1
|
Shin AE, Sugiura K, Kariuki SW, Cohen DA, Flashner SP, Klein-Szanto AJ, Nishiwaki N, De D, Vasan N, Gabre JT, Lengner CJ, Sims PA, Rustgi AK. LIN28B-mediated PI3K/AKT pathway activation promotes metastasis in colorectal cancer models. J Clin Invest 2025; 135:e186035. [PMID: 39808497 PMCID: PMC11996871 DOI: 10.1172/jci186035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Colorectal cancer (CRC) remains a leading cause of cancer death because of metastatic spread. LIN28B is overexpressed in 30% of CRCs and promotes metastasis, yet its mechanisms remain unclear. In this study, we genetically modified CRC cell lines to overexpress LIN28B, resulting in enhanced PI3K/AKT pathway activation and liver metastasis in mice. We developed genetically modified mouse models with constitutively active Pik3ca that form intestinal tumors progressing to liver metastases with an intact immune system, addressing the limitations of previous Pik3ca-mutant models, including long tumor latency, mixed histology, and lack of distant metastases. The PI3Kα-specific inhibitor alpelisib reduced migration and invasion in vitro and metastasis in vivo. We present a comprehensive analysis of vertical inhibition of the PI3K/AKT pathway in CRC using the FDA-approved drugs alpelisib and capivasertib (an AKT inhibitor) in combination with LY2584702 (a ribosomal protein S6 kinase inhibitor) in CRC cell lines and mouse- and patient-derived organoids. Tissue microarrays from patients with CRC verified that LIN28B and PI3K/AKT pathway activation correlate with CRC progression. These findings highlight the critical role of the LIN28B-mediated PI3K/AKT pathway in CRC metastasis, the therapeutic potential of targeted inhibition, and the promise of patient-derived organoids in precision medicine in metastatic CRC.
Collapse
Affiliation(s)
- Alice E. Shin
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Kensuke Sugiura
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | | | - David A. Cohen
- Department of Surgery, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons; Columbia University Irving Medical Center, New York, New York, USA
| | | | | | | | - Dechokyab De
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Neil Vasan
- Division of Hematology and Oncology, Department of Medicine, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Joel T. Gabre
- Division of Digestive and Liver Diseases, Department of Medicine, and
| | - Christopher J. Lengner
- Department of Biomedical Sciences, School of Veterinary Medicine, and Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter A. Sims
- Department of Systems Biology, Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, New York, New York, USA
| | - Anil K. Rustgi
- Division of Digestive and Liver Diseases, Department of Medicine, and
| |
Collapse
|
2
|
Reimão-Pinto MM, Castillo-Hair SM, Seelig G, Schier AF. The regulatory landscape of 5' UTRs in translational control during zebrafish embryogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.23.568470. [PMID: 38045294 PMCID: PMC10690280 DOI: 10.1101/2023.11.23.568470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
The 5' UTRs of mRNAs are critical for translation regulation, but their in vivo regulatory features are poorly characterized. Here, we report the regulatory landscape of 5' UTRs during early zebrafish embryogenesis using a massively parallel reporter assay of 18,154 sequences coupled to polysome profiling. We found that the 5' UTR is sufficient to confer temporal dynamics to translation initiation, and identified 86 motifs enriched in 5' UTRs with distinct ribosome recruitment capabilities. A quantitative deep learning model, DaniO5P, revealed a combined role for 5' UTR length, translation initiation site context, upstream AUGs and sequence motifs on in vivo ribosome recruitment. DaniO5P predicts the activities of 5' UTR isoforms and indicates that modulating 5' UTR length and motif grammar contributes to translation initiation dynamics. This study provides a first quantitative model of 5' UTR-based translation regulation in early vertebrate development and lays the foundation for identifying the underlying molecular effectors.
Collapse
Affiliation(s)
| | - Sebastian M Castillo-Hair
- Department of Electrical & Computer Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Georg Seelig
- Department of Electrical & Computer Engineering, University of Washington, Seattle, Washington 98195, United States
- Paul G. Allen School of Computer Science & Engineering, University of Washington, Seattle, Washington 98195, United States
| | - Alex F Schier
- Biozentrum, University of Basel, 4056 Basel, Switzerland
- Allen Discovery Center for Cell Lineage Tracing, Seattle, Washington 98195, United States
| |
Collapse
|
3
|
Yang K, Cui S, Wang J, Xu T, Du H, Yue H, Ye H, Guo J, Zhang J, Li P, Guo Y, Pan C, Pang J, Wang J, Yu X, Zhang C, Liu Z, Chen Y, Xu F. Early Progression of Abdominal Aortic Aneurysm is Decelerated by Improved Endothelial Barrier Function via ALDH2-LIN28B-ELK3 Signaling. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302231. [PMID: 37822152 PMCID: PMC10646281 DOI: 10.1002/advs.202302231] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 08/25/2023] [Indexed: 10/13/2023]
Abstract
The involvement of endothelial barrier function in abdominal aortic aneurysm (AAA) and its upstream regulators remains unknown. Single-cell RNA sequencing shows that disrupted endothelial focal junction is an early (3 days) and persistent (28 days) event during Angiotensin II (Ang II)-induced AAA progression. Consistently, mRNA sequencing on human aortic dissection tissues confirmed downregulated expression of endothelial barrier-related genes. Aldehyde dehydrogenase 2 (ALDH2), a negative regulator of AAA, is found to be upregulated in the intimal media of AAA samples, leading to testing its role in early-stage AAA. ALDH2 knockdown/knockout specifically in endothelial cells (ECs) significantly increases expression of EC barrier markers related to focal adhesion and tight junction, restores endothelial barrier integrity, and suppresses early aortic dilation of AAA (7 and 14 days post-Ang II). Mechanically, ELK3 acts as an ALDH2 downstream regulator for endothelial barrier function preservation. At the molecular level, ALDH2 directly binds to LIN28B, a regulator of ELK3 mRNA stability, hindering LIN28B binding to ELK3 mRNA, thereby depressing ELK3 expression and impairing endothelial barrier function. Therefore, preserving vascular endothelial barrier integrity via ALDH2-specific knockdown in ECs holds therapeutic potential in the early management of AAAs.
Collapse
|
4
|
Sahibdad I, Khalid S, Chaudhry GR, Salim A, Begum S, Khan I. Zinc enhances the cell adhesion, migration, and self-renewal potential of human umbilical cord derived mesenchymal stem cells. World J Stem Cells 2023; 15:751-767. [PMID: 37545753 PMCID: PMC10401417 DOI: 10.4252/wjsc.v15.i7.751] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 05/15/2023] [Accepted: 06/06/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND Zinc (Zn) is the second most abundant trace element after Fe, present in the human body. It is frequently reported in association with cell growth and proliferation, and its deficiency is considered to be a major disease contributing factor. AIM To determine the effect of Zn on in vitro growth and proliferation of human umbilical cord (hUC)-derived mesenchymal stem cells (MSCs). METHODS hUC-MSCs were isolated from human umbilical cord tissue and characterized based on immunocytochemistry, immunophenotyping, and tri-lineage differentiation. The impact of Zn on cytotoxicity and proliferation was determined by MTT and Alamar blue assay. To determine the effect of Zn on population doubling time (PDT), hUC-MSCs were cultured in media with and without Zn for several passages. An in vitro scratch assay was performed to analyze the effect of Zn on the wound healing and migration capability of hUC-MSCs. A cell adhesion assay was used to test the surface adhesiveness of hUC-MSCs. Transcriptional analysis of genes involved in the cell cycle, proliferation, migration, and self-renewal of hUC-MSCs was performed by quantitative real-time polymerase chain reaction. The protein expression of Lin28, a pluripotency marker, was analyzed by immunocytochemistry. RESULTS Zn at lower concentrations enhanced the rate of proliferation but at higher concentrations (> 100 µM), showed concentration dependent cytotoxicity in hUC-MSCs. hUC-MSCs treated with Zn exhibited a significantly greater healing and migration rate compared to untreated cells. Zn also increased the cell adhesion rate, and colony forming efficiency (CFE). In addition, Zn upregulated the expression of genes involved in the cell cycle (CDC20, CDK1, CCNA2, CDCA2), proliferation (transforming growth factor β1, GDF5, hypoxia-inducible factor 1α), migration (CXCR4, VCAM1, VEGF-A), and self-renewal (OCT4, SOX2, NANOG) of hUC-MSCs. Expression of Lin28 protein was significantly increased in cells treated with Zn. CONCLUSION Our findings suggest that zinc enhances the proliferation rate of hUC-MSCs decreasing the PDT, and maintaining the CFE. Zn also enhances the cell adhesion, migration, and self-renewal of hUC-MSCs. These results highlight the essential role of Zn in cell growth and development.
Collapse
Affiliation(s)
- Iqra Sahibdad
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Shumaila Khalid
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - G Rasul Chaudhry
- Department of Biological Sciences, Oakland University, Rochester, MI 48309, United States
| | - Asmat Salim
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan
| | - Sumreen Begum
- Stem Cell Research Laboratory (SCRL), Sindh Institute of Urology and Transplantation (SIUT), Karachi 74200, Sindh, Pakistan
| | - Irfan Khan
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Sindh, Pakistan.
| |
Collapse
|
5
|
LIN28 Family in Testis: Control of Cell Renewal, Maturation, Fertility and Aging. Int J Mol Sci 2022; 23:ijms23137245. [PMID: 35806250 PMCID: PMC9266904 DOI: 10.3390/ijms23137245] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 06/20/2022] [Accepted: 06/25/2022] [Indexed: 12/12/2022] Open
Abstract
Male reproductive development starts early in the embryogenesis with somatic and germ cell differentiation in the testis. The LIN28 family of RNA-binding proteins promoting pluripotency has two members—LIN28A and LIN28B. Their function in the testis has been investigated but many questions about their exact role based on the expression patterns remain unclear. LIN28 expression is detected in the gonocytes and the migrating, mitotically active germ cells of the fetal testis. Postnatal expression of LIN28 A and B showed differential expression, with LIN28A expressed in the undifferentiated spermatogonia and LIN28B in the elongating spermatids and Leydig cells. LIN28 interferes with many signaling pathways, leading to cell proliferation, and it is involved in important testicular physiological processes, such as cell renewal, maturation, fertility, and aging. In addition, aberrant LIN28 expression is associated with testicular cancer and testicular disorders, such as hypogonadotropic hypogonadism and Klinefelter’s syndrome. This comprehensive review encompasses current knowledge of the function of LIN28 paralogs in testis and other tissues and cells because many studies suggest LIN28AB as a promising target for developing novel therapeutic agents.
Collapse
|
6
|
Luo J, Zou H, Deng L, Sun X, Yuan P, Li P. Lin28 Inhibits the Differentiation from Mouse Embryonic Stem Cells to Glial Lineage Cells through Upregulation of Yap1. Stem Cells Int 2021; 2021:6674283. [PMID: 33688355 PMCID: PMC7920735 DOI: 10.1155/2021/6674283] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 02/05/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023] Open
Abstract
The RNA-binding protein Lin28 regulates neurogliogenesis in mammals, independently of the let-7 microRNA. However, the detailed regulatory mechanism remains obscured. Here, we established Lin28a or Lin28b overexpression mouse embryonic stem cells (ESCs) and found that these cells expressed similar levels of the core pluripotent factors, such as Oct4 and Sox2, and increased Yap1 but decreased lineage-specific markers compared to the control ESCs. Further differentiation of these ESCs to neuronal and glial lineage cells revealed that Lin28a/b overexpression did not affect the expression of neuronal marker βIII-tubulin, but dramatically inhibited the glial lineage markers, such as Gfap and Mbp. Interestingly, overexpression of Yap1 in mouse ESCs phenocopied Lin28a/b overexpression ESCs by showing defect in glial cell differentiation. Inhibition of Yap1/Tead-mediated transcription with verteporfin partially rescued the differentiation defect of Lin28a/b overexpression ESCs. Mechanistically, we demonstrated that Lin28 can directly bind to Yap1 mRNA, and the induction of Yap1 by Lin28a in mESCs is independent of Let7. Taken together, our results unravel a novel Lin28-Yap1 regulatory axis during mESC to glial lineage cell differentiation, which may shed light on glial cell generation in vitro.
Collapse
Affiliation(s)
- Juan Luo
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Hailin Zou
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Liang Deng
- Department of General Surgery, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| | - Xiang Sun
- Department of Medical Bioinformatics, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou 510275, China
- Center for Stem Cell Biology and Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou 510275, China
| | - Ping Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou 510655, China
- Guangdong Institute of Gastroenterology, Guangzhou, Guangdong 510655, China
| | - Peng Li
- Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, China
| |
Collapse
|
7
|
Sato T, Kataoka K, Ito Y, Yokoyama S, Inui M, Mori M, Takahashi S, Akita K, Takada S, Ueno-Kudoh H, Asahara H. Lin28a/let-7 pathway modulates the Hox code via Polycomb regulation during axial patterning in vertebrates. eLife 2020; 9:53608. [PMID: 32479258 PMCID: PMC7259951 DOI: 10.7554/elife.53608] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2019] [Accepted: 05/18/2020] [Indexed: 01/06/2023] Open
Abstract
The body plan along the anteroposterior axis and regional identities are specified by the spatiotemporal expression of Hox genes. Multistep controls are required for their unique expression patterns; however, the molecular mechanisms behind the tight control of Hox genes are not fully understood. In this study, we demonstrated that the Lin28a/let-7 pathway is critical for axial elongation. Lin28a–/– mice exhibited axial shortening with mild skeletal transformations of vertebrae, which were consistent with results in mice with tail bud-specific mutants of Lin28a. The accumulation of let-7 in Lin28a–/– mice resulted in the reduction of PRC1 occupancy at the Hox cluster loci by targeting Cbx2. Consistently, Lin28a loss in embryonic stem-like cells led to aberrant induction of posterior Hox genes, which was rescued by the knockdown of let-7. These results suggest that the Lin28/let-7 pathway is involved in the modulation of the ‘Hox code’ via Polycomb regulation during axial patterning.
Collapse
Affiliation(s)
- Tempei Sato
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Kensuke Kataoka
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Yoshiaki Ito
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Research Core, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shigetoshi Yokoyama
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,Laboratory of Metabolism, National Institutes of Health, Bethesda, United States
| | - Masafumi Inui
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,Laboratory of Animal Regeneration Systemology, Meiji University, Kanagawa, Japan
| | - Masaki Mori
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Medical Chemistry, Shiga University of Medical Science, Shiga, Japan
| | - Satoru Takahashi
- Department of Anatomy and Embryology, University of Tsukuba, Ibaraki, Japan
| | - Keiichi Akita
- Department of Clinical Anatomy, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shuji Takada
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Hiroe Ueno-Kudoh
- Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,Reproduction Center, Yokohama City University, Yokohama, Japan
| | - Hiroshi Asahara
- Department of Systems BioMedicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Systems BioMedicine, National Research Institute for Child Health and Development, Tokyo, Japan.,AMED-CREST, Japan Agency for Medical Research and Development (AMED), Tokyo, Japan.,Department of Molecular Medicine, The Scripps Research Institute, La Jolla, United States
| |
Collapse
|
8
|
Cao G, Gao Z, Jiang Y, Chu M. Lin28 gene and mammalian puberty. Mol Reprod Dev 2020; 87:525-533. [PMID: 32363678 DOI: 10.1002/mrd.23347] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 04/21/2020] [Indexed: 11/10/2022]
Abstract
Lin28a and Lin28b, homologs of the Caenorhabditis elegans Lin28 gene, play important roles in cell pluripotency, reprogramming, and tumorigenicity. Recently, genome-wide association and transgenic studies showed that Lin28a and/or Lin28b gene were involved in the onset of mammalian puberty, the stage representing the attainment of reproduction capacity; however, the detailed mechanism of these genes in mammalian puberty remains largely unknown. The present paper reviews the research progress on the roles of Lin28a/b genes in the onset of mammalian puberty by analyzing the results coming from gene expression patterns, mutations, and transgenic studies, and put forward possible pathways for further studies on their roles in animal reproduction.
Collapse
Affiliation(s)
- Guiling Cao
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China.,College of Agriculture, Liaocheng University, Liaocheng, China
| | - Zeyang Gao
- College of Agriculture, Liaocheng University, Liaocheng, China
| | - Yunliang Jiang
- College of Animal Science and Veterinary Medicine, Shandong Agricultural University, Tai'an, China
| | - Mingxing Chu
- Key Laboratory of Animal (Poultry) Genetics Breeding and Reproduction, Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China.,Key Laboratory of Farm Animal Genetic Resources and Germplasm Innovation of Ministry of Agriculture, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
9
|
Wang S, Chim B, Su Y, Khil P, Wong M, Wang X, Foroushani A, Smith PT, Liu X, Li R, Ganesan S, Kanellopoulou C, Hafner M, Muljo SA. Enhancement of LIN28B-induced hematopoietic reprogramming by IGF2BP3. Genes Dev 2019; 33:1048-1068. [PMID: 31221665 PMCID: PMC6672051 DOI: 10.1101/gad.325100.119] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 05/16/2019] [Indexed: 01/07/2023]
Abstract
Fetal hematopoietic stem and progenitor cells (HSPCs) hold promise to cure a wide array of hematological diseases, and we previously found a role for the RNA-binding protein (RBP) Lin28b in respecifying adult HSPCs to resemble their fetal counterparts. Here we show by single-cell RNA sequencing that Lin28b alone was insufficient for complete reprogramming of gene expression from the adult toward the fetal pattern. Using proteomics and in situ analyses, we found that Lin28b (and its closely related paralog, Lin28a) directly interacted with Igf2bp3, another RBP, and their enforced co-expression in adult HSPCs reactivated fetal-like B-cell development in vivo more efficiently than either factor alone. In B-cell progenitors, Lin28b and Igf2bp3 jointly stabilized thousands of mRNAs by binding at the same sites, including those of the B-cell regulators Pax5 and Arid3a as well as Igf2bp3 mRNA itself, forming an autoregulatory loop. Our results suggest that Lin28b and Igf2bp3 are at the center of a gene regulatory network that mediates the fetal-adult hematopoietic switch. A method to efficiently generate induced fetal-like hematopoietic stem cells (ifHSCs) will facilitate basic studies of their biology and possibly pave a path toward their clinical application.
Collapse
Affiliation(s)
- Saifeng Wang
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Bryan Chim
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Yijun Su
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Pavel Khil
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Madeline Wong
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Xiantao Wang
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Amir Foroushani
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Patrick T Smith
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Xiuhuai Liu
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Rui Li
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Chrysi Kanellopoulou
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Markus Hafner
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Stefan A Muljo
- Laboratory of Immune System Biology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
10
|
Lee M, Nguyen TMT, Kim K. In-depth study of lin-28 suggests selectively conserved let-7 independent mechanism in Drosophila. Gene 2019; 687:64-72. [DOI: 10.1016/j.gene.2018.11.017] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 11/03/2018] [Accepted: 11/07/2018] [Indexed: 12/13/2022]
|
11
|
Lin28 and let-7 regulate the timing of cessation of murine nephrogenesis. Nat Commun 2019; 10:168. [PMID: 30635573 PMCID: PMC6329821 DOI: 10.1038/s41467-018-08127-4] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/12/2018] [Indexed: 01/10/2023] Open
Abstract
In humans and in mice the formation of nephrons during embryonic development reaches completion near the end of gestation, after which no new nephrons are formed. The final nephron complement can vary 10-fold, with reduced nephron number predisposing individuals to hypertension, renal, and cardiovascular diseases in later life. While the heterochronic genes lin28 and let-7 are well-established regulators of developmental timing in invertebrates, their role in mammalian organogenesis is not fully understood. Here we report that the Lin28b/let-7 axis controls the duration of kidney development in mice. Suppression of let-7 miRNAs, directly or via the transient overexpression of LIN28B, can prolong nephrogenesis and enhance kidney function potentially via upregulation of the Igf2/H19 locus. In contrast, kidney-specific loss of Lin28b impairs renal development. Our study reveals mechanisms regulating persistence of nephrogenic mesenchyme and provides a rationale for therapies aimed at increasing nephron mass. Nephrogenesis ceases after postnatal day 2 in the mouse or after the 36th week of gestation in humans, but how this is regulated is unclear. Here, the authors identify a role for the RNA-binding protein Lin28 and suppression of let-7 microRNA in regulating the duration of nephrogenesis.
Collapse
|
12
|
Romer-Seibert JS, Hartman NW, Moss EG. The RNA-binding protein LIN28 controls progenitor and neuronal cell fate during postnatal neurogenesis. FASEB J 2018; 33:3291-3303. [PMID: 30423261 PMCID: PMC6404560 DOI: 10.1096/fj.201801118r] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The RNA-binding protein LIN28 is known to regulate cell fate, tissue growth, and pluripotency; however, a unified understanding of its role at the cellular level has not been achieved. Here, we address its developmental activity in mammalian postnatal neurogenesis. Constitutive expression of LIN28 in progenitor cells of the mouse subventricular zone (SVZ) caused several distinct effects: 1) the number of differentiated neurons in the olfactory bulb was dramatically reduced, whereas the relative abundance of 2 neuronal subtypes was significantly altered, 2) the population of proliferating neural progenitors in the SVZ was reduced, whereas the proportion of neuroblasts was increased, and 3) the number of astrocytes was reduced, occasionally causing them to appear early. Thus, LIN28 acts at a poststem cell/predifferentiation step, and its continuous expression caused a precocious phenotype unlike in other experimental systems. Furthermore, for the first time in a vertebrate system, we separate the majority of the biologic role of LIN28 from its known activity of blocking the microRNA let-7 by using a circular RNA sponge. We find that although LIN28 has a multifaceted role in the number and types of cells produced during postnatal neurogenesis, it appears that its action through let-7 is responsible for only a fraction of these effects.—Romer-Seibert, J. S., Hartman, N. W., Moss, E. G. The RNA-binding protein LIN28 controls progenitor and neuronal cell fate during postnatal neurogenesis.
Collapse
Affiliation(s)
- Jennifer S Romer-Seibert
- Department of Molecular Biology, Graduate School of Biomedical Sciences, Rowan University, Stratford, New Jersey, USA; and
| | - Nathaniel W Hartman
- School of Natural Sciences and Mathematics, Stockton University, Galloway, New Jersey, USA
| | - Eric G Moss
- Department of Molecular Biology, Graduate School of Biomedical Sciences, Rowan University, Stratford, New Jersey, USA; and
| |
Collapse
|
13
|
RNA-binding protein LIN28B inhibits apoptosis through regulation of the AKT2/FOXO3A/BIM axis in ovarian cancer cells. Signal Transduct Target Ther 2018; 3:23. [PMID: 30174831 PMCID: PMC6117292 DOI: 10.1038/s41392-018-0026-5] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 07/12/2018] [Accepted: 07/22/2018] [Indexed: 02/06/2023] Open
Abstract
LIN28B is an evolutionarily conserved RNA-binding protein that regulates mRNA translation and miRNA let-7 maturation in embryonic stem cells and developing tissues. Increasing evidence demonstrates that LIN28B is activated in cancer and serves as a critical oncogene. However, the underlying molecular mechanisms of LIN28B function in tumorigenesis are still largely unknown. Here we report that LIN28B was expressed in over half of the patients with epithelial ovarian cancer who were examined (n = 584). Functional experiments demonstrated that LIN28B inhibited ovarian cancer cell apoptosis. Furthermore, we showed that the proapoptotic factor BIM played an essential role in the antiapoptotic function of LIN28B. RNA-IP microarray analysis suggested that LIN28B binds to mRNAs that are associated with the DNA damage pathway, such as AKT2, in ovarian cancer cells. By binding to AKT2 mRNA and enhancing its protein expression, LIN28B regulated FOXO3A protein phosphorylation and decreased the transcriptional level of BIM, which antagonized the antiapoptosis activity of LIN28B. Taken together, these results mechanistically linked LIN28B and the AKT2/FOXO3A/BIM axis to the apoptosis pathway. The findings may have important implications in the diagnosis and therapeutics of ovarian cancer. Researchers in China have uncovered the molecular mechanism behind the activity of a gene related to ovarian cancer. Xiaomin Zhong’s team at Sun Yat-Sen University knocked down the gene, LIN28B, in cancer cell lines and discovered that this increased their response to a chemical, which induces programmed cell death (PCD). By contrast, increasing LIN28B expression reduced PCD sensitivity, leading to the conclusion that LIN28B inhibits PCD in cancer cells. Examining gene expression in the knockdown lines revealed that LIN28B suppresses the activity of the PCD-related gene BIM. Further experiments showed that this happens through the modulation of genes upstream of BIM. These results demonstrate that LIN28B acts by regulating a genetic pathway, which regulates PCD. Altogether, this improved understanding of LIN28B may help with the diagnosis and therapy of ovarian cancer.
Collapse
|
14
|
Lightfoot HL, Miska EA, Balasubramanian S. Identification of small molecule inhibitors of the Lin28-mediated blockage of pre-let-7g processing. Org Biomol Chem 2018; 14:10208-10216. [PMID: 27731469 PMCID: PMC5433426 DOI: 10.1039/c6ob01945e] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Small molecules enhance Dicer processing of a let-7 miRNA precursor through antagonization of the Lin28–pre-let-7 interaction.
The protein Lin28 and microRNA let-7 play critical roles in mammalian development and human disease. Lin28 inhibits let-7 biogenesis through direct interaction with let-7 precursors (pre-let-7). Accumulating evidence in vitro and in vivo suggests this interaction plays a dominant role in embryonic stem cell self-renewal and tumorigenesis. Thus the Lin28–let-7 interaction might be an attractive drug target, if not for the well-known difficulties in targeting protein–RNA interactions with drugs. The identification and development of suitable probe molecules to further elucidate therapeutic potential, as well as mechanistic details of this pathway will be valuable. We report the development and application of a biophysical high-throughput screening assay for the identification of small molecule inhibitors of the Lin28–pre-let-7 interaction. A library of pharmacologically active small molecules was screened and several small molecule inhibitors were identified and biochemically validated. Of these four validated inhibitors, two compounds successfully restored processing of pre-let-7g in the presence of Lin28, validating the concept. Thus, we have identified examples of small molecule inhibitors of the interaction between Lin28 and pre-let-7. This study provides a proof of concept for small molecule inhibitors that antagonise the effects of Lin28 and enhance processing of let-7 miRNA.
Collapse
Affiliation(s)
- Helen L Lightfoot
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK.
| | - Eric A Miska
- Wellcome Trust Cancer Research UK Gurdon Institute, University of Cambridge, Tennis Court Rd, Cambridge, CB2 1QN, UK.
| | - Shankar Balasubramanian
- Department of Chemistry, University of Cambridge, Lensfield Road, Cambridge, CB2 1EW, UK. and Cancer Research UK, Cambridge Institute, Li Ka Shing Centre, Robinson Way, Cambridge, CB2 0RE, UK
| |
Collapse
|
15
|
Lee T, Pelletier J. The biology of DHX9 and its potential as a therapeutic target. Oncotarget 2018; 7:42716-42739. [PMID: 27034008 PMCID: PMC5173168 DOI: 10.18632/oncotarget.8446] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 03/16/2016] [Indexed: 12/25/2022] Open
Abstract
DHX9 is member of the DExD/H-box family of helicases with a “DEIH” sequence at its eponymous DExH-box motif. Initially purified from human and bovine cells and identified as a homologue of the Drosophila Maleless (MLE) protein, it is an NTP-dependent helicase consisting of a conserved helicase core domain, two double-stranded RNA-binding domains at the N-terminus, and a nuclear transport domain and a single-stranded DNA-binding RGG-box at the C-terminus. With an ability to unwind DNA and RNA duplexes, as well as more complex nucleic acid structures, DHX9 appears to play a central role in many cellular processes. Its functions include regulation of DNA replication, transcription, translation, microRNA biogenesis, RNA processing and transport, and maintenance of genomic stability. Because of its central role in gene regulation and RNA metabolism, there are growing implications for DHX9 in human diseases and their treatment. This review will provide an overview of the structure, biochemistry, and biology of DHX9, its role in cancer and other human diseases, and the possibility of targeting DHX9 in chemotherapy.
Collapse
Affiliation(s)
- Teresa Lee
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada.,Department of Oncology, McGill University, Montreal, Quebec, Canada.,Department of Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
16
|
Balzeau J, Menezes MR, Cao S, Hagan JP. The LIN28/let-7 Pathway in Cancer. Front Genet 2017; 8:31. [PMID: 28400788 PMCID: PMC5368188 DOI: 10.3389/fgene.2017.00031] [Citation(s) in RCA: 344] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 02/27/2017] [Indexed: 01/12/2023] Open
Abstract
Among all tumor suppressor microRNAs, reduced let-7 expression occurs most frequently in cancer and typically correlates with poor prognosis. Activation of either LIN28A or LIN28B, two highly related RNA binding proteins (RBPs) and proto-oncogenes, is responsible for the global post-transcriptional downregulation of the let-7 microRNA family observed in many cancers. Specifically, LIN28A binds the terminal loop of precursor let-7 and recruits the Terminal Uridylyl Transferase (TUTase) ZCCHC11 that polyuridylates pre-let-7, thereby blocking microRNA biogenesis and tumor suppressor function. For LIN28B, the precise mechanism responsible for let-7 inhibition remains controversial. Functionally, the decrease in let-7 microRNAs leads to overexpression of their oncogenic targets such as MYC, RAS, HMGA2, BLIMP1, among others. Furthermore, mouse models demonstrate that ectopic LIN28 expression is sufficient to drive and/or accelerate tumorigenesis via a let-7 dependent mechanism. In this review, the LIN28/let-7 pathway is discussed, emphasizing its role in tumorigenesis, cancer stem cell biology, metabolomics, metastasis, and resistance to ionizing radiation and several chemotherapies. Also, emerging evidence will be presented suggesting that molecular targeting of this pathway may provide therapeutic benefit in cancer.
Collapse
Affiliation(s)
- Julien Balzeau
- Department of Neurosurgery, University of Texas Health Science Center at Houston Houston, TX, USA
| | - Miriam R Menezes
- Department of Neurosurgery, University of Texas Health Science Center at Houston Houston, TX, USA
| | - Siyu Cao
- Department of Neurosurgery, University of Texas Health Science Center at Houston Houston, TX, USA
| | - John P Hagan
- Department of Neurosurgery, University of Texas Health Science Center at Houston Houston, TX, USA
| |
Collapse
|
17
|
Ma F, Zhou Z, Li N, Zheng L, Wu C, Niu B, Tang F, He X, Li G, Hua J. Lin28a promotes self-renewal and proliferation of dairy goat spermatogonial stem cells (SSCs) through regulation of mTOR and PI3K/AKT. Sci Rep 2016; 6:38805. [PMID: 27941834 PMCID: PMC5150521 DOI: 10.1038/srep38805] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 11/15/2016] [Indexed: 12/14/2022] Open
Abstract
Lin28a is a conserved RNA-binding protein that plays an important role in development, pluripotency, stemness maintenance, proliferation and self-renewal. Early studies showed that Lin28a serves as a marker of spermatogonial stem cells (SSCs) and promotes the proliferation capacity of mouse SSCs. However, there is little information about Lin28a in livestock SSCs. In this study, we cloned Capra hircus Lin28a CDS and found that it is evolutionarily conserved. Lin28a is widely expressed in different tissues of Capra hircus, but is expressed at a high level in the testis. Lin28a is specifically located in the cytoplasm of Capra hircus spermatogonial stem cells and may also be a marker of dairy goat spermatogonial stem cells. Lin28a promoted proliferation and maintained the self-renewal of GmGSCs-I-SB in vivo and in vitro. Lin28a-overexpressing GmGSCs-I-SB showed an enhanced proliferation rate, which might be due to increased PCNA expression. Moreover, Lin28a maintained the self-renewal of GmGSCs-I-SB by up-regulating the expression of OCT4, SOX2, GFRA1, PLZF and ETV5. Furthermore, we found that Lin28a may activate the AKT, ERK, and mTOR signaling pathways to promote the proliferation and maintain the self-renewal of GmGSCs-I-SB.
Collapse
Affiliation(s)
- Fanglin Ma
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Zhe Zhou
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Na Li
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Liming Zheng
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Chongyang Wu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Bowen Niu
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Furong Tang
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Xin He
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| | - Guangpeng Li
- Key Laboratory for Mammalian Reproductive Biology and Biotechnology, Ministry of Education, Inner Mongolia University, Hohhot, 010021, China
| | - Jinlian Hua
- College of Veterinary Medicine, Shaanxi Centre of Stem Cells Engineering &Technology, Northwest A&F University, Yangling, Shaanxi, 712100 China
| |
Collapse
|
18
|
Stage-Specific Timing of the microRNA Regulation of lin-28 by the Heterochronic Gene lin-14 in Caenorhabditis elegans. Genetics 2016; 205:251-262. [PMID: 27815363 DOI: 10.1534/genetics.116.195040] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Accepted: 10/20/2016] [Indexed: 01/01/2023] Open
Abstract
In normal development, the order and synchrony of diverse developmental events must be explicitly controlled. In the nematode Caenorhabditis elegans, the timing of larval events is regulated by hierarchy of proteins and microRNAs (miRNAs) known as the heterochronic pathway. These regulators are organized in feedforward and feedback interactions to form a robust mechanism for specifying the timing and execution of cell fates at successive stages. One member of this pathway is the RNA binding protein LIN-28, which promotes pluripotency and cell fate decisions in successive stages. Two genetic circuits control LIN-28 abundance: it is negatively regulated by the miRNA lin-4, and positively regulated by the transcription factor LIN-14 through a mechanism that was previously unknown. In this report, we used animals that lack lin-4 to elucidate LIN-14's activity in this circuit. We demonstrate that three let-7 family miRNAs-miR-48, miR-84, and miR-241-inhibit lin-28 expression. Furthermore, we show genetically that these miRNAs act between lin-14 and lin-28, and that they comprise the pathway by which lin-14 positively regulates lin-28 We also show that the lin-4 family member mir-237, also regulates early cell fates. Finally, we show that the expression of these miRNAs is directly inhibited by lin-14 activity, making them the first known targets of lin-14 that act in the heterochronic pathway.
Collapse
|
19
|
Bronevetsky Y, Burt TD, McCune JM. Lin28b Regulates Fetal Regulatory T Cell Differentiation through Modulation of TGF-β Signaling. THE JOURNAL OF IMMUNOLOGY 2016; 197:4344-4350. [PMID: 27793996 DOI: 10.4049/jimmunol.1601070] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 09/30/2016] [Indexed: 01/08/2023]
Abstract
Immune tolerance between the fetus and mother represents an active process by which the developing fetus must not mount immune responses to noninherited Ags on chimeric maternal cells that reside in fetal tissue. This is, in part, mediated by the suppressive influence of CD4+FOXP3+CD25+ regulatory T cells (Tregs). Fetal secondary lymphoid organs have an increased frequency of Tregs and, as compared with adult T cells, fetal naive CD4+ T cells exhibit a strong predisposition to differentiate into Tregs when stimulated. This effect is mediated by the TCR and TGF-β pathways, and fetal T cells show significantly increased Treg differentiation in response to anti-CD3 and TGF-β stimulation. Naive fetal T cells also exhibit increased signaling through the TGF-β pathway, with these cells demonstrating increased expression of the signaling mediators TGF-βRI, TGF-βRIII, and SMAD2, and higher levels of SMAD2/SMAD3 phosphorylation. Increased fetal Treg differentiation is mediated by the RNA-binding protein Lin28b, which is overexpressed in fetal T cells as compared with adult cells. When Lin28b expression is decreased in naive fetal T cells, they exhibit decreased Treg differentiation that is associated with decreased TGF-β signaling and lowered expression of TGF-βRI, TGF-βRIII, and SMAD2. Lin28b regulates the maturation of let-7 microRNAs, and these TGF-β signaling mediators are let-7 targets. We hypothesize that loss of Lin28b expression in fetal T cells leads to increased mature let-7, which causes decreased expression of TGF-βRI, TGF-βRIII, and SMAD2 proteins. A reduction in TGF-β signaling leads to reduced Treg numbers.
Collapse
Affiliation(s)
- Yelena Bronevetsky
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110;
| | - Trevor D Burt
- Division of Neonatology, Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94110; and.,Eli and Edythe Broad Center of Regeneration Medicine and Stem Cell Research, University of California, San Francisco, San Francisco, CA 94143
| | - Joseph M McCune
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94110
| |
Collapse
|
20
|
Corre C, Shinoda G, Zhu H, Cousminer DL, Crossman C, Bellissimo C, Goldenberg A, Daley GQ, Palmert MR. Sex-specific regulation of weight and puberty by the Lin28/let-7 axis. J Endocrinol 2016; 228:179-91. [PMID: 26698568 PMCID: PMC4772724 DOI: 10.1530/joe-15-0360] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/22/2015] [Indexed: 12/18/2022]
Abstract
Growth and pubertal timing differ in boys and girls. Variants in/near LIN28B associate with age at menarche (AAM) in genome-wide association studies and some AAM-related variants associate with growth in a sex-specific manner. Sex-specific growth patterns in response to Lin28b perturbation have been detected in mice, and overexpression of Lin28a has been shown to alter pubertal timing in female mice. To investigate further how Lin28a and Lin28b affect growth and puberty in both males and females, we evaluated Lin28b loss-of-function (LOF) mice and Lin28a gain-of-function (GOF) mice. Because both Lin28a and Lin28b can act via the conserved microRNA let-7, we also examined let-7 GOF mice. As reported previously, Lin28b LOF led to lighter body weights only in male mice while Lin28a GOF yielded heavier mice of both sexes. Let-7 GOF mice weighed less than controls, and males were more affected than females. Timing of puberty was assessed by vaginal opening (VO) and preputial separation (PS). Male Lin28b LOF and male let-7 GOF, but not female, mice displayed alteration of pubertal timing, with later PS than controls. In contrast, both male and female Lin28a GOF mice displayed late onset of puberty. Together, these data point toward a complex system of regulation by Lin28a, Lin28b, and let-7, in which Lin28b and let-7 can impact both puberty and growth in a sex-specific manner, raising the possibility that this pathway may contribute to differential regulation of male and female growth and puberty in humans.
Collapse
Affiliation(s)
- Christina Corre
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Gen Shinoda
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Hao Zhu
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Diana L Cousminer
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Christine Crossman
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Christian Bellissimo
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Anna Goldenberg
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - George Q Daley
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| | - Mark R Palmert
- Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada Division of EndocrinologyThe Hospital for Sick Children, 555 University Avenue, Toronto ON, M5G 1X8, CanadaDivision of Hematology/OncologyBoston Children's Hospital, Boston, Massachusetts, USADepartments of Pediatrics and Internal MedicineChildren's Research Institute, University of Texas Southwestern Medical Center, Dallas, Texas, USAInstitute for Molecular MedicineFinland (FIMM), University of Helsinki, Helsinki, FinlandGenetics and Genome Biology ProgramThe Hospital for Sick Children, Toronto, Ontario, CanadaDepartment of Computer ScienceUniversity of Toronto, Toronto, Ontario, CanadaDepartments of Paediatrics and PhysiologyThe University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
21
|
Abstract
LIN28 is an RNA-binding protein that is best known for its roles in promoting pluripotency via regulation of the microRNA let-7. However, recent studies have uncovered new roles for LIN28 and have revealed how it functions, suggesting that it is more than just a regulator of miRNA biogenesis. Together, these findings imply a new paradigm for LIN28 - as a gatekeeper molecule that regulates the transition between pluripotency and committed cell lineages, in both let-7-dependent and let-7-independent manners. Here, we provide an overview of LIN28 function in development and disease.
Collapse
Affiliation(s)
- Jennifer Tsialikas
- Department of Molecular Biology, Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| | - Jennifer Romer-Seibert
- Department of Molecular Biology, Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ 08084, USA
| |
Collapse
|
22
|
Sasaki K, Liu Y, Kim MH, Imai R. An RNA chaperone, AtCSP2, negatively regulates salt stress tolerance. PLANT SIGNALING & BEHAVIOR 2015; 10:e1042637. [PMID: 26252779 PMCID: PMC4623246 DOI: 10.1080/15592324.2015.1042637] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Abstract
Cold shock domain (CSD) proteins are RNA chaperones that destabilize RNA secondary structures. Arabidopsis Cold Shock Domain Protein 2 (AtCSP2), one of the 4 CSD proteins (AtCSP1-AtCSP4) in Arabidopsis, is induced during cold acclimation but negatively regulates freezing tolerance. Here, we analyzed the function of AtCSP2 in salt stress tolerance. A double mutant, with reduced AtCSP2 and no AtCSP4 expression (atcsp2-3 atcsp4-1), displayed higher survival rates after salt stress. In addition, overexpression of AtCSP2 resulted in reduced salt stress tolerance. These data demonstrate that AtCSP2 acts as a negative regulator of salt stress tolerance in Arabidopsis.
Collapse
Affiliation(s)
- Kentaro Sasaki
- Hokkaido Agricultural Research Center (HARC); National Agriculture and Food Research Organization (NARO); Toyohira-ku, Sapporo, Japan
- These authors contributed equally to this work
| | - Yuelin Liu
- Graduate School of Agriculture; Hokkaido University; Kita-ku, Sapporo, Japan
- These authors contributed equally to this work
| | - Myung-Hee Kim
- Hokkaido Agricultural Research Center (HARC); National Agriculture and Food Research Organization (NARO); Toyohira-ku, Sapporo, Japan
- Center for Plant Aging Research; Institute for Basic Science (IBS), Daegu, Republic of Korea
| | - Ryozo Imai
- Hokkaido Agricultural Research Center (HARC); National Agriculture and Food Research Organization (NARO); Toyohira-ku, Sapporo, Japan
- Graduate School of Agriculture; Hokkaido University; Kita-ku, Sapporo, Japan
- Correspondence to: Ryozo Imai;
| |
Collapse
|
23
|
New Proteomic Insights on the Role of NPR-A in Regulating Self-Renewal of Embryonic Stem Cells. Stem Cell Rev Rep 2014; 10:561-72. [DOI: 10.1007/s12015-014-9517-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
24
|
Madison BB, Liu Q, Zhong X, Hahn CM, Lin N, Emmett MJ, Stanger BZ, Lee JS, Rustgi AK. LIN28B promotes growth and tumorigenesis of the intestinal epithelium via Let-7. Genes Dev 2013; 27:2233-45. [PMID: 24142874 PMCID: PMC3814644 DOI: 10.1101/gad.224659.113] [Citation(s) in RCA: 99] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The RNA-binding proteins LIN28A and LIN28B have diverse functions in cellular reprogramming, growth, and oncogenesis. Madison et al. discover that intestine targeted expression of LIN28B causes intestinal hypertrophy, crypt expansion, and adenocarcinoma formation. Modulation of Let-7 levels via deletion of the mirLet7c2/mirLet7b genes recapitulated these effects, and intestine-specific Let-7 expression reversed the hypertrophy and Paneth cell depletion caused by Lin28b. These results demonstrate that Let-7 miRNAs are critical for repressing intestinal tissue growth and that LIN28B can act as an oncogene. The RNA-binding proteins LIN28A and LIN28B have diverse functions in embryonic stem cells, cellular reprogramming, growth, and oncogenesis. Many of these effects occur via direct inhibition of Let-7 microRNAs (miRNAs), although Let-7-independent effects have been surmised. We report that intestine targeted expression of LIN28B causes intestinal hypertrophy, crypt expansion, and Paneth cell loss. Furthermore, LIN28B fosters intestinal polyp and adenocarcinoma formation. To examine potential Let-7-independent functions of LIN28B, we pursued ribonucleoprotein cross-linking, immunoprecipitation, and high-throughput sequencing (CLIP-seq) to identify direct RNA targets. This revealed that LIN28B bound a substantial number of mRNAs and modestly augmented protein levels of these target mRNAs in vivo. Conversely, Let-7 had a profound effect; modulation of Let-7 levels via deletion of the mirLet7c2/mirLet7b genes recapitulated effects of Lin28b overexpression. Furthermore, intestine-specific Let-7 expression could reverse hypertrophy and Paneth cell depletion caused by Lin28b. This was independent of effects on insulin–PI3K–mTOR signaling. Our study reveals that Let-7 miRNAs are critical for repressing intestinal tissue growth and promoting Paneth cell differentiation. Let-7-dependent effects of LIN28B may supersede Let-7-independent effects on intestinal tissue growth. In summary, LIN28B can definitively act as an oncogene in the absence of canonical genetic alterations.
Collapse
Affiliation(s)
- Blair B Madison
- Division of Gastroenterology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
TAN CHENG, LI WENFEI, WANG WEI. SPECIFIC BINDING OF A SHORT miRNA SEQUENCE BY ZINC KNUCKLES OF Lin28: A MOLECULAR DYNAMICS SIMULATION STUDY. JOURNAL OF THEORETICAL & COMPUTATIONAL CHEMISTRY 2013. [DOI: 10.1142/s0219633613410150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Protein Lin28 recognizes pre-let-7 microRNAs (miRNAs) through direct interactions between its zinc-knuckle type zinc-finger (ZnF) domains and the terminal loop of pre-let-7, resulting in the inhibition of the synthesis of mature let-7 miRNAs. Despite the physiological importance, the involved conformational changes and energetic factors contributing to the binding affinity and specificity remain unclear. We conducted molecular dynamics (MD) simulations in conjunction with molecular mechanics/generalized born surface area (MM/GBSA) and energy decomposition calculations to investigate the RNA binding-induced conformational changes of the ZnFs and the residual level energetic factors that influence the binding affinity and specificity. We showed that the binding of the RNA results in the inter-domain conformational changes of the two ZnF domains, including changes of the spatial relationships of several nucleobase-binding amino acids. We also observed mutation-induced weakening of the affinity of the Lin28–pre-let-7 binding, which reveals the importance of the stacking interactions between the side-chains of Tyr140, His148, His162 and the bases of nucleic acid G2 and G5 in the specific recognition of pre-let-7 by the ZnFs of Lin28.
Collapse
Affiliation(s)
- CHENG TAN
- National Laboratory of Solid State Microstructure and Department of Physics, Nanjing University, Nanjing, 210093, P. R. China
| | - WENFEI LI
- National Laboratory of Solid State Microstructure and Department of Physics, Nanjing University, Nanjing, 210093, P. R. China
| | - WEI WANG
- National Laboratory of Solid State Microstructure and Department of Physics, Nanjing University, Nanjing, 210093, P. R. China
| |
Collapse
|
26
|
Spence T, Perotti C, Sin-Chan P, Picard D, Wu W, Singh A, Anderson C, Blough MD, Cairncross JG, Lafay-Cousin L, Strother D, Hawkins C, Narendran A, Huang A, Chan JA. A novel C19MC amplified cell line links Lin28/let-7 to mTOR signaling in embryonal tumor with multilayered rosettes. Neuro Oncol 2013; 16:62-71. [PMID: 24311633 DOI: 10.1093/neuonc/not162] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
BACKGROUND Embryonal tumor with multilayered rosettes (ETMR) is an aggressive central nervous system primitive neuroectodermal tumor (CNS-PNET) variant. ETMRs have distinctive histology, amplification of the chromosome 19 microRNA cluster (C19MC) at chr19q13.41-42, expression of the RNA binding protein Lin28, and dismal prognosis. Functional and therapeutic studies of ETMR have been limited by a lack of model systems. METHODS We have established a first cell line, BT183, from a case of ETMR and characterized its molecular and cellular features. LIN28 knockdown was performed in BT183 to examine the potential role of Lin28 in regulating signaling pathway gene expression in ETMR. Cell line findings were corroborated with immunohistochemical studies in ETMR tissues. A drug screen of 73 compounds was performed to identify potential therapeutic targets. RESULTS The BT183 line maintains C19MC amplification, expresses C19MC-encoded microRNAs, and is tumor initiating. ETMRs, including BT183, have high LIN28 expression and low let-7 miRNA expression, and show evidence of mTOR pathway activation. LIN28 knockdown increases let-7 expression and decreases expression of IGF/PI3K/mTOR pathway components. Pharmacologic inhibition of the mTOR pathway reduces BT183 cell viability. CONCLUSIONS BT183 retains key genetic and histologic features of ETMR. In ETMR, Lin28 is not only a diagnostic marker but also a regulator of genes involved in growth and metabolism. Our findings indicate that inhibitors of the IGF/PI3K/mTOR pathway may be promising novel therapies for these fatal embryonal tumors. As the first patient-derived cell line of these rare tumors, BT183 is an important, unique reagent for investigating ETMR biology and therapeutics.
Collapse
Affiliation(s)
- Tara Spence
- Corresponding authors: Jennifer A. Chan, MD, Department of Pathology & Laboratory Medicine, University of Calgary, HRIC 2A25, 3330 Hospital Drive NW, Calgary, Alberta, Canada T2N 4N1. ); Annie Huang, MD, PhD, Division of Hematology-Oncology, Department of Pediatrics, The Hospital for Sick Children, TMDT, 11-401P, 101 College St., Toronto, Ontario, Canada M5G 1L7 (
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Seabrook JL, Cantlon JD, Cooney AJ, McWhorter EE, Fromme BA, Bouma GJ, Anthony RV, Winger QA. Role of LIN28A in mouse and human trophoblast cell differentiation. Biol Reprod 2013; 89:95. [PMID: 24006280 DOI: 10.1095/biolreprod.113.109868] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Proper regulation of trophoblast proliferation, differentiation, and function are critical for placenta development and function. The RNA-binding protein, LIN28A, has been well characterized as a potent regulator of differentiation in embryonic stem cells; however, little is known about the function of LIN28A in the placenta. We assessed LIN28A in vitro using mouse trophoblast stem (mTS) cells and human trophoblast cells (ACH-3P). We observed that LIN28A decreased and let-7 miRNA increased when mTS cells were induced to differentiate into mouse trophoblast giant cells (mTGCs) upon the removal of FGF4, heparin and conditioned medium. Similarly, we observed that LIN28A decreased in ACH-3P cells induced to syncytialize with forskolin treatment. To assess LIN28A in vivo we examined Embryonic Day 11.5 mouse placenta and observed abundant LIN28A in the chorioallantoic interface and labyrinth layer, with little LIN28A staining in spongiotrophoblast or differentiated mTGCs. Additionally, shRNA-mediated LIN28A knockdown in ACH-3P cells resulted in increased spontaneous syncytialization, and increased levels of syncytiotrophoblast markers hCG, LGALS13, and ERVW-1 mRNA. Additionally, targeted degradation of LIN28A mRNA increased responsiveness to forskolin-induced differentiation. In contrast, targeted degradation of Lin28a mRNA in mTS cells did not alter cell phenotype when maintained under proliferative culture conditions. Together, these data establish that LIN28A has a functional role in regulating trophoblast differentiation and function, and that loss of LIN28A in human trophoblast is sufficient to induce differentiation, but does not induce differentiation in the mouse.
Collapse
Affiliation(s)
- Jill L Seabrook
- Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Mayr F, Heinemann U. Mechanisms of Lin28-mediated miRNA and mRNA regulation--a structural and functional perspective. Int J Mol Sci 2013; 14:16532-53. [PMID: 23939427 PMCID: PMC3759924 DOI: 10.3390/ijms140816532] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 07/22/2013] [Accepted: 07/25/2013] [Indexed: 12/14/2022] Open
Abstract
Lin28 is an essential RNA-binding protein that is ubiquitously expressed in embryonic stem cells. Its physiological function has been linked to the regulation of differentiation, development, and oncogenesis as well as glucose metabolism. Lin28 mediates these pleiotropic functions by inhibiting let-7 miRNA biogenesis and by modulating the translation of target mRNAs. Both activities strongly depend on Lin28’s RNA-binding domains (RBDs), an N-terminal cold-shock domain (CSD) and a C-terminal Zn-knuckle domain (ZKD). Recent biochemical and structural studies revealed the mechanisms of how Lin28 controls let-7 biogenesis. Lin28 binds to the terminal loop of pri- and pre-let-7 miRNA and represses their processing by Drosha and Dicer. Several biochemical and structural studies showed that the specificity of this interaction is mainly mediated by the ZKD with a conserved GGAGA or GGAGA-like motif. Further RNA crosslinking and immunoprecipitation coupled to high-throughput sequencing (CLIP-seq) studies confirmed this binding motif and uncovered a large number of new mRNA binding sites. Here we review exciting recent progress in our understanding of how Lin28 binds structurally diverse RNAs and fulfills its pleiotropic functions.
Collapse
Affiliation(s)
- Florian Mayr
- Crystallography, Max-Delbrück Center for Molecular Medicine, Robert-Rössle Straße 10, Berlin 13125, Germany; E-Mail:
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 6, Berlin 14195, Germany
| | - Udo Heinemann
- Crystallography, Max-Delbrück Center for Molecular Medicine, Robert-Rössle Straße 10, Berlin 13125, Germany; E-Mail:
- Institute for Chemistry and Biochemistry, Freie Universität Berlin, Takustraße 6, Berlin 14195, Germany
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +49-30-9406-3420; Fax: +49-30-9406-2548
| |
Collapse
|
29
|
Graf R, Munschauer M, Mastrobuoni G, Mayr F, Heinemann U, Kempa S, Rajewsky N, Landthaler M. Identification of LIN28B-bound mRNAs reveals features of target recognition and regulation. RNA Biol 2013; 10:1146-59. [PMID: 23770886 PMCID: PMC3849162 DOI: 10.4161/rna.25194] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The conserved human LIN28 RNA-binding proteins function in development, maintenance of pluripotency and oncogenesis. We used PAR-CLIP and a newly developed variant of this method, iDo-PAR-CLIP, to identify LIN28B targets as well as sites bound by the individual RNA-binding domains of LIN28B in the human transcriptome at nucleotide resolution. The position of target binding sites reflected the known structural relative orientation of individual LIN28B-binding domains, validating iDo-PAR-CLIP. Our data suggest that LIN28B directly interacts with most expressed mRNAs and members of the let-7 microRNA family. The Lin28-binding motif detected in pre-let-7 was enriched in mRNA sequences bound by LIN28B. Upon LIN28B knockdown, cell proliferation and the cell cycle were strongly impaired. Quantitative shotgun proteomics of LIN28B depleted cells revealed significant reduction of protein synthesis from its RNA targets. Computational analyses provided evidence that the strength of protein synthesis reduction correlated with the location of LIN28B binding sites within target transcripts.
Collapse
Affiliation(s)
- Robin Graf
- Systems Biology of Gene Regulatory Elements; Max-Delbrück-Center for Molecular Medicine; Berlin, Germany
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Zhou J, Ng SB, Chng WJ. LIN28/LIN28B: an emerging oncogenic driver in cancer stem cells. Int J Biochem Cell Biol 2013; 45:973-978. [PMID: 23420006 DOI: 10.1016/j.biocel.2013.02.006] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2012] [Revised: 01/31/2013] [Accepted: 02/07/2013] [Indexed: 10/27/2022]
Abstract
LIN28 (LIN28A) is a reprogramming factor and conserved RNA-binding protein. LIN28B is the only homolog of LIN28 in humans, sharing structure and certain function. LIN28/LIN28B has been identified to be overexpressed in a wide range of solid tumors and hematological malignancies. Blockage of let-7 miRNA biogensis and subsequent derepression of let-7 miRNA target genes by LIN28/LIN28B play important roles in cancer progression and metastasis. We will first provide an overview of LIN28/LIN28B gene and protein structures, followed by summary of the studies that showed their aberrant expression in primary human cancers and relevant clinical significance with emphasis on their roles in formation of cancer stem cells. Next, we will highlight the current knowledge of LIN28/LIN28B regulators and molecular mechanisms of LIN28/LIN28B-mediated oncogenesis. The potential medical applications for targeting LIN28/LIN28B will also be discussed in this review.
Collapse
Affiliation(s)
- Jianbiao Zhou
- Cancer Science Institute of Singapore, National University of Singapore, Singapore.
| | | | | |
Collapse
|
31
|
Hafner M, Max KEA, Bandaru P, Morozov P, Gerstberger S, Brown M, Molina H, Tuschl T. Identification of mRNAs bound and regulated by human LIN28 proteins and molecular requirements for RNA recognition. RNA (NEW YORK, N.Y.) 2013; 19:613-26. [PMID: 23481595 PMCID: PMC3677277 DOI: 10.1261/rna.036491.112] [Citation(s) in RCA: 133] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Human LIN28A and LIN28B are RNA-binding proteins (RBPs) conserved in animals with important roles during development and stem cell reprogramming. We used Photoactivatable-Ribonucleoside-Enhanced Crosslinking and Immunoprecipitation (PAR-CLIP) in HEK293 cells and identified a largely overlapping set of ∼3000 mRNAs at ∼9500 sites located in the 3' UTR and CDS. In vitro and in vivo, LIN28 preferentially bound single-stranded RNA containing a uridine-rich element and one or more flanking guanosines and appeared to be able to disrupt base-pairing to access these elements when embedded in predicted secondary structure. In HEK293 cells, LIN28 protein binding mildly stabilized target mRNAs and increased protein abundance. The top targets were its own mRNAs and those of other RBPs and cell cycle regulators. Alteration of LIN28 protein levels also negatively regulated the abundance of some but not all let-7 miRNA family members, indicating sequence-specific binding of let-7 precursors to LIN28 proteins and competition with cytoplasmic miRNA biogenesis factors.
Collapse
Affiliation(s)
- Markus Hafner
- Howard Hughes Medical Institute and Laboratory for RNA Molecular Biology, The Rockefeller University, New York, New York 10065, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Gaytan F, Sangiao-Alvarellos S, Manfredi-Lozano M, García-Galiano D, Ruiz-Pino F, Romero-Ruiz A, León S, Morales C, Cordido F, Pinilla L, Tena-Sempere M. Distinct expression patterns predict differential roles of the miRNA-binding proteins, Lin28 and Lin28b, in the mouse testis: studies during postnatal development and in a model of hypogonadotropic hypogonadism. Endocrinology 2013; 154:1321-36. [PMID: 23337528 DOI: 10.1210/en.2012-1745] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Lin28 (also termed Lin28a) and Lin28b are related RNA-binding proteins, involved in the control of microRNA synthesis, especially of the let-7 family, with putative functions in early (embryo) development. However, their roles during postnatal maturation remain ill defined. Despite the general assumption that Lin28 and Lin28b share similar targets and functions, conclusive demonstration of such redundancy is still missing. In addition, recent observations suggest a role of Lin28 proteins in mammalian reproduction, which is yet to be defined. We document herein the patterns of RNA expression and protein distribution of Lin28 and Lin28b in mouse testis during postnatal development and in a model of hypogonadotropic hypogonadism as a result of inactivation of the kisspeptin receptor, Gpr54. Lin28 and Lin28b mRNAs were expressed in mouse testis across postnatal maturation, but their levels disparately varied between neonatal and pubertal periods, with peak Lin28 levels in infantile testes and sustained elevation of Lin28b mRNA in young adult male gonads, where relative levels of let-7a and let-7b miRNAs were significantly suppressed. In addition, Lin28 peptides displayed totally different patterns of cellular distribution in mouse testis: Lin28 was located in undifferentiated and type-A1 spermatogonia, whereas Lin28b was confined to spermatids and interstitial Leydig cells. These profiles were perturbed in Gpr54 null mouse testis, which showed preserved but irregular Lin28 signal and absence of Lin28b peptide, which was rescued by administration of gonadotropins, mainly hCG (as super-agonist of LH). In addition, increased relative levels of Lin28, but not Lin28b, mRNA and of let-7a/let-7b miRNAs were observed in Gpr54 KO mouse testes. Altogether, our data are the first to document the divergent patterns of cellular distribution and mRNA expression of Lin28 and Lin28b in the mouse testis along postnatal maturation and their alteration in a model of congenital hypogonadotropic hypogonadism. Our findings suggest distinct functional roles of these two related, but not overlapping, miRNA-binding proteins in the male gonad.
Collapse
MESH Headings
- Animals
- Base Sequence
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Disease Models, Animal
- Gene Expression Regulation, Developmental
- Hypogonadism/congenital
- Hypogonadism/genetics
- Hypogonadism/metabolism
- Immunohistochemistry
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Models, Biological
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Receptors, G-Protein-Coupled/deficiency
- Receptors, G-Protein-Coupled/genetics
- Receptors, Kisspeptin-1
- Reverse Transcriptase Polymerase Chain Reaction
- Spermatogenesis/genetics
- Spermatogenesis/physiology
- Testis/growth & development
- Testis/metabolism
Collapse
Affiliation(s)
- Francisco Gaytan
- Department of Cell Biology, Physiology and Immunology, Faculty of Medicine, University of Córdoba, Avda. Menéndez Pidal s/n, 14004 Córdoba, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Faas L, Warrander FC, Maguire R, Ramsbottom SA, Quinn D, Genever P, Isaacs HV. Lin28 proteins are required for germ layer specification in Xenopus. Development 2013; 140:976-86. [PMID: 23344711 DOI: 10.1242/dev.089797] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Lin28 family proteins share a unique structure, with both zinc knuckle and cold shock RNA-binding domains, and were originally identified as regulators of developmental timing in Caenorhabditis elegans. They have since been implicated as regulators of pluripotency in mammalian stem cells in culture. Using Xenopus tropicalis, we have undertaken the first analysis of the effects on the early development of a vertebrate embryo resulting from global inhibition of the Lin28 family. The Xenopus genome contains two Lin28-related genes, lin28a and lin28b. lin28a is expressed zygotically, whereas lin28b is expressed both zygotically and maternally. Both lin28a and lin28b are expressed in pluripotent cells of the Xenopus embryo and are enriched in cells that respond to mesoderm-inducing signals. The development of axial and paraxial mesoderm is severely abnormal in lin28 knockdown (morphant) embryos. In culture, the ability of pluripotent cells from the embryo to respond to the FGF and activin/nodal-like mesoderm-inducing pathways is compromised following inhibition of lin28 function. Furthermore, there are complex effects on the temporal regulation of, and the responses to, mesoderm-inducing signals in lin28 morphant embryos. We provide evidence that Xenopus lin28 proteins play a key role in choreographing the responses of pluripotent cells in the early embryo to the signals that regulate germ layer specification, and that this early function is probably independent of the recognised role of Lin28 proteins in negatively regulating let-7 miRNA biogenesis.
Collapse
Affiliation(s)
- Laura Faas
- Area 11, Department of Biology, University of York, York, YO10 5DD, UK
| | | | | | | | | | | | | |
Collapse
|
34
|
Hepatitis B virus X protein upregulates Lin28A/Lin28B through Sp-1/c-Myc to enhance the proliferation of hepatoma cells. Oncogene 2013; 33:449-60. [PMID: 23318446 DOI: 10.1038/onc.2012.618] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2012] [Revised: 11/02/2012] [Accepted: 11/14/2012] [Indexed: 12/17/2022]
Abstract
Hepatitis B virus X protein (HBx) plays critical roles in the pathogenesis of hepatocellular carcinoma (HCC). Here, we were interested in knowing whether the oncogene Lin28A and its homolog Lin28B are involved in the hepatocarcinogenesis mediated by HBx. We showed that the expression levels of Lin28A and Lin28B were increased in clinical HCC tissues, HepG2.2.15 cell line and liver tissues of p21-HBx transgenic mice. Interestingly, the expression levels of HBx were positively associated with those of Lin28A/Lin28B in clinical HCC tissues. Moreover, the overexpression of HBx resulted in the upregulation of Lin28A/Lin28B in hepatoma HepG2/H7402 cell lines by transient transfection, suggesting that HBx was able to upregulate Lin28A and Lin28B. Then, we examined the mechanism by which HBx upregulated Lin28A and Lin28B. We identified that the promoter region of Lin28A regulated by HBx was located at nt -235/-66 that contained Sp-1 binding element. Co-immunoprecipitation showed that HBx was able to interact with Sp-1 in HepG2-X cells. Moreover, chromatin immunoprecipitation (ChIP) demonstrated that HBx could bind to the promoter of Lin28A, which failed to work when Sp-1 was silenced. Electrophoretic mobility shift assay (EMSA) further identified that HBx was able to interact with Sp-1 element in Lin28A promoter via transcription factor Sp-1. In addition, we found that c-Myc was involved in the activation of Lin28B mediated by HBx. In function, Lin28A/Lin28B played important roles in HBx-enhanced proliferation of hepatoma cells in vitro and in vivo. In conclusion, HBx activates Lin28A/Lin28B through Sp-1/c-Myc in hepatoma cells. Lin28A/Lin28B serves as key driver genes in HBx-induced hepatocarcinogenesis.
Collapse
|
35
|
Kallen AN, Ma J, Huang Y. Does Lin28 Antagonize miRNA-Mediated Repression by Displacing miRISC from Target mRNAs? Front Genet 2012; 3:240. [PMID: 23162570 PMCID: PMC3499807 DOI: 10.3389/fgene.2012.00240] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2012] [Accepted: 10/20/2012] [Indexed: 12/26/2022] Open
Abstract
Lin28 is a developmentally regulated RNA-binding protein that plays important roles in diverse physiological and pathological processes including oncogenesis and brain synaptic function. These pleiotropic roles of Lin28 are mechanistically linked both to its ability to directly stimulate translation of genes involved primarily in cell growth and metabolism and to its ability to block biogenesis of a subset of miRNAs including the let-7 family of miRNAs. In the case of direct stimulation of gene expression, Lin28 binds to targeted mRNAs through recognition of Lin28-responsive elements (LREs) within mRNAs and recruits RNA helicase A (RHA) to promote translation. RHA belongs to the DEAD-box protein family of RNA helicases, which generally catalyze ATP-dependent unwinding of RNA duplexes or remodeling of ribonucleoprotein complexes (RNPs). Since any given mRNA can potentially be inhibited by miRNAs bearing complementary sequences, we hypothesize that binding of Lin28 to LREs not only nucleates the binding of multiple Lin28 molecules to the same mRNA, but also leads to remodeling of RNPs through recruitment of RHA and causes release of inhibitory miRNA-induced silencing complexes bound to the mRNA. This mode of action may contribute to Lin28-mediated stimulation of translation in both tumor and neuronal cells.
Collapse
Affiliation(s)
- Amanda N Kallen
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale Stem Cell Center, Yale University School of Medicine New Haven, CT, USA
| | | | | |
Collapse
|
36
|
Cho J, Chang H, Kwon SC, Kim B, Kim Y, Choe J, Ha M, Kim YK, Kim VN. LIN28A is a suppressor of ER-associated translation in embryonic stem cells. Cell 2012; 151:765-777. [PMID: 23102813 DOI: 10.1016/j.cell.2012.10.019] [Citation(s) in RCA: 183] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Revised: 09/08/2012] [Accepted: 10/01/2012] [Indexed: 02/07/2023]
Abstract
LIN28 plays a critical role in developmental transition, glucose metabolism, and tumorigenesis. At the molecular level, LIN28 is known to repress maturation of let-7 microRNAs and enhance translation of certain mRNAs. In this study, we obtain a genome-wide view of the molecular function of LIN28A in mouse embryonic stem cells by carrying out RNA crosslinking-immunoprecipitation-sequencing (CLIP-seq) and ribosome footprinting. We find that, in addition to let-7 precursors, LIN28A binds to a large number of spliced mRNAs. LIN28A recognizes AAGNNG, AAGNG, and less frequently UGUG, which are located in the terminal loop of a small hairpin. LIN28A is localized to the periendoplasmic reticulum (ER) area and inhibits translation of mRNAs that are destined for the ER, reducing the synthesis of transmembrane proteins, ER or Golgi lumen proteins, and secretory proteins. Our study suggests a selective regulatory mechanism for ER-associated translation and reveals an unexpected role of LIN28A as a global suppressor of genes in the secretory pathway.
Collapse
Affiliation(s)
- Jun Cho
- Institute for Basic Science, Seoul National University, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - Hyeshik Chang
- Institute for Basic Science, Seoul National University, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - S Chul Kwon
- Institute for Basic Science, Seoul National University, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - Baekgyu Kim
- Institute for Basic Science, Seoul National University, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - Yoosik Kim
- Institute for Basic Science, Seoul National University, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - Junho Choe
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | - Minju Ha
- Institute for Basic Science, Seoul National University, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea
| | - Yoon Ki Kim
- School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Korea
| | - V Narry Kim
- Institute for Basic Science, Seoul National University, Seoul 151-742, Korea; School of Biological Sciences, Seoul National University, Seoul 151-742, Korea.
| |
Collapse
|
37
|
Brzuzan P, Woźny M, Wolińska L, Piasecka A. Expression profiling in vivo demonstrates rapid changes in liver microRNA levels of whitefish (Coregonus lavaretus) following microcystin-LR exposure. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2012; 122-123:188-196. [PMID: 22819808 DOI: 10.1016/j.aquatox.2012.07.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2012] [Revised: 06/04/2012] [Accepted: 07/02/2012] [Indexed: 06/01/2023]
Abstract
At present, little is known about the role of miRNAs in liver response of fish to the cyanobacterial hepatotoxin microcystin-LR (MC-LR) treatment, despite the fact that the exposure is thought to underlie multiple acute and chronic effects. To address this question, we used the Real-Time PCR method to examine the differential expression of 6 miRNAs putatively playing roles in signal transduction (let-7c, miR-9b), apoptosis and cell cycle (miR-16a, miR-21a, miR-34a) and fatty acid metabolism (miR-122) in whitefish (Coregonus lavaretus) liver, during the first 48h after intraperitoneal injection of MC-LR (100 μg/kg body weight). In addition, we analyzed expression levels of 8 mRNAs and p53 protein, known to be involved in the cell response on the exposure to environmental stressors. Following the challenge we observed a rapid and transient increase in the mean (n=5) levels of individual miRNA expression (from 2.7-fold for miR-122 to 6.8-fold for let-7c), compared to the respective levels in control fish, which mostly peaked at 24h of the experiment. This increase was correlated with a reduction in the expression of mRNAs of genes coding for ferritin H (frih) and HNK Ras -like protein (p-ras) and an overexpression of mRNAs of genes coding for bcl2-associated X protein (bax), cyclin dependent kinase inhibitor 1a (cdkn1a), dicer (dcr), histone 2A (h2a) and p53. Expression of the remaining caspase 6 (cas6) mRNA did not change over 48 h of the treatment. Moreover, exposure to MC-LR did not alter whitefish p53 protein levels. Bearing in mind a variety of likely silencing targets for, and the onset of, the aberrant miRNA expression it may be concluded that they are involved in molecular pathways, such as liver cell metabolism, cell cycle regulation and apoptosis, and may contribute to the early phase of MC-LR induced hepatotoxicity.
Collapse
Affiliation(s)
- P Brzuzan
- Department of Environmental Biotechnology, Faculty of Environmental Sciences, University of Warmia and Mazury in Olsztyn, Olsztyn, Poland.
| | | | | | | |
Collapse
|
38
|
Qiao C, Ma J, Xu J, Xie M, Ma W, Huang Y. Drosha mediates destabilization of Lin28 mRNA targets. Cell Cycle 2012; 11:3590-8. [PMID: 22935707 DOI: 10.4161/cc.21871] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Lin28 plays important roles in development, stem cell maintenance, oncogenesis and metabolism. As an RNA-binding protein, it blocks the biogenesis primarily of let-7 family miRNAs and also promotes translation of a cohort of mRNAs involved in cell growth, metabolism and pluripotency, likely through recognition of distinct sequence and structural motifs within mRNAs. Here, we show that one such motif, shared by multiple Lin28-responsive elements (LREs) present in Lin28 mRNA targets also participates in a Drosha-dependent regulation and may contribute to destabilization of its cognate mRNAs. We further show that the same mutations in the LREs known to abolish Lin28 binding and stimulation of translation also abrogate Drosha-dependent mRNA destabilization, and that this effect is independent of miRNAs, uncovering a previously unsuspected coupling between Drosha-dependent destabilization and Lin28-mediated regulation. Thus, Lin28-dependent stimulation of translation of target mRNAs may, in part, serve to compensate for their intrinsic instability, thereby ensuring optimal levels of expression of genes critical for cell viability, metabolism and pluripotency.
Collapse
Affiliation(s)
- Chong Qiao
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | |
Collapse
|
39
|
Li N, Zhong X, Lin X, Guo J, Zou L, Tanyi JL, Shao Z, Liang S, Wang LP, Hwang WT, Katsaros D, Montone K, Zhao X, Zhang L. Lin-28 homologue A (LIN28A) promotes cell cycle progression via regulation of cyclin-dependent kinase 2 (CDK2), cyclin D1 (CCND1), and cell division cycle 25 homolog A (CDC25A) expression in cancer. J Biol Chem 2012; 287:17386-17397. [PMID: 22467868 PMCID: PMC3366838 DOI: 10.1074/jbc.m111.321158] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2011] [Revised: 03/29/2012] [Indexed: 02/05/2023] Open
Abstract
The RNA-binding protein LIN28A regulates the translation and stability of a large number of mRNAs as well as the biogenesis of certain miRNAs in embryonic stem cells and developing tissues. Increasing evidence indicates that LIN28A functions as an oncogene promoting cancer cell growth. However, little is known about its molecular mechanism of cell cycle regulation in cancer. Using tissue microarrays, we found that strong LIN28A expression was reactivated in about 10% (7.1-17.1%) of epithelial tumors (six tumor types, n = 369). Both in vitro and in vivo experiments demonstrate that LIN28A promotes cell cycle progression in cancer cells. Genome-wide RNA-IP-chip experiments indicate that LIN28A binds to thousands of mRNAs, including a large group of cell cycle regulatory mRNAs in cancer and embryonic stem cells. Furthermore, the ability of LIN28A to stimulate translation of LIN28A-binding mRNAs, such as CDK2, was validated in vitro and in vivo. Finally, using a combined gene expression microarray and bioinformatics approach, we found that LIN28A also regulates CCND1 and CDC25A expression and that this is mediated by inhibiting the biogenesis of let-7 miRNA. Taken together, these results demonstrate that LIN28A is reactivated in about 10% of epithelial tumors and promotes cell cycle progression by regulation of both mRNA translation (let-7-independent) and miRNA biogenesis (let-7-dependent).
Collapse
Affiliation(s)
- Ning Li
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Xiaomin Zhong
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Xiaojuan Lin
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104; Department of Obstetrics and Gynecology, Second West China Hospital of Sichuan University, Chengdu, China
| | - Jinyi Guo
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Lian Zou
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104; Department of Obstetrics and Gynecology, Second West China Hospital of Sichuan University, Chengdu, China
| | - Janos L Tanyi
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Zhongjun Shao
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Shun Liang
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Li-Ping Wang
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Wei-Ting Hwang
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Dionyssios Katsaros
- Department of Obstetrics and Gynecology, University of Turin, Turin 10124, Italy
| | - Kathleen Montone
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104
| | - Xia Zhao
- Department of Obstetrics and Gynecology, Second West China Hospital of Sichuan University, Chengdu, China
| | - Lin Zhang
- Ovarian Cancer Research Center and Department of Obstetrics and Gynecology, University of Pennsylvania, Philadelphia, Pennsylvania 19104.
| |
Collapse
|
40
|
Mayr F, Schütz A, Döge N, Heinemann U. The Lin28 cold-shock domain remodels pre-let-7 microRNA. Nucleic Acids Res 2012; 40:7492-506. [PMID: 22570413 PMCID: PMC3424542 DOI: 10.1093/nar/gks355] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The RNA-binding protein Lin28 regulates the processing of a developmentally important group of microRNAs, the let-7 family. Lin28 blocks the biogenesis of let-7 in embryonic stem cells and thereby prevents differentiation. It was shown that both RNA-binding domains (RBDs) of this protein, the cold-shock domain (CSD) and the zinc-knuckle domain (ZKD) are indispensable for pri- or pre-let-7 binding and blocking its maturation. Here, we systematically examined the nucleic acid-binding preferences of the Lin28 RBDs and determined the crystal structure of the Lin28 CSD in the absence and presence of nucleic acids. Both RNA-binding domains bind to single-stranded nucleic acids with the ZKD mediating specific binding to a conserved GGAG motif and the CSD showing only limited sequence specificity. However, only the isolated Lin28 CSD, but not the ZKD, can bind with a reasonable affinity to pre-let-7 and thus is able to remodel the terminal loop of pre-let-7 including the Dicer cleavage site. Further mutagenesis studies reveal that the Lin28 CSD induces a conformational change in the terminal loop of pre-let-7 and thereby facilitates a subsequent specific binding of the Lin28 ZKD to the conserved GGAG motif.
Collapse
Affiliation(s)
- Florian Mayr
- Crystallography, Max-Delbrück Center for Molecular Medicine, Robert-Rössle Straße 10, 13125 Berlin, Germany
| | | | | | | |
Collapse
|
41
|
Huang Y. A mirror of two faces: Lin28 as a master regulator of both miRNA and mRNA. WILEY INTERDISCIPLINARY REVIEWS-RNA 2012; 3:483-94. [PMID: 22467269 DOI: 10.1002/wrna.1112] [Citation(s) in RCA: 72] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Lin28 is an evolutionarily conserved RNA-binding protein that plays important roles in development, pluripotency, tumorigenesis, and metabolism. Emerging evidence suggests that the pleiotropic roles of Lin28 in the diverse physiological and pathological processes are mechanistically linked to its ability to modulate not only the biogenesis of miRNAs, particularly the let-7 family miRNAs, but also the translation of mRNAs important for cell growth and metabolism. Let-7 negatively regulates the translation of oncogenes, cell cycle regulators, and metabolic pathway components. Lin28 relieves this repression by blocking the production of mature let-7. Lin28 binds to the terminal loops of let-7 precursors, leading to inhibition of processing and the induction of uridylation and precursor degradation. Lin28 also is a direct translational regulator: it selectively binds to a cohort of mRNAs and stimulates their translation. Recent advances in our understanding of Lin28-mediated mechanisms of posttranscriptional regulation of gene expression reveal important roles of this protein in the fields of development, stem cells, metabolic diseases, and cancer.
Collapse
Affiliation(s)
- Yingqun Huang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
42
|
Vadla B, Kemper K, Alaimo J, Heine C, Moss EG. lin-28 controls the succession of cell fate choices via two distinct activities. PLoS Genet 2012; 8:e1002588. [PMID: 22457637 PMCID: PMC3310729 DOI: 10.1371/journal.pgen.1002588] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Accepted: 01/25/2012] [Indexed: 12/29/2022] Open
Abstract
lin-28 is a conserved regulator of cell fate succession in animals. In Caenorhabditis elegans, it is a component of the heterochronic gene pathway that governs larval developmental timing, while its vertebrate homologs promote pluripotency and control differentiation in diverse tissues. The RNA binding protein encoded by lin-28 can directly inhibit let-7 microRNA processing by a novel mechanism that is conserved from worms to humans. We found that C. elegans LIN-28 protein can interact with four distinct let-7 family pre-microRNAs, but in vivo inhibits the premature accumulation of only let-7. Surprisingly, however, lin-28 does not require let-7 or its relatives for its characteristic promotion of second larval stage cell fates. In other words, we find that the premature accumulation of mature let-7 does not account for lin-28's precocious phenotype. To explain let-7's role in lin-28 activity, we provide evidence that lin-28 acts in two steps: first, the let-7-independent positive regulation of hbl-1 through its 3'UTR to control L2 stage-specific cell fates; and second, a let-7-dependent step that controls subsequent fates via repression of lin-41. Our evidence also indicates that let-7 functions one stage earlier in C. elegans development than previously thought. Importantly, lin-28's two-step mechanism resembles that of the heterochronic gene lin-14, and the overlap of their activities suggests a clockwork mechanism for developmental timing. Furthermore, this model explains the previous observation that mammalian Lin28 has two genetically separable activities. Thus, lin-28's two-step mechanism may be an essential feature of its evolutionarily conserved role in cell fate succession.
Collapse
Affiliation(s)
- Bhaskar Vadla
- Department of Molecular Biology, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
- Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
| | - Kevin Kemper
- Department of Molecular Biology, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
| | - Jennifer Alaimo
- Department of Molecular Biology, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
- Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
| | - Christian Heine
- Department of Molecular Biology, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
- Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
| | - Eric G. Moss
- Department of Molecular Biology, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
- Graduate School of Biomedical Sciences, University of Medicine and Dentistry of New Jersey, Stratford, New Jersey, United States of America
- * E-mail:
| |
Collapse
|
43
|
Childs AJ, Kinnell HL, He J, Anderson RA. LIN28 is selectively expressed by primordial and pre-meiotic germ cells in the human fetal ovary. Stem Cells Dev 2012; 21:2343-9. [PMID: 22296229 DOI: 10.1089/scd.2011.0730] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
Abstract
Germ cell development requires timely transition from primordial germ cell (PGC) self-renewal to meiotic differentiation. This is associated with widespread changes in gene expression, including downregulation of stem cell-associated genes, such as OCT4 and KIT, and upregulation of markers of germ cell differentiation and meiosis, such as VASA, STRA8, and SYCP3. The stem cell-expressed RNA-binding protein Lin28 has recently been demonstrated to be essential for PGC specification in mice, and LIN28 is expressed in human germ cell tumors with phenotypic similarities to human fetal germ cells. We have therefore examined the expression of LIN28 during normal germ cell development in the human fetal ovary, from the PGC stage, through meiosis to the initiation of follicle formation. LIN28 transcript levels were highest when the gonad contained only PGCs, and decreased significantly with increasing gestation, coincident with the onset of germ cell differentiation. Immunohistochemistry revealed LIN28 protein expression to be germ cell-specific at all stages examined. All PGCs expressed LIN28, but at later gestations expression was restricted to a subpopulation of germ cells, which we demonstrate to be primordial and premeiotic germ cells based on immunofluorescent colocalization of LIN28 and OCT4, and absence of overlap with the meiosis marker SYCP3. We also demonstrate the expression of the LIN28 target precursor pri-microRNA transcripts pri-LET7a/f/d and pri-LET-7g in the human fetal ovary, and that expression of these is highest at the PGC stage, mirroring that of LIN28. The spatial and temporal restriction of LIN28 expression and coincident peaks of expression of LIN28 and target pri-microRNAs suggest important roles for this protein in the maintenance of the germline stem cell state and the regulation of microRNA activity in the developing human ovary.
Collapse
Affiliation(s)
- Andrew J Childs
- MRC Centre for Reproductive Health, The Queen's Medical Research Institute, The University of Edinburgh, Edinburgh, United Kingdom.
| | | | | | | |
Collapse
|
44
|
MicroRNA expression profile in bovine cumulus–oocyte complexes: Possible role of let-7 and miR-106a in the development of bovine oocytes. Anim Reprod Sci 2012; 130:16-26. [DOI: 10.1016/j.anireprosci.2011.12.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2011] [Revised: 12/14/2011] [Accepted: 12/23/2011] [Indexed: 12/21/2022]
|
45
|
Kim CW, Vo MT, Kim HK, Lee HH, Yoon NA, Lee BJ, Min YJ, Joo WD, Cha HJ, Park JW, Cho WJ. Ectopic over-expression of tristetraprolin in human cancer cells promotes biogenesis of let-7 by down-regulation of Lin28. Nucleic Acids Res 2011; 40:3856-69. [PMID: 22210895 PMCID: PMC3351177 DOI: 10.1093/nar/gkr1302] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tristetraprolin (TTP) is a AU-rich element (ARE) binding protein and exhibits suppressive effects on cell growth through down-regulation of ARE-containing oncogenes. The let-7 microRNA has emerged as a significant factor in tumor suppression. Both TTP and let-7 are often repressed in human cancers, thereby promoting oncogenesis by derepressing their target genes. In this work, an unexpected link between TTP and let-7 has been found in human cancer cells. TTP promotes an increase in expression of mature let-7, which leads to the inhibition of let-7 target gene CDC34 expression and suppresses cell growth. This event is associated with TTP-mediated inhibition of Lin28, which has emerged as a negative modulator of let-7. Lin28 mRNA contains ARE within its 3′-UTR and TTP enhances the decay of Lin28 mRNA through binding to its 3′-UTR. This suggests that the TTP-mediated down-regulation of Lin28 plays a key role in let-7 miRNA biogenesis in cancer cells.
Collapse
Affiliation(s)
- Chae Won Kim
- Department of Biological Sciences, University of Ulsan, Ulsan 680-749, Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Lei XX, Xu J, Ma W, Qiao C, Newman MA, Hammond SM, Huang Y. Determinants of mRNA recognition and translation regulation by Lin28. Nucleic Acids Res 2011; 40:3574-84. [PMID: 22210884 PMCID: PMC3333870 DOI: 10.1093/nar/gkr1279] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Lin28 is critical for stem cell maintenance and is also associated with advanced human malignancies. Our recent genome-wide studies mark Lin28 as a master post-transcriptional regulator of a subset of messenger RNAs important for cell growth and metabolism. However, the molecular basis underpinning the selective mRNA target regulation is unclear. Here, we provide evidence that Lin28 recognizes a unique motif in multiple target mRNAs, characterized by a small but critical ‘A’ bulge flanked by two G:C base pairs embedded in a complex secondary structure. This motif mediates Lin28-dependent stimulation of translation. As Lin28 is also known to inhibit the biogenesis of a cohort of miRNAs including let-7, we propose that Lin28 binding to different RNA types (precursor miRNAs versus mRNAs) may facilitate recruitment of different co-factors, leading to distinct regulatory outcomes. Our findings uncover a putative yet unexpected motif that may constitute a mechanistic base for the multitude of functions regulated by Lin28 in both stem cells and cancer cells.
Collapse
Affiliation(s)
- Xin-Xiang Lei
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT 06510, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Structural basis of pre-let-7 miRNA recognition by the zinc knuckles of pluripotency factor Lin28. Nat Struct Mol Biol 2011; 19:84-9. [PMID: 22157959 DOI: 10.1038/nsmb.2202] [Citation(s) in RCA: 106] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2011] [Accepted: 11/10/2011] [Indexed: 12/11/2022]
Abstract
Lin28 inhibits the biogenesis of let-7 miRNAs through a direct interaction with the terminal loop of pre-let-7. This interaction requires the zinc-knuckle domains of Lin28. We show that the zinc knuckle domains of Lin28 are sufficient to provide binding selectivity for pre-let-7 miRNAs and present the NMR structure of human Lin28 zinc knuckles bound to the short sequence 5'-AGGAGAU-3'. The structure reveals that each zinc knuckle recognizes an AG dinucleotide separated by a single nucleotide spacer. This defines a new 5'-NGNNG-3' consensus motif that explains how Lin28 selectively recognizes pre-let-7 family members. Binding assays in cell lysates and functional assays in cultured cells demonstrate that the interactions observed in the solution structure also occur between the full-length protein and members of the pre-let-7 family. The consensus sequence explains several seemingly disparate previously published observations on the binding properties of Lin28.
Collapse
|
48
|
Zhu H, Shyh-Chang N, Segrè AV, Shinoda G, Shah SP, Einhorn WS, Takeuchi A, Engreitz JM, Hagan JP, Kharas MG, Urbach A, Thornton JE, Triboulet R, Gregory RI, Altshuler D, Daley GQ. The Lin28/let-7 axis regulates glucose metabolism. Cell 2011; 147:81-94. [PMID: 21962509 DOI: 10.1016/j.cell.2011.08.033] [Citation(s) in RCA: 732] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 05/09/2011] [Accepted: 08/05/2011] [Indexed: 12/15/2022]
Abstract
The let-7 tumor suppressor microRNAs are known for their regulation of oncogenes, while the RNA-binding proteins Lin28a/b promote malignancy by inhibiting let-7 biogenesis. We have uncovered unexpected roles for the Lin28/let-7 pathway in regulating metabolism. When overexpressed in mice, both Lin28a and LIN28B promote an insulin-sensitized state that resists high-fat-diet induced diabetes. Conversely, muscle-specific loss of Lin28a or overexpression of let-7 results in insulin resistance and impaired glucose tolerance. These phenomena occur, in part, through the let-7-mediated repression of multiple components of the insulin-PI3K-mTOR pathway, including IGF1R, INSR, and IRS2. In addition, the mTOR inhibitor, rapamycin, abrogates Lin28a-mediated insulin sensitivity and enhanced glucose uptake. Moreover, let-7 targets are enriched for genes containing SNPs associated with type 2 diabetes and control of fasting glucose in human genome-wide association studies. These data establish the Lin28/let-7 pathway as a central regulator of mammalian glucose metabolism.
Collapse
Affiliation(s)
- Hao Zhu
- Stem Cell Transplantation Program, Division of Pediatric Hematology/Oncology, Children's Hospital Boston and Dana Farber Cancer Institute, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Desjardins A, Yang A, Bouvette J, Omichinski JG, Legault P. Importance of the NCp7-like domain in the recognition of pre-let-7g by the pluripotency factor Lin28. Nucleic Acids Res 2011; 40:1767-77. [PMID: 22013165 PMCID: PMC3287177 DOI: 10.1093/nar/gkr808] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The pluripotency factor Lin28 is a highly conserved protein comprising a unique combination of RNA-binding motifs, an N-terminal cold-shock domain and a C-terminal region containing two retroviral-type CCHC zinc-binding domains. An important function of Lin28 is to inhibit the biogenesis of the let-7 family of microRNAs through a direct interaction with let-7 precursors. Here, we systematically characterize the determinants of the interaction between Lin28 and pre-let-7g by investigating the effect of protein and RNA mutations on in vitro binding. We determine that Lin28 binds with high affinity to the extended loop of pre-let-7g and that its C-terminal domain contributes predominantly to the affinity of this interaction. We uncover remarkable similarities between this C-terminal domain and the NCp7 protein of HIV-1, not only in terms of primary structure but also in their modes of RNA binding. This NCp7-like domain of Lin28 recognizes a G-rich bulge within pre-let-7g, which is adjacent to one of the Dicer cleavage sites. We hypothesize that the NCp7-like domain initiates RNA binding and partially unfolds the RNA. This partial unfolding would then enable multiple copies of Lin28 to bind the extended loop of pre-let-7g and protect the RNA from cleavage by the pre-microRNA processing enzyme Dicer.
Collapse
Affiliation(s)
- Alexandre Desjardins
- Département de Biochimie, Université de Montréal, CP 6128, Succursale Centre-Ville, Montréal, QC, Canada H3C 3J7
| | | | | | | | | |
Collapse
|
50
|
Peng S, Chen LL, Lei XX, Yang L, Lin H, Carmichael GG, Huang Y. Genome-wide studies reveal that Lin28 enhances the translation of genes important for growth and survival of human embryonic stem cells. Stem Cells 2011; 29:496-504. [PMID: 21425412 DOI: 10.1002/stem.591] [Citation(s) in RCA: 158] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Lin28 inhibits the expression of let-7 microRNAs but also exhibits let-7-independent functions. Using immunoprecipitation and deep sequencing, we show here that Lin28 preferentially associates with a small subset of cellular mRNAs. Of particular interest are those for ribosomal proteins and metabolic enzymes, the expression levels of which are known to be coupled to cell growth and survival. Polysome profiling and reporter analyses suggest that Lin28 stimulates the translation of many or most of these targets. Moreover, Lin28-responsive elements were found within the coding regions of all target genes tested. Finally, a mutant Lin28 that still binds RNA but fails to interact with RNA helicase A (RHA), acts as a dominant-negative inhibitor of Lin28-dependent stimulation of translation. We suggest that Lin28, working in concert with RHA, enhances the translation of genes important for the growth and survival of human embryonic stem cells.
Collapse
Affiliation(s)
- Shuping Peng
- Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | | | | | | |
Collapse
|