testis to the vas deferens. Its proper functions in sperm transport, storage, and maturation are essential for male reproduction. One of the genes predominantly expressed in the epididymis is ADAM7 (a disintegrin and metalloprotease 7). Previous studies have shown that ADAM7 synthesized in the epididymis is secreted into the epididymal lumen and is then transferred to sperm membranes, where it forms a chaperone complex that is potentially involved in sperm fertility. In this study, we generated and analyzed mice with a targeted disruption in the Adam7 gene. We found that the fertility of male mice was modestly but significantly reduced by knockout of Adam7. Histological analyses revealed that the cell heights of the epithelium were dramatically decreased in the caput of the epididymis of Adam7-null mice, suggesting a requirement for ADAM7 in maintaining the integrity of the epididymal epithelium. We found that sperm from Adam7-null mice exhibit decreased motility, tail deformation, and altered tyrosine phosphorylation, indicating that the absence of ADAM7 leads to abnormal sperm functions and morphology. Western blot analyses revealed reduced levels of integral membrane protein 2B (ITM2B) and ADAM2 in sperm from Adam7-null mice, suggesting a requirement for ADAM7 in normal expression of sperm membrane proteins involved in sperm functions. Collectively, our study demonstrates for the first time that ADAM7 is required for normal fertility and is important for the maintenance of epididymal integrity and for sperm morphology, motility, and membrane proteins.


Subject(s)
ADAM Proteins/genetics , Epididymis/pathology , Infertility, Male/genetics , Infertility, Male/pathology , Membrane Proteins/genetics , Spermatozoa/pathology , Adaptor Proteins, Signal Transducing , Animals , Epithelial Cells/pathology , Epithelial Cells/ultrastructure , Female , Male , Mice , Mice, Knockout , Sperm Capacitation/genetics , Sperm Head/pathology , Sperm Motility/genetics , Sperm Tail/pathology
14.
Nature ; 520(7549): 697-701, 2015 Apr 30.
Article in English | MEDLINE | ID: mdl-25901683

ABSTRACT

TP53, a well-known tumour suppressor gene that encodes p53, is frequently inactivated by mutation or deletion in most human tumours. A tremendous effort has been made to restore p53 activity in cancer therapies. However, no effective p53-based therapy has been successfully translated into clinical cancer treatment owing to the complexity of p53 signalling. Here we demonstrate that genomic deletion of TP53 frequently encompasses essential neighbouring genes, rendering cancer cells with hemizygous TP53 deletion vulnerable to further suppression of such genes. POLR2A is identified as such a gene that is almost always co-deleted with TP53 in human cancers. It encodes the largest and catalytic subunit of the RNA polymerase II complex, which is specifically inhibited by α-amanitin. Our analysis of The Cancer Genome Atlas (TCGA) and Cancer Cell Line Encyclopedia (CCLE) databases reveals that POLR2A expression levels are tightly correlated with its gene copy numbers in human colorectal cancer. Suppression of POLR2A with α-amanitin or small interfering RNAs selectively inhibits the proliferation, survival and tumorigenic potential of colorectal cancer cells with hemizygous TP53 loss in a p53-independent manner. Previous clinical applications of α-amanitin have been limited owing to its liver toxicity. However, we found that α-amanitin-based antibody-drug conjugates are highly effective therapeutic agents with reduced toxicity. Here we show that low doses of α-amanitin-conjugated anti-epithelial cell adhesion molecule (EpCAM) antibody lead to complete tumour regression in mouse models of human colorectal cancer with hemizygous deletion of POLR2A. We anticipate that inhibiting POLR2A will be a new therapeutic approach for human cancers containing such common genomic alterations.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Genes, p53/genetics , Tumor Suppressor Protein p53/deficiency , Alpha-Amanitin/adverse effects , Alpha-Amanitin/chemistry , Alpha-Amanitin/pharmacology , Alpha-Amanitin/therapeutic use , Animals , Antibodies/chemistry , Antibodies/immunology , Antigens, Neoplasm/immunology , Catalytic Domain , Cell Adhesion Molecules/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Databases, Genetic , Disease Models, Animal , Epithelial Cell Adhesion Molecule , Female , Gene Deletion , Gene Dosage/genetics , Humans , Immunoconjugates/adverse effects , Immunoconjugates/chemistry , Immunoconjugates/immunology , Immunoconjugates/therapeutic use , Mice , Protein Subunits/chemistry , Protein Subunits/deficiency , Protein Subunits/genetics , RNA Polymerase II/antagonists & inhibitors , RNA Polymerase II/chemistry , RNA Polymerase II/deficiency , RNA Polymerase II/genetics , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays
15.
Cell Rep ; 8(5): 1447-60, 2014 Sep 11.
Article in English | MEDLINE | ID: mdl-25176654

ABSTRACT

Posttranscriptional maturation is a critical step in microRNA (miRNA) biogenesis that determines mature miRNA levels. In addition to core components (Drosha and DGCR8 [DiGeorge syndrome critical region gene 8]) in the microprocessor, regulatory RNA-binding proteins may confer the specificity for recruiting and processing of individual primary miRNAs (pri-miRNAs). Here, we identify DDX1 as a regulatory protein that promotes the expression of a subset of miRNAs, including five members in the microRNA-200 (miR-200) family and four miRNAs in an eight-miRNA signature of a mesenchymal ovarian cancer subtype. A majority of DDX1-dependent miRNAs are induced after DNA damage. This induction is facilitated by the ataxia telangiectasia mutated (ATM)-mediated phosphorylation of DDX1. Inhibiting DDX1 promotes ovarian tumor growth and metastasis in a syngeneic mouse model. Analysis of The Cancer Genome Atlas (TCGA) reveals that low DDX1 levels are associated with poor clinical outcome in patients with serous ovarian cancer. These findings suggest that DDX1 is a key modulator in miRNA maturation and ovarian tumor suppression.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , DEAD-box RNA Helicases/metabolism , MicroRNAs/metabolism , Ovarian Neoplasms/metabolism , Adenocarcinoma/diagnosis , Animals , Biomarkers, Tumor/genetics , Cell Line, Tumor , DEAD-box RNA Helicases/genetics , DNA Damage , Female , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , MicroRNAs/genetics , Ovarian Neoplasms/diagnosis
16.
Antioxid Redox Signal ; 20(4): 655-77, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-23879367

ABSTRACT

SIGNIFICANCE: The well-studied sequences in the human genome are those of protein-coding genes, which account for only 1%-2% of the total genome. However, with the advent of high-throughput transcriptome sequencing technology, we now know that about 90% of our genome is extensively transcribed and that the vast majority of them are transcribed into noncoding RNAs (ncRNAs). It is of great interest and importance to decipher the functions of these ncRNAs in humans. RECENT ADVANCES: In the last decade, it has become apparent that ncRNAs play a crucial role in regulating gene expression in normal development, in stress responses to internal and environmental stimuli, and in human diseases. CRITICAL ISSUES: In addition to those constitutively expressed structural RNA, such as ribosomal and transfer RNAs, regulatory ncRNAs can be classified as microRNAs (miRNAs), Piwi-interacting RNAs (piRNAs), small interfering RNAs (siRNAs), small nucleolar RNAs (snoRNAs), and long noncoding RNAs (lncRNAs). However, little is known about the biological features and functional roles of these ncRNAs in DNA repair and genome instability, although a number of miRNAs and lncRNAs are regulated in the DNA damage response. FUTURE DIRECTIONS: A major goal of modern biology is to identify and characterize the full profile of ncRNAs with regard to normal physiological functions and roles in human disorders. Clinically relevant ncRNAs will also be evaluated and targeted in therapeutic applications.


Subject(s)
DNA Repair , Genomic Instability , RNA, Untranslated/physiology , Animals , DNA Damage , Humans , Neoplasms/genetics , RNA Interference , RNA Transport , Reactive Oxygen Species/metabolism , Signal Transduction
17.
EMBO J ; 32(21): 2833-47, 2013 Oct 30.
Article in English | MEDLINE | ID: mdl-24097061

ABSTRACT

A prompt and efficient DNA damage response (DDR) eliminates the detrimental effects of DNA lesions in eukaryotic cells. Basic and preclinical studies suggest that the DDR is one of the primary anti-cancer barriers during tumorigenesis. The DDR involves a complex network of processes that detect and repair DNA damage, in which long non-coding RNAs (lncRNAs), a new class of regulatory RNAs, may play an important role. In the current study, we identified a novel lncRNA, lncRNA-JADE, that is induced after DNA damage in an ataxia-telangiectasia mutated (ATM)-dependent manner. LncRNA-JADE transcriptionally activates Jade1, a key component in the HBO1 (human acetylase binding to ORC1) histone acetylation complex. Consequently, lncRNA-JADE induces histone H4 acetylation in the DDR. Markedly higher levels of lncRNA-JADE were observed in human breast tumours in comparison with normal breast tissues. Knockdown of lncRNA-JADE significantly inhibited breast tumour growth in vivo. On the basis of these results, we propose that lncRNA-JADE is a key functional link that connects the DDR to histone H4 acetylation, and that dysregulation of lncRNA-JADE may contribute to breast tumorigenesis.


Subject(s)
Breast Neoplasms/genetics , DNA Damage , Histones/metabolism , Homeodomain Proteins/genetics , RNA, Long Noncoding/genetics , Tumor Suppressor Proteins/genetics , Acetylation , Animals , Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation , Female , Gene Silencing , HEK293 Cells , Humans , MCF-7 Cells , Mice , Mice, SCID , NIH 3T3 Cells , Tissue Array Analysis
18.
Cell Rep ; 3(6): 2100-12, 2013 Jun 27.
Article in English | MEDLINE | ID: mdl-23791529

ABSTRACT

Expression of microRNAs (miRNAs) involves transcription of miRNA genes and maturation of the primary transcripts. Recent studies have shown that posttranscriptional processing of primary and precursor miRNAs is induced after DNA damage through regulatory RNA-binding proteins in the Drosha and Dicer complexes, such as DDX5 and KSRP. However, little is known about the regulation of nuclear export of pre-miRNAs in the DNA-damage response, a critical step in miRNA maturation. Here, we show that nuclear export of pre-miRNAs is accelerated after DNA damage in an ATM-dependent manner. The ATM-activated AKT kinase phosphorylates Nup153, a key component of the nucleopore, leading to enhanced interaction between Nup153 and Exportin-5 (XPO5) and increased nuclear export of pre-miRNAs. These findings define an important role of DNA-damage signaling in miRNA transport and maturation.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , DNA Damage , MicroRNAs/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Active Transport, Cell Nucleus , Amino Acid Sequence , Animals , Cell Line, Tumor , Cell Nucleus/metabolism , HCT116 Cells , Humans , Karyopherins/genetics , Karyopherins/metabolism , MicroRNAs/biosynthesis , MicroRNAs/genetics , Molecular Sequence Data , Mutation , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/genetics , Sequence Homology, Amino Acid , Signal Transduction
19.
Cell Mol Life Sci ; 69(17): 2895-906, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22430204

ABSTRACT

MicroRNAs (miRNAs) are a family of small, non-coding RNAs that control gene expression at the post-transcriptional level by destabilizing and inhibiting translation of their target messenger RNAs. MiRNAs are involved in the regulation of a number of fundamental biological processes, and their dysregulation is thought to contribute to several disease processes. Emerging evidence suggests that miRNAs also play a critical role in protecting the heritable genome by contributing to the regulation of the DNA damage response. Consequently, much recent investigative effort has been directed towards an improved understanding of how miRNAs are regulated in response to DNA damage. In this review, we discuss the most recent findings regarding the regulation of miRNA expression and the functional roles of miRNAs in the DNA damage response.


Subject(s)
Cell Physiological Phenomena , DNA Damage/genetics , Gene Expression Regulation , MicroRNAs/genetics , Animals , Humans
20.
J Cell Physiol ; 226(5): 1186-95, 2011 May.
Article in English | MEDLINE | ID: mdl-20945367

ABSTRACT

In mammals, sperm acquire their motility and ability to fertilize eggs in the epididymis. This maturation process involves the acquisition of particular proteins from the epididymis. One such secretory protein specifically expressed in the epididymis is Adam7 (a disintegrin and metalloprotease 7). Previous studies have shown that Adam7 that resides in an intracellular compartment of epididymal cells is transferred to sperm membranes, where its levels are dependent on the expression of Adam2 and Adam3, which have critical roles in fertilization. Here, using a proteomics approach based on mass spectrometry, we identified proteins that interact with Adam7 in sperm membranes. This analysis revealed that Adam7 forms complexes with calnexin (Canx), heat shock protein 5 (Hspa5), and integral membrane protein 2B (Itm2b). Canx and Hspa5 are molecular chaperones, and Itm2b is a type II integral membrane protein implicated in neurodegeneration. The interaction of Adam7 with these proteins was confirmed by immunoprecipitation-Western blot analysis. We found that Adam7 and Itm2b are located in detergent-resistant regions known to be highly correlated with membrane lipid rafts. We further found that the association of Adam7 with Itm2b is remarkably promoted during sperm capacitation owing to a conformational change of Adam7 that occurs in concert with the capacitation process. Thus, our results suggest that Adam7 functions in fertilization through the formation of a chaperone complex and enhanced association with Itm2b during capacitation in sperm.


Subject(s)
ADAM Proteins/metabolism , Calnexin/metabolism , Cell Membrane/metabolism , Heat-Shock Proteins/metabolism , Membrane Proteins/metabolism , Sperm Capacitation , Spermatozoa/metabolism , Adaptor Proteins, Signal Transducing , Animals , Blotting, Western , Endoplasmic Reticulum Chaperone BiP , Immunoprecipitation , Male , Mice , Protein Binding , Proteomics/methods , Signal Transduction , Tandem Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...