Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 91
Filter
1.
Nat Rev Mol Cell Biol ; 24(6): 430-447, 2023 06.
Article in English | MEDLINE | ID: mdl-36596869

ABSTRACT

Genes specifying long non-coding RNAs (lncRNAs) occupy a large fraction of the genomes of complex organisms. The term 'lncRNAs' encompasses RNA polymerase I (Pol I), Pol II and Pol III transcribed RNAs, and RNAs from processed introns. The various functions of lncRNAs and their many isoforms and interleaved relationships with other genes make lncRNA classification and annotation difficult. Most lncRNAs evolve more rapidly than protein-coding sequences, are cell type specific and regulate many aspects of cell differentiation and development and other physiological processes. Many lncRNAs associate with chromatin-modifying complexes, are transcribed from enhancers and nucleate phase separation of nuclear condensates and domains, indicating an intimate link between lncRNA expression and the spatial control of gene expression during development. lncRNAs also have important roles in the cytoplasm and beyond, including in the regulation of translation, metabolism and signalling. lncRNAs often have a modular structure and are rich in repeats, which are increasingly being shown to be relevant to their function. In this Consensus Statement, we address the definition and nomenclature of lncRNAs and their conservation, expression, phenotypic visibility, structure and functions. We also discuss research challenges and provide recommendations to advance the understanding of the roles of lncRNAs in development, cell biology and disease.


Subject(s)
RNA, Long Noncoding , RNA, Long Noncoding/genetics , Cell Nucleus/genetics , Chromatin/genetics , Regulatory Sequences, Nucleic Acid , RNA Polymerase II/genetics
2.
Cell ; 172(3): 393-407, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29373828

ABSTRACT

Over the last decade, it has been increasingly demonstrated that the genomes of many species are pervasively transcribed, resulting in the production of numerous long noncoding RNAs (lncRNAs). At the same time, it is now appreciated that many types of DNA regulatory elements, such as enhancers and promoters, regularly initiate bi-directional transcription. Thus, discerning functional noncoding transcripts from a vast transcriptome is a paramount priority, and challenge, for the lncRNA field. In this review, we aim to provide a conceptual and experimental framework for classifying and elucidating lncRNA function. We categorize lncRNA loci into those that regulate gene expression in cis versus those that perform functions in trans and propose an experimental approach to dissect lncRNA activity based on these classifications. These strategies to further understand lncRNAs promise to reveal new and unanticipated biology with great potential to advance our understanding of normal physiology and disease.


Subject(s)
RNA, Long Noncoding/genetics , Animals , Humans , RNA, Long Noncoding/classification , RNA, Long Noncoding/metabolism , Regulatory Sequences, Nucleic Acid , Transcription, Genetic
3.
Mol Cell ; 84(4): 776-790.e5, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38211588

ABSTRACT

TANK-binding kinase 1 (TBK1) is a potential therapeutic target in multiple cancers, including clear cell renal cell carcinoma (ccRCC). However, targeting TBK1 in clinical practice is challenging. One approach to overcome this challenge would be to identify an upstream TBK1 regulator that could be targeted therapeutically in cancer specifically. In this study, we perform a kinome-wide small interfering RNA (siRNA) screen and identify doublecortin-like kinase 2 (DCLK2) as a TBK1 regulator in ccRCC. DCLK2 binds to and directly phosphorylates TBK1 on Ser172. Depletion of DCLK2 inhibits anchorage-independent colony growth and kidney tumorigenesis in orthotopic xenograft models. Conversely, overexpression of DCLK2203, a short isoform that predominates in ccRCC, promotes ccRCC cell growth and tumorigenesis in vivo. Mechanistically, DCLK2203 elicits its oncogenic signaling via TBK1 phosphorylation and activation. Taken together, these results suggest that DCLK2 is a TBK1 activator and potential therapeutic target for ccRCC.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Carcinogenesis/genetics , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Doublecortin-Like Kinases , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
4.
Cell ; 164(1-2): 69-80, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26724866

ABSTRACT

Long noncoding RNAs (lncRNAs) have emerged as regulators of diverse biological processes. Here, we describe the initial functional analysis of a poorly characterized human lncRNA (LINC00657) that is induced after DNA damage, which we termed "noncoding RNA activated by DNA damage", or NORAD. NORAD is highly conserved and abundant, with expression levels of approximately 500-1,000 copies per cell. Remarkably, inactivation of NORAD triggers dramatic aneuploidy in previously karyotypically stable cell lines. NORAD maintains genomic stability by sequestering PUMILIO proteins, which repress the stability and translation of mRNAs to which they bind. In the absence of NORAD, PUMILIO proteins drive chromosomal instability by hyperactively repressing mitotic, DNA repair, and DNA replication factors. These findings introduce a mechanism that regulates the activity of a deeply conserved and highly dosage-sensitive family of RNA binding proteins and reveal unanticipated roles for a lncRNA and PUMILIO proteins in the maintenance of genomic stability.


Subject(s)
Genomic Instability , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/metabolism , Animals , Base Sequence , Chromosomal Instability , HCT116 Cells , Humans , Mice , Ploidies , RNA, Long Noncoding/chemistry , RNA, Long Noncoding/genetics
5.
Genes Dev ; 37(5-6): 191-203, 2023 03 01.
Article in English | MEDLINE | ID: mdl-36859340

ABSTRACT

Subcellular localization of messenger RNA (mRNA) is a widespread phenomenon that can impact the regulation and function of the encoded protein. In nonneuronal cells, specific mRNAs localize to cell protrusions, and proper mRNA localization is required for cell migration. However, the mechanisms by which mRNA localization regulates protein function in this setting remain unclear. Here, we examined the functional consequences of localization of the mRNA encoding KIF1C. KIF1C is a kinesin motor protein required for cell migration and mRNA trafficking, including trafficking of its own mRNA. We show that Kif1c mRNA localization does not regulate KIF1C's protein abundance, distribution, or ability to traffic other mRNAs. Conversely, Kif1c mRNA localization to protrusions is required for directed cell migration. We used mass spectrometry to identify binding partners of endogenous KIF1C, which revealed dramatic dysregulation of the number and identity of KIF1C interactors in response to Kif1c mRNA mislocalization. These results therefore uncovered a mechanistic connection between mRNA localization to cell protrusions and the specificity of protein-protein interactions. We anticipate that this mechanism is not limited to Kif1c and is likely to be a general principle that impacts the functions of proteins encoded by protrusion-enriched mRNAs in nonneuronal cells.


Subject(s)
Kinesins , Proteins , RNA, Messenger/metabolism , Proteins/metabolism , Kinesins/genetics , Kinesins/metabolism , Dyneins/metabolism , Cell Movement/genetics
6.
Genes Dev ; 37(13-14): 661-674, 2023 07 01.
Article in English | MEDLINE | ID: mdl-37553261

ABSTRACT

MicroRNAs (miRNAs) are post-transcriptional regulators of gene expression that play critical roles in development and disease. Target-directed miRNA degradation (TDMD), a pathway in which miRNAs that bind to specialized targets with extensive complementarity are rapidly decayed, has emerged as a potent mechanism of controlling miRNA levels. Nevertheless, the biological role and scope of miRNA regulation by TDMD in mammals remains poorly understood. To address these questions, we generated mice with constitutive or conditional deletion of Zswim8, which encodes an essential TDMD factor. Loss of Zswim8 resulted in developmental defects in the heart and lungs, growth restriction, and perinatal lethality. Small RNA sequencing of embryonic tissues revealed widespread miRNA regulation by TDMD and greatly expanded the known catalog of miRNAs regulated by this pathway. These experiments also uncovered novel features of TDMD-regulated miRNAs, including their enrichment in cotranscribed clusters and examples in which TDMD underlies "arm switching," a phenomenon wherein the dominant strand of a miRNA precursor changes in different tissues or conditions. Importantly, deletion of two miRNAs, miR-322 and miR-503, rescued growth of Zswim8-null embryos, directly implicating the TDMD pathway as a regulator of mammalian body size. These data illuminate the broad landscape and developmental role of TDMD in mammals.


Subject(s)
MicroRNAs , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Mammals/genetics , Base Sequence
7.
Genes Dev ; 36(9-10): 550-565, 2022 05 01.
Article in English | MEDLINE | ID: mdl-35589130

ABSTRACT

Although splicing is a major driver of RNA nuclear export, many intronless RNAs are efficiently exported to the cytoplasm through poorly characterized mechanisms. For example, GC-rich sequences promote nuclear export in a splicing-independent manner, but how GC content is recognized and coupled to nuclear export is unknown. Here, we developed a genome-wide screening strategy to investigate the mechanism of export of NORAD, an intronless cytoplasmic long noncoding RNA (lncRNA). This screen revealed an RNA binding protein, RBM33, that directs the nuclear export of NORAD and numerous other transcripts. RBM33 directly binds substrate transcripts and recruits components of the TREX-NXF1/NXT1 RNA export pathway. Interestingly, high GC content emerged as the feature that specifies RBM33-dependent nuclear export. Accordingly, RBM33 directly binds GC-rich elements in target transcripts. These results provide a broadly applicable strategy for the genetic dissection of nuclear export mechanisms and reveal a long-sought nuclear export pathway for transcripts with GC-rich sequences.


Subject(s)
Nucleocytoplasmic Transport Proteins , RNA, Viral , Active Transport, Cell Nucleus , Cell Nucleus/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , RNA Transport , RNA, Viral/metabolism
8.
Cell ; 157(5): 1104-16, 2014 May 22.
Article in English | MEDLINE | ID: mdl-24855947

ABSTRACT

Downregulation of the miR-143/145 microRNA (miRNA) cluster has been repeatedly reported in colon cancer and other epithelial tumors. In addition, overexpression of these miRNAs inhibits tumorigenesis, leading to broad consensus that they function as cell-autonomous epithelial tumor suppressors. We generated mice with deletion of miR-143/145 to investigate the functions of these miRNAs in intestinal physiology and disease in vivo. Although intestinal development proceeded normally in the absence of these miRNAs, epithelial regeneration after injury was dramatically impaired. Surprisingly, we found that miR-143/145 are expressed and function exclusively within the mesenchymal compartment of intestine. Defective epithelial regeneration in miR-143/145-deficient mice resulted from the dysfunction of smooth muscle and myofibroblasts and was associated with derepression of the miR-143 target Igfbp5, which impaired IGF signaling after epithelial injury. These results provide important insights into the regulation of epithelial wound healing and argue against a cell-autonomous tumor suppressor role for miR-143/145 in colon cancer.


Subject(s)
Intestinal Mucosa/physiology , MicroRNAs/metabolism , Animals , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Dextran Sulfate , Humans , Insulin-Like Growth Factor Binding Protein 5/genetics , Intestinal Mucosa/cytology , Mesoderm/metabolism , Mice , MicroRNAs/genetics , Myofibroblasts/metabolism , Paracrine Communication , Regeneration , Somatomedins/metabolism
9.
Trends Biochem Sci ; 49(2): 101-104, 2024 02.
Article in English | MEDLINE | ID: mdl-37949765

ABSTRACT

Intrinsically disordered regions (IDRs) within human proteins play critical roles in cellular information processing, including signaling, transcription, stress response, DNA repair, genome organization, and RNA processing. Here, we summarize current challenges in the field and propose cutting-edge approaches to address them in physiology and disease processes, with a focus on cancer.


Subject(s)
Intrinsically Disordered Proteins , Humans , Intrinsically Disordered Proteins/metabolism , Biophysics , Biology
10.
Mol Cell ; 77(5): 1044-1054.e3, 2020 03 05.
Article in English | MEDLINE | ID: mdl-31924448

ABSTRACT

Antisense oligonucleotides (ASOs) that trigger RNase-H-mediated cleavage are commonly used to knock down transcripts for experimental or therapeutic purposes. In particular, ASOs are frequently used to functionally interrogate long noncoding RNAs (lncRNAs) and discriminate lncRNA loci that produce functional RNAs from those whose activity is attributable to the act of transcription. Transcription termination is triggered by cleavage of nascent transcripts, generally during polyadenylation, resulting in degradation of the residual RNA polymerase II (Pol II)-associated RNA by XRN2 and dissociation of elongating Pol II. Here, we show that ASOs act upon nascent transcripts and, consequently, induce premature transcription termination downstream of the cleavage site in an XRN2-dependent manner. Targeting the transcript 3' end with ASOs, however, allows transcript knockdown while preserving Pol II association with the gene body. These results demonstrate that the effects of ASOs on transcription must be considered for appropriate experimental and therapeutic use of these reagents.


Subject(s)
Chromatin/metabolism , Oligonucleotides, Antisense/metabolism , RNA Precursors/metabolism , RNA Stability , RNA, Messenger/metabolism , Transcription Termination, Genetic , Chromatin/genetics , Exoribonucleases/metabolism , HCT116 Cells , HEK293 Cells , Humans , Models, Genetic , Oligonucleotides, Antisense/genetics , RNA Polymerase II/genetics , RNA Polymerase II/metabolism , RNA Precursors/genetics , RNA, Messenger/genetics , Time Factors
11.
Trends Biochem Sci ; 48(1): 26-39, 2023 01.
Article in English | MEDLINE | ID: mdl-35811249

ABSTRACT

MicroRNAs (miRNAs) post-transcriptionally repress gene expression by guiding Argonaute (AGO) proteins to target mRNAs. While much is known about the regulation of miRNA biogenesis, miRNA degradation pathways are comparatively poorly understood. Although miRNAs generally exhibit slow turnover, they can be rapidly degraded through regulated mechanisms that act in a context- or sequence-specific manner. Recent work has revealed a particularly important role for specialized target interactions in controlling rates of miRNA degradation. Engagement of these targets is associated with the addition and removal of nucleotides from the 3' ends of miRNAs, a process known as tailing and trimming. Here we review these mechanisms of miRNA modification and turnover, highlighting the contexts in which they impact miRNA stability and discussing important questions that remain unanswered.


Subject(s)
MicroRNAs , MicroRNAs/genetics , Argonaute Proteins/genetics , Argonaute Proteins/metabolism , RNA Stability , Nucleotides/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Cell ; 148(6): 1172-87, 2012 Mar 16.
Article in English | MEDLINE | ID: mdl-22424228

ABSTRACT

Disease is often the result of an aberrant or inadequate response to physiologic and pathophysiologic stress. Studies over the last 10 years have uncovered a recurring paradigm in which microRNAs (miRNAs) regulate cellular behavior under these conditions, suggesting an especially significant role for these small RNAs in pathologic settings. Here, we review emerging principles of miRNA regulation of stress signaling pathways and apply these concepts to our understanding of the roles of miRNAs in disease. These discussions further highlight the unique challenges and opportunities associated with the mechanistic dissection of miRNA functions and the development of miRNA-based therapeutics.


Subject(s)
Disease/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Signal Transduction , Animals , Humans
13.
Nature ; 595(7866): 303-308, 2021 07.
Article in English | MEDLINE | ID: mdl-34108682

ABSTRACT

Liquid-liquid phase separation is a major mechanism of subcellular compartmentalization1,2. Although the segregation of RNA into phase-separated condensates broadly affects RNA metabolism3,4, whether and how specific RNAs use phase separation to regulate interacting factors such as RNA-binding proteins (RBPs), and the phenotypic consequences of such regulatory interactions, are poorly understood. Here we show that RNA-driven phase separation is a key mechanism through which a long noncoding RNA (lncRNA) controls the activity of RBPs and maintains genomic stability in mammalian cells. The lncRNA NORAD prevents aberrant mitosis by inhibiting Pumilio (PUM) proteins5-8. We show that NORAD can out-compete thousands of other PUM-binding transcripts to inhibit PUM by nucleating the formation of phase-separated PUM condensates, termed NP bodies. Dual mechanisms of PUM recruitment, involving multivalent PUM-NORAD and PUM-PUM interactions, enable NORAD to competitively sequester a super-stoichiometric amount of PUM in NP bodies. Disruption of NORAD-driven PUM phase separation leads to PUM hyperactivity and genome instability that is rescued by synthetic RNAs that induce the formation of PUM condensates. These results reveal a mechanism by which RNA-driven phase separation can regulate RBP activity and identify an essential role for this process in genome maintenance. The repetitive sequence architecture of NORAD and other lncRNAs9-11 suggests that phase separation may be a widely used mechanism of lncRNA-mediated regulation.


Subject(s)
Genomic Instability , Phase Transition , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA-Binding Proteins/chemistry , RNA-Binding Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Cell Line , Cytoplasm/chemistry , Cytoplasm/genetics , Cytoplasm/metabolism , Humans , RNA/chemistry , RNA/genetics , RNA/metabolism , RNA, Long Noncoding/chemistry
14.
Genes Dev ; 33(19-20): 1367-1380, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31488578

ABSTRACT

Fat storage in adult mammals is a highly regulated process that involves the mobilization of adipocyte progenitor cells (APCs) that differentiate to produce new adipocytes. Here we report a role for the broadly conserved miR-26 family of microRNAs (miR-26a-1, miR-26a-2, and miR-26b) as major regulators of APC differentiation and adipose tissue mass. Deletion of all miR-26-encoding loci in mice resulted in a dramatic expansion of adipose tissue in adult animals fed normal chow. Conversely, transgenic overexpression of miR-26a protected mice from high-fat diet-induced obesity. These effects were attributable to a cell-autonomous function of miR-26 as a potent inhibitor of APC differentiation. miR-26 blocks adipogenesis, at least in part, by repressing expression of Fbxl19, a conserved miR-26 target without a previously known role in adipocyte biology that encodes a component of SCF-type E3 ubiquitin ligase complexes. These findings have therefore revealed a novel pathway that plays a critical role in regulating adipose tissue formation in vivo and suggest new potential therapeutic targets for obesity and related disorders.


Subject(s)
Adipogenesis/genetics , Cell Differentiation/genetics , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , F-Box Proteins/genetics , F-Box Proteins/metabolism , MicroRNAs/metabolism , Obesity/genetics , Stem Cells/cytology , Animals , Diet, High-Fat , Gene Expression , Gene Knockdown Techniques , Mice , MicroRNAs/genetics
15.
Nature ; 582(7810): 134, 2020 06.
Article in English | MEDLINE | ID: mdl-32483375

ABSTRACT

A Retraction to this paper has been published and can be accessed via a link at the top of the paper.

16.
Genes Dev ; 32(13-14): 903-908, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29950491

ABSTRACT

Loss of function of the DIS3L2 exoribonuclease is associated with Wilms tumor and the Perlman congenital overgrowth syndrome. LIN28, a Wilms tumor oncoprotein, triggers the DIS3L2-mediated degradation of the precursor of let-7, a microRNA that inhibits Wilms tumor development. These observations have led to speculation that DIS3L2-mediated tumor suppression is attributable to let-7 regulation. Here we examine new DIS3L2-deficient cell lines and mouse models, demonstrating that DIS3L2 loss has no effect on mature let-7 levels. Rather, analysis of Dis3l2-null nephron progenitor cells, a potential cell of origin of Wilms tumors, reveals up-regulation of Igf2, a growth-promoting gene strongly associated with Wilms tumorigenesis. These findings nominate a new potential mechanism underlying the pathology associated with DIS3L2 deficiency.


Subject(s)
Exoribonucleases/genetics , Fetal Macrosomia/genetics , Insulin-Like Growth Factor II/genetics , Up-Regulation , Wilms Tumor/genetics , Animals , Cell Line , Disease Models, Animal , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , Mutation , Nephrons/cytology , Nephrons/physiopathology , Stem Cells
17.
Genes Dev ; 32(15-16): 996-1007, 2018 08 01.
Article in English | MEDLINE | ID: mdl-30026293

ABSTRACT

Many childhood Wilms tumors are driven by mutations in the microRNA biogenesis machinery, but the mechanism by which these mutations drive tumorigenesis is unknown. Here we show that the transcription factor pleomorphic adenoma gene 1 (PLAG1) is a microRNA target gene that is overexpressed in Wilms tumors with mutations in microRNA processing genes. Wilms tumors can also overexpress PLAG1 through copy number alterations, and PLAG1 expression correlates with prognosis in Wilms tumors. PLAG1 overexpression accelerates growth of Wilms tumor cells in vitro and induces neoplastic growth in the developing mouse kidney in vivo. In both settings, PLAG1 transactivates insulin-like growth factor 2 (IGF2), a key Wilms tumor oncogene, and drives mammalian target of rapamycin complex 1 (mTORC1) signaling. These data link microRNA impairment to the PLAG1-IGF2 pathway, providing new insight into the manner in which common Wilms tumor mutations drive disease pathogenesis.


Subject(s)
DNA-Binding Proteins/genetics , Insulin-Like Growth Factor II/biosynthesis , MicroRNAs/metabolism , Mutation , Transcription Factors/genetics , Wilms Tumor/genetics , Animals , Cell Line, Tumor , DNA Copy Number Variations , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Humans , Kidney/metabolism , Mice , RNA Processing, Post-Transcriptional , TOR Serine-Threonine Kinases/metabolism , Transcription Factors/metabolism , Wilms Tumor/metabolism , Wilms Tumor/pathology
18.
Nature ; 570(7761): E51, 2019 Jun.
Article in English | MEDLINE | ID: mdl-31127195

ABSTRACT

Change history: In this Letter, the citation to 'Fig. 4e, f' in the main text should be 'Fig. 3e, f'. This has not been corrected online.

19.
Cell ; 137(4): 606-8, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19450510

ABSTRACT

MicroRNAs have been implicated as regulators of embryonic stem (ES) cell self-renewal and pluripotency. In this issue, Xu et al. (2009) demonstrate that miR-145 facilitates ES cell differentiation by repressing the core pluripotency factors OCT4, SOX2, and KLF4, thereby silencing the self-renewal program.


Subject(s)
Cell Differentiation , Embryonic Stem Cells/cytology , MicroRNAs/metabolism , Animals , Gene Silencing , Humans , Kruppel-Like Factor 4
20.
Cell ; 137(6): 1005-17, 2009 Jun 12.
Article in English | MEDLINE | ID: mdl-19524505

ABSTRACT

Therapeutic strategies based on modulation of microRNA (miRNA) activity hold great promise due to the ability of these small RNAs to potently influence cellular behavior. In this study, we investigated the efficacy of a miRNA replacement therapy for liver cancer. We demonstrate that hepatocellular carcinoma (HCC) cells exhibit reduced expression of miR-26a, a miRNA that is normally expressed at high levels in diverse tissues. Expression of this miRNA in liver cancer cells in vitro induces cell-cycle arrest associated with direct targeting of cyclins D2 and E2. Systemic administration of this miRNA in a mouse model of HCC using adeno-associated virus (AAV) results in inhibition of cancer cell proliferation, induction of tumor-specific apoptosis, and dramatic protection from disease progression without toxicity. These findings suggest that delivery of miRNAs that are highly expressed and therefore tolerated in normal tissues but lost in disease cells may provide a general strategy for miRNA replacement therapies.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/therapy , Liver Neoplasms/genetics , Liver Neoplasms/therapy , MicroRNAs/therapeutic use , Animals , Cyclin D2 , Cyclins/metabolism , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors , Mice , MicroRNAs/genetics , MicroRNAs/metabolism , Proto-Oncogene Proteins c-myc/genetics
SELECTION OF CITATIONS
SEARCH DETAIL