Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
1.
Nat Rev Mol Cell Biol ; 24(2): 123-141, 2023 02.
Article in English | MEDLINE | ID: mdl-36104626

ABSTRACT

PIWI-interacting RNAs (piRNAs) are a class of small non-coding RNAs that associate with proteins of the PIWI clade of the Argonaute family. First identified in animal germ line cells, piRNAs have essential roles in germ line development. The first function of PIWI-piRNA complexes to be described was the silencing of transposable elements, which is crucial for maintaining the integrity of the germ line genome. Later studies provided new insights into the functions of PIWI-piRNA complexes by demonstrating that they regulate protein-coding genes. Recent studies of piRNA biology, including in new model organisms such as golden hamsters, have deepened our understanding of both piRNA biogenesis and piRNA function. In this Review, we discuss the most recent advances in our understanding of piRNA biogenesis, the molecular mechanisms of piRNA function and the emerging roles of piRNAs in germ line development mainly in flies and mice, and in infertility, cancer and neurological diseases in humans.


Subject(s)
Argonaute Proteins , Piwi-Interacting RNA , Animals , Mice , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , DNA Transposable Elements , Germ Cells/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
2.
Cell ; 180(6): 1212-1227.e14, 2020 03 19.
Article in English | MEDLINE | ID: mdl-32169215

ABSTRACT

The paternal genome undergoes a massive exchange of histone with protamine for compaction into sperm during spermiogenesis. Upon fertilization, this process is potently reversed, which is essential for parental genome reprogramming and subsequent activation; however, it remains poorly understood how this fundamental process is initiated and regulated. Here, we report that the previously characterized splicing kinase SRPK1 initiates this life-beginning event by catalyzing site-specific phosphorylation of protamine, thereby triggering protamine-to-histone exchange in the fertilized oocyte. Interestingly, protamine undergoes a DNA-dependent phase transition to gel-like condensates and SRPK1-mediated phosphorylation likely helps open up such structures to enhance protamine dismissal by nucleoplasmin (NPM2) and enable the recruitment of HIRA for H3.3 deposition. Remarkably, genome-wide assay for transposase-accessible chromatin sequencing (ATAC-seq) analysis reveals that selective chromatin accessibility in both sperm and MII oocytes is largely erased in early pronuclei in a protamine phosphorylation-dependent manner, suggesting that SRPK1-catalyzed phosphorylation initiates a highly synchronized reorganization program in both parental genomes.


Subject(s)
Chromatin/metabolism , Protamines/metabolism , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Cycle Proteins/metabolism , Cell Nucleus/metabolism , Chromatin/physiology , Chromatin Assembly and Disassembly/genetics , Chromatin Assembly and Disassembly/physiology , Fertilization/genetics , Histones/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Oocytes/metabolism , Oocytes/physiology , Phosphorylation , Protamine Kinase/genetics , Protamine Kinase/metabolism , Protamines/genetics , Protein Serine-Threonine Kinases/physiology , RNA Splicing/genetics , RNA Splicing/physiology , Spermatozoa/metabolism , Transcription Factors/metabolism , Zygote/metabolism
3.
Cell ; 179(7): 1566-1581.e16, 2019 12 12.
Article in English | MEDLINE | ID: mdl-31835033

ABSTRACT

Spermiogenesis is a highly orchestrated developmental process during which chromatin condensation decouples transcription from translation. Spermiogenic mRNAs are transcribed earlier and stored in a translationally inert state until needed for translation; however, it remains largely unclear how such repressed mRNAs become activated during spermiogenesis. We previously reported that the MIWI/piRNA machinery is responsible for mRNA elimination during late spermiogenesis in preparation for spermatozoa production. Here we unexpectedly discover that the same machinery is also responsible for activating translation of a subset of spermiogenic mRNAs to coordinate with morphological transformation into spermatozoa. Such action requires specific base-pairing interactions of piRNAs with target mRNAs in their 3' UTRs, which activates translation through coupling with cis-acting AU-rich elements to nucleate the formation of a MIWI/piRNA/eIF3f/HuR super-complex in a developmental stage-specific manner. These findings reveal a critical role of the piRNA system in translation activation, which we show is functionally required for spermatid development.


Subject(s)
Argonaute Proteins/metabolism , Peptide Chain Initiation, Translational , RNA, Small Interfering/metabolism , Spermatogenesis , 3' Untranslated Regions , Animals , Argonaute Proteins/genetics , Base Pairing , Cells, Cultured , ELAV-Like Protein 1/metabolism , Eukaryotic Initiation Factor-3/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Small Interfering/genetics
4.
Cell ; 169(6): 1090-1104.e13, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28552346

ABSTRACT

Genetic studies have elucidated critical roles of Piwi proteins in germline development in animals, but whether Piwi is an actual disease gene in human infertility remains unknown. We report germline mutations in human Piwi (Hiwi) in patients with azoospermia that prevent its ubiquitination and degradation. By modeling such mutations in Piwi (Miwi) knockin mice, we demonstrate that the genetic defects are directly responsible for male infertility. Mechanistically, we show that MIWI binds the histone ubiquitin ligase RNF8 in a Piwi-interacting RNA (piRNA)-independent manner, and MIWI stabilization sequesters RNF8 in the cytoplasm of late spermatids. The resulting aberrant sperm show histone retention, abnormal morphology, and severely compromised activity, which can be functionally rescued via blocking RNF8-MIWI interaction in spermatids with an RNF8-N peptide. Collectively, our findings identify Piwi as a factor in human infertility and reveal its role in regulating the histone-to-protamine exchange during spermiogenesis.


Subject(s)
Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Azoospermia/genetics , Mutation , Animals , Azoospermia/metabolism , Chromatin/metabolism , DNA Mutational Analysis , DNA-Binding Proteins/metabolism , Disease Models, Animal , Female , Gene Knock-In Techniques , Histones/metabolism , Humans , Introns , Male , Mice , Pedigree , Protamines/metabolism , Proteolysis , Spermatogenesis , Ubiquitin-Protein Ligases , Ubiquitination
7.
Mol Cell ; 77(5): 1014-1031.e13, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32017898

ABSTRACT

The La-related protein 7 (LARP7) forms a complex with the nuclear 7SK RNA to regulate RNA polymerase II transcription. It has been implicated in cancer and the Alazami syndrome, a severe developmental disorder. Here, we report a so far unknown role of this protein in RNA modification. We show that LARP7 physically connects the spliceosomal U6 small nuclear RNA (snRNA) with a distinct subset of box C/D small nucleolar RNAs (snoRNAs) guiding U6 2'-O-methylation. Consistently, these modifications are severely compromised in the absence of LARP7. Although general splicing remains largely unaffected, transcriptome-wide analysis revealed perturbations in alternative splicing in LARP7-depleted cells. Importantly, we identified defects in 2'-O-methylation of the U6 snRNA in Alazami syndrome siblings carrying a LARP7 mutation. Our data identify LARP7 as a bridging factor for snoRNA-guided modification of the U6 snRNA and suggest that alterations in splicing fidelity contribute to the etiology of the Alazami syndrome.


Subject(s)
Alternative Splicing , Developmental Disabilities/metabolism , RNA, Small Nuclear/metabolism , Ribonucleoproteins/metabolism , Spliceosomes/metabolism , Binding Sites , Cell Line, Tumor , Child , Child, Preschool , Conserved Sequence , Developmental Disabilities/genetics , Female , Genetic Predisposition to Disease , HEK293 Cells , Humans , Male , Methylation , Middle Aged , Mutation , Nucleic Acid Conformation , Protein Binding , Protein Interaction Domains and Motifs , RNA, Small Nuclear/genetics , Ribonucleoproteins/genetics , Spliceosomes/genetics
8.
Mol Cell ; 77(5): 999-1013.e6, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32017896

ABSTRACT

U6 snRNA, as an essential component of the catalytic core of the pre-mRNA processing spliceosome, is heavily modified post-transcriptionally, with 2'-O-methylation being most common. The role of these modifications in pre-mRNA splicing as well as their physiological function in mammals have remained largely unclear. Here we report that the La-related protein LARP7 functions as a critical cofactor for 2'-O-methylation of U6 in mouse male germ cells. Mechanistically, LARP7 promotes U6 loading onto box C/D snoRNP, facilitating U6 2'-O-methylation by box C/D snoRNP. Importantly, ablation of LARP7 in the male germline causes defective U6 2'-O-methylation, massive alterations in pre-mRNA splicing, and spermatogenic failure in mice, which can be rescued by ectopic expression of wild-type LARP7 but not an U6-loading-deficient mutant LARP7. Our data uncover a novel role of LARP7 in regulating U6 2'-O-methylation and demonstrate the functional requirement of such modification for splicing fidelity and spermatogenesis in mice.


Subject(s)
RNA Precursors/metabolism , RNA Splicing , RNA, Messenger/metabolism , RNA, Small Nuclear/metabolism , RNA-Binding Proteins/metabolism , Spermatogenesis , Spermatozoa/metabolism , Spliceosomes/metabolism , Animals , Fertility , Gene Expression Regulation, Developmental , HEK293 Cells , Humans , Male , Methylation , Mice, Inbred C57BL , Mice, Knockout , RNA Precursors/genetics , RNA, Messenger/genetics , RNA, Small Nuclear/genetics , RNA-Binding Proteins/genetics , Ribonucleoproteins, Small Nucleolar/genetics , Ribonucleoproteins, Small Nucleolar/metabolism , Signal Transduction , Spermatogenesis/genetics , Spliceosomes/genetics
9.
J Cell Physiol ; 238(12): 2765-2777, 2023 12.
Article in English | MEDLINE | ID: mdl-37812578

ABSTRACT

Glucose metabolic reprogramming, known as the Warburg effect, is one of the metabolic hallmarks of tumor cells. Cancer cells preferentially metabolize glucose by glycolysis rather than mitochondrial oxidative phosphorylation regardless of oxygen availability, but the regulatory mechanism underlying this switch has been incompletely understood. Here, we report that the circular RNA circ ankyrin repeat domain 17 (ANKRD17) functions as a key regulator for glycolysis to promote cell growth, migration, invasion, and cell-cycle progression in breast cancer (BC) cells. We further show that circANKRD17 acts to accelerate glycolysis in BC cells by acting as a sponge for miR-143 and in turn overrides the repressive effect of miR-143, a well-documented glycolytic repressor, on hexokinase 2 in BC cells, thus resulting in enhanced glycolysis in BC cells. These data suggest the circANKRD17-miR-143 cascade as a novel mechanism in controlling glucose metabolic reprogramming in BC cells and suggest circANKRD17 as a promising therapeutic target to interrupt cancerous glycolysis.


Subject(s)
Breast Neoplasms , MicroRNAs , Humans , Female , MicroRNAs/genetics , MicroRNAs/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Glycolysis/genetics , Cell Proliferation/genetics , Glucose/metabolism , RNA-Binding Proteins/metabolism
10.
Development ; 146(13)2019 07 10.
Article in English | MEDLINE | ID: mdl-31189663

ABSTRACT

Epigenetic regulation, including histone-to-protamine exchanges, controls spermiogenesis. However, the underlying mechanisms of this regulation are largely unknown. Here, we report that PHF7, a testis-specific PHD and RING finger domain-containing protein, is essential for histone-to-protamine exchange in mice. PHF7 is specifically expressed during spermiogenesis. PHF7 deletion results in male infertility due to aberrant histone retention and impaired protamine replacement in elongated spermatids. Mechanistically, PHF7 can simultaneously bind histone H2A and H3; its PHD domain, a histone code reader, can specifically bind H3K4me3/me2, and its RING domain, a histone writer, can ubiquitylate H2A. Thus, our study reveals that PHF7 is a novel E3 ligase that can specifically ubiquitylate H2A through binding H3K4me3/me2 prior to histone-to-protamine exchange.


Subject(s)
Histones/metabolism , Protamines/metabolism , Spermatogenesis/genetics , Ubiquitin-Protein Ligases/physiology , Ubiquitination/genetics , Animals , Cells, Cultured , Chromatin Assembly and Disassembly/physiology , Female , Gene Expression Regulation, Developmental , Infertility, Male/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/genetics , Testis/metabolism , Ubiquitin-Protein Ligases/genetics
11.
Biol Reprod ; 107(1): 101-108, 2022 07 25.
Article in English | MEDLINE | ID: mdl-35403682

ABSTRACT

PIWI proteins and PIWI-interacting RNAs (piRNAs) are specifically expressed in animal germlines and play essential roles during gametogenesis in animals. The primary function of PIWI/piRNAs is known to silence transposable elements for protecting genome integrity in animal germlines, while their roles beyond silencing transposons are also documented by us and others. In particular, we show that mouse PIWIL1 (MIWI)/piRNAs play a dual role in regulating protein-coding genes in mouse spermatids through interacting with different protein factors in a developmental stage-dependent manner, including translationally activating a subset of AU-rich element-containing mRNAs in round spermatids and inducing massive mRNA degradation in late spermatids. We further show that MIWI is eliminated through the ubiquitin-26S proteasome pathway during late spermiogenesis. By exploring the biological function of MIWI ubiquitination by APC/C, we identified ubiquitination-deficient mutations in human PIWIL1 of infertile men and further established their causative role in male infertility in mouse model, supporting PIWIL1 as a human male infertility-relevant gene. Additionally, we reported that PIWIL1, aberrantly induced in human tumors, functions as an oncoprotein in a piRNA-independent manner in cancer cells. In the current review, we summarize our latest findings regarding the roles and mechanisms of PIWIL1 and piRNAs in mouse spermatids and human diseases, and discuss the related works in the field.


Subject(s)
Argonaute Proteins , Infertility, Male , Animals , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , Germ Cells/metabolism , Humans , Infertility, Male/genetics , Infertility, Male/metabolism , Male , Mice , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Spermatids/metabolism , Spermatogenesis/genetics
12.
Nucleic Acids Res ; 48(9): 4780-4796, 2020 05 21.
Article in English | MEDLINE | ID: mdl-32232334

ABSTRACT

Previously, we have shown that human sperm Prohibitin (PHB) expression is significantly negatively correlated with mitochondrial ROS levels but positively correlated with mitochondrial membrane potential and motility. However, the possible role of PHB in mammalian spermatogenesis has not been investigated. Here we document the presence of PHB in spermatocytes and its functional roles in meiosis by generating the first male germ cell-specific Phb-cKO mouse. Loss of PHB in spermatocytes resulted in complete male infertility, associated with not only meiotic pachytene arrest with accompanying apoptosis, but also apoptosis resulting from mitochondrial morphology and function impairment. Our mechanistic studies show that PHB in spermatocytes regulates the expression of STAG3, a key component of the meiotic cohesin complex, via a non-canonical JAK/STAT pathway, and consequently promotes meiotic DSB repair and homologous recombination. Furthermore, the PHB/JAK2 axis was found as a novel mechanism in the maintenance of stabilization of meiotic STAG3 cohesin complex and the modulation of heterochromatin formation in spermatocytes during meiosis. The observed JAK2-mediated epigenetic changes in histone modifications, reflected in a reduction of histone 3 tyrosine 41 phosphorylation (H3Y41ph) and a retention of H3K9me3 at the Stag3 locus, could be responsible for Stag3 dysregulation in spermatocytes with the loss of PHB.


Subject(s)
Histone Code , Meiosis/genetics , Repressor Proteins/physiology , Spermatocytes/metabolism , Spermatogenesis/genetics , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line , Chromosome Pairing , Epigenome , Histones/metabolism , Homologous Recombination , Infertility/genetics , Janus Kinase 2/metabolism , Janus Kinases/metabolism , Male , Mice , Mice, Knockout , Mitochondria/physiology , Mitochondria/ultrastructure , Pachytene Stage , Phosphorylation , Prohibitins , Repressor Proteins/genetics , Repressor Proteins/metabolism , STAT Transcription Factors/metabolism , Signal Transduction , Spermatocytes/enzymology , Spermatocytes/ultrastructure , Testis/metabolism
14.
EMBO J ; 34(21): 2671-85, 2015 Nov 03.
Article in English | MEDLINE | ID: mdl-26346275

ABSTRACT

Glucose metabolic reprogramming is a hallmark of cancer. Cancer cells rapidly adjust their energy source from oxidative phosphorylation to glycolytic metabolism in order to efficiently proliferate in a hypoxic environment, but the mechanism underlying this switch is still incompletely understood. Here, we report that hypoxia potently induces the RNA-binding protein HuR to specifically bind primary miR-199a transcript to block miR-199a maturation in hepatocellular carcinoma (HCC) cells. We demonstrate that this hypoxia-suppressed miR-199a plays a decisive role in limiting glycolysis in HCC cells by targeting hexokinase-2 (Hk2) and pyruvate kinase-M2 (Pkm2). Furthermore, systemically delivered cholesterol-modified agomiR-199a inhibits [(18)F]-fluorodeoxyglucose uptake and attenuates tumor growth in HCC tumor-bearing mice. These data reveal a novel mechanism of reprogramming of cancer energy metabolism in which HuR suppresses miR-199a maturation to link hypoxia to the Warburg effect and suggest a promising therapeutic strategy that targets miR-199a to interrupt cancerous aerobic glycolysis.


Subject(s)
Carcinoma, Hepatocellular/genetics , ELAV-Like Protein 1/physiology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , MicroRNAs/genetics , Animals , Base Sequence , Carcinoma, Hepatocellular/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cell Hypoxia , Cell Line, Tumor , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Humans , Liver Neoplasms/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/metabolism , Neoplasm Transplantation , Protein Binding , Thyroid Hormones/genetics , Thyroid Hormones/metabolism , Thyroid Hormone-Binding Proteins
16.
Cell Mol Life Sci ; 74(16): 2929-2941, 2017 08.
Article in English | MEDLINE | ID: mdl-28321489

ABSTRACT

The reprogramming of glucose metabolism from oxidative to glycolytic metabolism, known as the Warburg effect, is an anomalous characteristic of cancer cell metabolism. Recent studies have revealed a subset of microRNAs (miRNAs) that play critical roles in regulating the reprogramming of glucose metabolism in cancer cells. These miRNAs regulate cellular glucose metabolism by directly targeting multiple metabolic genes, including those encoding key glycolytic enzymes. In the first part of this review, we summarized the recent knowledge of miRNA regulation in the reprogramming of glucose metabolism in cancer cells and discussed the potential utilization of the key miRNA regulators as metabolic targets for developing new antitumor agents. Then, we summarized recent advances in methods and techniques for studying miRNA regulation in cancer cell metabolism.


Subject(s)
Gene Expression Regulation, Neoplastic , Glucose/metabolism , MicroRNAs/metabolism , Neoplasms/metabolism , Animals , Glucose/genetics , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Glycolysis , Hexokinase/genetics , Hexokinase/metabolism , Humans , L-Lactate Dehydrogenase/genetics , L-Lactate Dehydrogenase/metabolism , Magnetic Resonance Spectroscopy/methods , Mass Spectrometry/methods , MicroRNAs/genetics , Molecular Probe Techniques , Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase , Pyruvate Kinase/genetics , Pyruvate Kinase/metabolism
17.
Reproduction ; 154(6): 723-733, 2017 12.
Article in English | MEDLINE | ID: mdl-28871057

ABSTRACT

The chromatoid body (CB) is a specific cloud-like structure in the cytoplasm of haploid spermatids. Recent findings indicate that CB is identified as a male germ cell-specific RNA storage and processing center, but its function has remained elusive for decades. In somatic cells, KH-type splicing regulatory protein (KSRP) is involved in regulating gene expression and maturation of select microRNAs (miRNAs). However, the function of KSRP in spermatogenesis remains unclear. In this study, we showed that KSRP partly localizes in CB, as a component of CB. KSRP interacts with proteins (mouse VASA homolog (MVH), polyadenylate-binding protein 1 (PABP1) and polyadenylate-binding protein 2 (PABP2)), mRNAs (Tnp2 and Odf1) and microRNAs (microRNA-182) in mouse CB. Moreover, KSRP may regulate the integrity of CB via DDX5-miRNA-182 pathway. In addition, we found abnormal expressions of CB component in testes of Ksrp-knockout mice and of patients with hypospermatogenesis. Thus, our results provide mechanistic insight into the role of KSRP in spermatogenesis.


Subject(s)
RNA-Binding Proteins/metabolism , Spermatids/metabolism , Spermatogenesis , Trans-Activators/metabolism , Adult , Animals , Argonaute Proteins/deficiency , Argonaute Proteins/genetics , Case-Control Studies , Cells, Cultured , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , DNA-Binding Proteins , Gene Expression Regulation, Developmental , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Male , Mice, Knockout , MicroRNAs/genetics , MicroRNAs/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Oligospermia/genetics , Oligospermia/metabolism , Poly(A)-Binding Protein I/genetics , Poly(A)-Binding Protein I/metabolism , Poly(A)-Binding Protein II/genetics , Poly(A)-Binding Protein II/metabolism , Protein Binding , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , Signal Transduction , Trans-Activators/deficiency , Trans-Activators/genetics , Young Adult
18.
EMBO J ; 31(8): 1985-98, 2012 Apr 18.
Article in English | MEDLINE | ID: mdl-22354042

ABSTRACT

Cancer cells preferentially metabolize glucose through aerobic glycolysis. This phenomenon, known as the Warburg effect, is an anomalous characteristic of glucose metabolism in cancer cells. Chronic inflammation is a key promoting factor of tumourigenesis. It remains, however, largely unexplored whether and how pro-tumourigenic inflammation regulates glucose metabolism in cancer cells. Here, we show that pro-inflammatory cytokines promote glycolysis in breast cancer cells, and that the inflammation-induced miR-155 functions as an important mediator in this process. We further show that miR-155 acts to upregulate hexokinase 2 (hk2), through two distinct mechanisms. First, miR-155 promotes hk2 transcription by activation of signal transducer and activator of transcription 3 (STAT3), a transcriptional activator for hk2. Second, via targeting C/EBPß (a transcriptional activator for mir-143), miR-155 represses mir-143, a negative regulator of hk2, thus resulting in upregulation of hk2 expression at the post-transcriptional level. The miR-155-mediated hk2 upregulation also appears to operate in other types of cancer cells examined. We suggest that the miR-155/miR-143/HK2 axis may represent a common mechanism linking inflammation to the altered metabolism in cancer cells.


Subject(s)
Glycolysis , Hexokinase/biosynthesis , MicroRNAs/metabolism , Animals , Breast Neoplasms/physiopathology , Cell Line, Tumor , Humans , Mice
19.
Proc Natl Acad Sci U S A ; 108(25): 10144-9, 2011 Jun 21.
Article in English | MEDLINE | ID: mdl-21646541

ABSTRACT

MicroRNA 21 (miR-21) is overexpressed in virtually all types of carcinomas and various types of hematological malignancies. To determine whether miR-21 promotes tumor development in vivo, we knocked out the miR-21 allele in mice. In response to the 7,12-dimethylbenz[a]anthracene (DMBA)/12-O-tetradecanoylphorbol-13-acetate mouse skin carcinogenesis protocol, miR-21-null mice showed a significant reduction in papilloma formation compared with wild-type mice. We revealed that cellular apoptosis was elevated and cell proliferation was decreased in mice deficient of miR-21 compared to wild-type animals. In addition, we found that a large number of validated or predicted miR-21 target genes were up-regulated in miR-21-null keratinocytes, which are precursor cells to skin papillomas. Specifically, up-regulation of Spry1, Pten, and Pdcd4 when miR-21 was ablated coincided with reduced phosphorylation of ERK, AKT, and JNK, three major downstream effectors of Ras activation that plays a predominant role in DMBA-initiated skin carcinogenesis. These results provide in vivo evidence that miR-21 exerts its oncogenic function through negatively regulating its target genes.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/metabolism , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Apoptosis/genetics , Carcinogens/pharmacology , Cell Proliferation , Epidermal Cells , Epidermis/drug effects , Epidermis/pathology , Epidermis/physiology , Female , Keratinocytes/cytology , Keratinocytes/drug effects , Keratinocytes/physiology , Mice , Mice, Knockout , MicroRNAs/genetics , Signal Transduction/physiology , Skin Neoplasms/chemically induced , Skin Neoplasms/pathology , Skin Neoplasms/physiopathology , Tetradecanoylphorbol Acetate/pharmacology , ras Proteins/genetics , ras Proteins/metabolism
20.
J Genet Genomics ; 51(2): 111-132, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38181897

ABSTRACT

Previous studies on genetic diseases predominantly focused on protein-coding variations, overlooking the vast noncoding regions in the human genome. The development of high-throughput sequencing technologies and functional genomics tools has enabled the systematic identification of functional noncoding variants. These variants can impact gene expression, regulation, and chromatin conformation, thereby contributing to disease pathogenesis. Understanding the mechanisms that underlie the impact of noncoding variants on genetic diseases is indispensable for the development of precisely targeted therapies and the implementation of personalized medicine strategies. The intricacies of noncoding regions introduce a multitude of challenges and research opportunities. In this review, we introduce a spectrum of noncoding variants involved in genetic diseases, along with research strategies and advanced technologies for their precise identification and in-depth understanding of the complexity of the noncoding genome. We will delve into the research challenges and propose potential solutions for unraveling the genetic basis of rare and complex diseases.


Subject(s)
Genetic Variation , Genomics , Humans , Genetic Variation/genetics , Precision Medicine , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study
SELECTION OF CITATIONS
SEARCH DETAIL