1
|
Ogawa A, Izumikawa K, Tate S, Isoyama S, Mori M, Fujiwara K, Watanabe S, Ohga T, Jo U, Taniyama D, Kitajima S, Tanaka S, Onji H, Kageyama SI, Yamamoto G, Saito H, Morita TY, Okada M, Natsumeda M, Nagahama M, Kobayashi J, Ohashi A, Sasanuma H, Higashiyama S, Dan S, Pommier Y, Murai J. SLFN11-mediated ribosome biogenesis impairment induces TP53-independent apoptosis. Mol Cell 2025; 85:894-912.e10. [PMID: 39909041 PMCID: PMC11890970 DOI: 10.1016/j.molcel.2025.01.008] [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: 05/14/2024] [Revised: 12/11/2024] [Accepted: 01/10/2025] [Indexed: 02/07/2025]
Abstract
Impairment of ribosome biogenesis (RiBi) triggered by inhibition of ribosomal RNA (rRNA) synthesis and processing leads to various biological effects. We report that Schlafen 11 (SLFN11) induces TP53-independent apoptosis through RiBi impairment. Upon replication stress, SLFN11 inhibits rRNA synthesis with RNA polymerase I accumulation and increased chromatin accessibility in the ribosomal DNA (rDNA) genes. SLFN11-dependent RiBi impairment preferentially depletes short-lived proteins, particularly MCL1, leading to apoptosis in response to replication stress. SLFN11's Walker B motif (E669), DNA-binding site (K652), dephosphorylation site for single-strand DNA binding (S753), and RNase sites (E209/E214) are all required for the SLFN11-mediated RiBi impairment. Comparable effects were obtained with direct RNA polymerase I inhibitors and other RiBi inhibitory conditions regardless of SLFN11. These findings were extended across 34 diverse human cancer cell lines. Thus, we demonstrate that RiBi impairment is a robust inactivator of MCL1 and an additional proapoptotic mechanism by which SLFN11 sensitizes cancer cells to chemotherapeutic agents.
Collapse
Affiliation(s)
- Akane Ogawa
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Keiichi Izumikawa
- Laboratory of Molecular and Cellular Biochemistry, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Sota Tate
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Sho Isoyama
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Masaru Mori
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Kohei Fujiwara
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Soyoka Watanabe
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Takayuki Ohga
- Laboratory of Molecular and Cellular Biochemistry, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Ukhyun Jo
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Daiki Taniyama
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814, USA
| | - Shojiro Kitajima
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Soichiro Tanaka
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan
| | - Hiroshi Onji
- Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan
| | - Shun-Ichiro Kageyama
- Division of Radiation Oncology and Particle Therapy, National Cancer Center Hospital East, Chiba 277-8577, Japan
| | - Gaku Yamamoto
- Division of Collaborative Research and Development, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Hitoshi Saito
- Division of Collaborative Research and Development, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Tomoko Yamamori Morita
- Division of Collaborative Research and Development, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Masayasu Okada
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; Department of Brain Tumor Biology, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Manabu Natsumeda
- Department of Neurosurgery, Brain Research Institute, Niigata University, Niigata 951-8585, Japan; Advanced Treatment of Neurological Diseases Branch, Brain Research Institute, Niigata University, Niigata 951-8585, Japan
| | - Masami Nagahama
- Laboratory of Molecular and Cellular Biochemistry, Meiji Pharmaceutical University, Tokyo 204-8588, Japan
| | - Junya Kobayashi
- Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan; Department of Radiological Sciences, School of Health Sciences at Narita, International University of Health and Welfare, Narita, Tokyo 286-0048, Japan
| | - Akihiro Ohashi
- Division of Collaborative Research and Development, Exploratory Oncology Research & Clinical Trial Center, National Cancer Center, Chiba 277-8577, Japan
| | - Hiroyuki Sasanuma
- Department of Genome Medicine, Tokyo Metropolitan Institute of Medical Science, Tokyo 156-0057, Japan
| | - Shigeki Higashiyama
- Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan; Department of Oncogenesis and Tumor Regulation, Osaka International Cancer Institute, Osaka 103-0027, Japan
| | - Shingo Dan
- Division of Molecular Pharmacology, Cancer Chemotherapy Center, Japanese Foundation for Cancer Research, Tokyo 135-8550, Japan
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20814, USA.
| | - Junko Murai
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata 997-0017, Japan; Division of Cell Growth and Tumor Regulation, Proteo-Science Center, Toon, Ehime 791-0295, Japan; Department of Biochemistry and Molecular Genetics, Ehime University Graduate School of Medicine, Toon, Ehime 791-0295, Japan; Radiation Biology Center, Graduate School of Biostudies, Kyoto University, Kyoto 606-8501, Japan.
| |
Collapse
|
2
|
Challa S, Nandu T, Kim HB, Gong X, Renshaw CW, Li WC, Tan X, Aljardali MW, Camacho CV, Chen J, Kraus WL. RACK1 MARylation regulates translation and stress granules in ovarian cancer cells. J Cell Biol 2025; 224:e202401101. [PMID: 39760726 DOI: 10.1083/jcb.202401101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 09/09/2024] [Accepted: 11/11/2024] [Indexed: 01/07/2025] Open
Abstract
Mono(ADP-ribosyl)ation (MARylation) is emerging as a critical regulator of ribosome function and translation. Herein, we demonstrate that RACK1, an integral component of the ribosome, is MARylated by the mono(ADP-ribosyl) transferase (MART) PARP14 in ovarian cancer cells. MARylation of RACK1 is required for stress granule formation and promotes the colocalization of RACK1 in stress granules with G3BP1, eIF3η, and 40S ribosomal proteins. In parallel, we observed reduced translation of a subset of mRNAs, including those encoding key cancer regulators (e.g., AKT). Treatment with a PARP14 inhibitor or mutation of the sites of MARylation on RACK1 blocks these outcomes, as well as the growth of ovarian cancer cells in culture and in vivo. To reset the system after prolonged stress and recovery, the ADP-ribosyl hydrolase TARG1 deMARylates RACK1, leading to the dissociation of the stress granules and the restoration of translation. Collectively, our results demonstrate a therapeutically targetable pathway that controls polysome assembly, translation, and stress granule dynamics in ovarian cancer cells.
Collapse
Affiliation(s)
- Sridevi Challa
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tulip Nandu
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hyung Bum Kim
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Graduate Program in Genetics, Development, and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xuan Gong
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charles W Renshaw
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Wan-Chen Li
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xinrui Tan
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Marwa W Aljardali
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Cristel V Camacho
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jin Chen
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - W Lee Kraus
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Graduate Program in Genetics, Development, and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Section of Laboratory Research, Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
3
|
Challa S, Nandu T, Kim HB, Gong X, Renshaw CW, Li WC, Tan X, Aljardali MW, Camacho CV, Chen J, Kraus WL. A PARP14/TARG1-Regulated RACK1 MARylation Cycle Drives Stress Granule Dynamics in Ovarian Cancer Cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.10.13.562273. [PMID: 37873085 PMCID: PMC10592810 DOI: 10.1101/2023.10.13.562273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Mono(ADP-ribosyl)ation (MARylation) is emerging as a critical regulator of ribosome function and translation. Herein, we demonstrate that RACK1, an integral component of the ribosome, is MARylated on three acidic residues by the mono(ADP-ribosyl) transferase (MART) PARP14 in ovarian cancer cells. MARylation of RACK1 is required for stress granule formation and promotes the colocalization of RACK1 in stress granules with G3BP1, eIF3η, and 40S ribosomal proteins. In parallel, we observed reduced translation of a subset of mRNAs, including those encoding key cancer regulators (e.g., AKT). Treatment with a PARP14 inhibitor or mutation of the sites of MARylation on RACK1 blocks these outcomes, as well as the growth of ovarian cancer cells in culture and in vivo. To re-set the system after prolonged stress and recovery, the ADP-ribosyl hydrolase TARG1 deMARylates RACK1, leading to the dissociation of the stress granules and the restoration of translation. Collectively, our results demonstrate a therapeutically targetable pathway that controls stress granule assembly and disassembly in ovarian cancer cells.
Collapse
Affiliation(s)
- Sridevi Challa
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Current address: Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637
| | - Tulip Nandu
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hyung Bum Kim
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Graduate Program in Genetics, Development, and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xuan Gong
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Current address: Department of Bone Marrow Transplantation and Cellular Therapy, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Charles W. Renshaw
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Wan-Chen Li
- Altos Labs, Bay Area Institute of Science, Redwood City, CA 94403
| | - Xinrui Tan
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Marwa W. Aljardali
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cristel V. Camacho
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jin Chen
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Altos Labs, Bay Area Institute of Science, Redwood City, CA 94403
| | - W. Lee Kraus
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Graduate Program in Genetics, Development, and Disease, Graduate School of Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
4
|
Yang Y, Li Y, Sears RC, Sun XX, Dai MS. SUMOylation regulation of ribosome biogenesis: Emerging roles for USP36. FRONTIERS IN RNA RESEARCH 2024; 2:1389104. [PMID: 38764604 PMCID: PMC11101209 DOI: 10.3389/frnar.2024.1389104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2024]
Abstract
Ribosome biogenesis is essential for cell growth, proliferation, and animal development. Its deregulation leads to various human disorders such as ribosomopathies and cancer. Thus, tight regulation of ribosome biogenesis is crucial for normal cell homeostasis. Emerging evidence suggests that posttranslational modifications such as ubiquitination and SUMOylation play a crucial role in regulating ribosome biogenesis. Our recent studies reveal that USP36, a nucleolar deubiquitinating enzyme (DUB), acts also as a SUMO ligase to regulate nucleolar protein group SUMOylation, thereby being essential for ribosome biogenesis. Here, we provide an overview of the current understanding of the SUMOylation regulation of ribosome biogenesis and discuss the role of USP36 in nucleolar SUMOylation.
Collapse
Affiliation(s)
- Yunhan Yang
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Yanping Li
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Rosalie C. Sears
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Xiao-Xin Sun
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| | - Mu-Shui Dai
- Department of Molecular & Medical Genetics, Oregon Health & Science University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA
| |
Collapse
|
5
|
Goutam J, Sharma G, Yadav V, Pathak G, Kharwar RN, Sharma D. A Focused Review of the Pharmacological Potentials of Terrein as an Anticancer Agent. Nat Prod Commun 2023; 18. [DOI: 10.1177/1934578x231174128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025] Open
Abstract
Terrein is one of the most important biomolecules of fungal origin being studied from a medicinal perspective. Secondary metabolites are the intermediate products produced during the metabolism of organisms for a large number of functions, for example, defense and communication signals. From the outset, terrein has largely been studied as an anticancer secondary biomolecule. Aspergillus terreus is the only fungal source of some valuable drugs and mycotoxins. From the beginning, a few species of Aspergillus were known to be viable chemical factories. Terrein is a potent biological molecule present in the fungus that is responsible for its medicinal and agricultural values. Numerous evaluations conducted on terrein showed it to have marked biological activities (antimicrobial, antiproliferative, anti-oxidative, and others). To date, terrein has emerged as a very attractive therapeutic regimen against cancer due to its dual targeting nature; tumor angiogenesis and cell proliferation. This focused review provides details of the therapeutic value of terrein and its modes of action as an anticancer agent. Besides this, terrein has other marked bioactivities and manifold uses in the field of medicine, which have also been discussed here.
Collapse
Affiliation(s)
- Jyoti Goutam
- Mycopathology and Microbial Technology Laboratory, Centre of Advance Study in Botany, Banaras Hindu University, Varanasi, India
| | - Gunjan Sharma
- Immunology and Cancer Research, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Vandana Yadav
- Central Animal House Facility, ICMR-National Institute of Pathology, New Delhi, India
| | - Gauri Pathak
- Department of Microbiology, Maulana Azad Medical College, New Delhi, India
| | - Ravindra Nath Kharwar
- Mycopathology and Microbial Technology Laboratory, Centre of Advance Study in Botany, Banaras Hindu University, Varanasi, India
| | - Divakar Sharma
- Department of Microbiology, Lady Hardinge Medical College, New Delhi, India
| |
Collapse
|
6
|
Matsuzaki Y, Naito Y, Miura N, Mori T, Watabe Y, Yoshimoto S, Shibahara T, Takano M, Honda K. RIOK2 Contributes to Cell Growth and Protein Synthesis in Human Oral Squamous Cell Carcinoma. Curr Oncol 2022; 30:381-391. [PMID: 36661680 PMCID: PMC9857684 DOI: 10.3390/curroncol30010031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Ribosomes are responsible for the protein synthesis that maintains cellular homeostasis and is required for the rapid cellular division of cancer cells. However, the role of ribosome biogenesis mediators in the malignant behavior of tongue squamous cell carcinoma (TSCC) is unknown. In this study, we found that the expression of RIOK2, a key enzyme involved in the maturation steps of the pre-40S ribosomal complex, was significantly associated with poorer overall survival in patients with TSCC. Further, multivariate analysis revealed that RIOK2 is an independent prognostic factor (hazard ratio, 3.53; 95% confidence interval, 1.19-10.91). Inhibition of RIOK2 expression by siRNA decreased cell growth and S6 ribosomal protein expression in oral squamous cell carcinoma cell lines. RIOK2 knockdown also led to a significant decrease in the protein synthesis in cancer cells. RIOK2 has potential application as a novel therapeutic target for TSCC treatment.
Collapse
Affiliation(s)
- Yusuke Matsuzaki
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo 113-8602, Japan
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Yutaka Naito
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo 113-8602, Japan
| | - Nami Miura
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo 113-8602, Japan
| | - Taisuke Mori
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Yukio Watabe
- Department of Dentistry and Oral Surgery, Tokyo Metropolitan Tama Medical Center, Tokyo 183-8524, Japan
| | - Seiichi Yoshimoto
- Department of Head and Neck Surgery, National Cancer Center Hospital, Tokyo 104-0045, Japan
| | - Takahiko Shibahara
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Masayuki Takano
- Department of Oral and Maxillofacial Surgery, Tokyo Dental College, Tokyo 101-0061, Japan
| | - Kazufumi Honda
- Department of Bioregulation, Institute for Advanced Medical Sciences, Nippon Medical School, Tokyo 113-8602, Japan
- Department of Bioregulation, Graduate School of Medicine, Nippon Medical School, Tokyo 113-8602, Japan
| |
Collapse
|
7
|
Targeting Ribosome Biogenesis in Cancer: Lessons Learned and Way Forward. Cancers (Basel) 2022; 14:cancers14092126. [PMID: 35565259 PMCID: PMC9100539 DOI: 10.3390/cancers14092126] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 01/05/2023] Open
Abstract
Simple Summary Cells need to produce ribosomes to sustain continuous proliferation and expand in numbers, a feature that is even more prominent in uncontrollably proliferating cancer cells. Certain cancer cell types are expected to depend more on ribosome biogenesis based on their genetic background, and this potential vulnerability can be exploited in designing effective, targeted cancer therapies. This review provides information on anti-cancer molecules that target the ribosome biogenesis machinery and indicates avenues for future research. Abstract Rapid growth and unrestrained proliferation is a hallmark of many cancers. To accomplish this, cancer cells re-wire and increase their biosynthetic and metabolic activities, including ribosome biogenesis (RiBi), a complex, highly energy-consuming process. Several chemotherapeutic agents used in the clinic impair this process by interfering with the transcription of ribosomal RNA (rRNA) in the nucleolus through the blockade of RNA polymerase I or by limiting the nucleotide building blocks of RNA, thereby ultimately preventing the synthesis of new ribosomes. Perturbations in RiBi activate nucleolar stress response pathways, including those controlled by p53. While compounds such as actinomycin D and oxaliplatin effectively disrupt RiBi, there is an ongoing effort to improve the specificity further and find new potent RiBi-targeting compounds with improved pharmacological characteristics. A few recently identified inhibitors have also become popular as research tools, facilitating our advances in understanding RiBi. Here we provide a comprehensive overview of the various compounds targeting RiBi, their mechanism of action, and potential use in cancer therapy. We discuss screening strategies, drug repurposing, and common problems with compound specificity and mechanisms of action. Finally, emerging paths to discovery and avenues for the development of potential biomarkers predictive of therapeutic outcomes across cancer subtypes are also presented.
Collapse
|
8
|
Ding GB, Zhu C, Wang Q, Cao H, Li BC, Yang P, Stauber RH, Nie G, Li Z. Molecularly engineered tumor acidity-responsive plant toxin gelonin for safe and efficient cancer therapy. Bioact Mater 2022; 18:42-55. [PMID: 35387163 PMCID: PMC8961304 DOI: 10.1016/j.bioactmat.2022.02.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 01/17/2022] [Accepted: 02/07/2022] [Indexed: 12/11/2022] Open
Abstract
Due to the unsatisfactory therapeutic efficacy and inexorable side effects of small molecule antineoplastic agents, extensive efforts have been devoted to the development of more potent macromolecular agents with high specificity. Gelonin is a plant-derived protein toxin that exhibits robust antitumor effect via inactivating ribosomes and inhibiting protein synthesis. Nonetheless, its poor internalization ability to tumor cells has compromised the therapeutic promise of gelonin. In this study, a tumor acidity-responsive intracellular protein delivery system ─ functional gelonin (Trx-pHLIP-Gelonin, TpG) composed of a thioredoxin (Trx) tag, a pH low insertion peptide (pHLIP) and gelonin, was designed and obtained by genetic recombination technique for the first time. TpG could effectively enter into tumor cells under weakly acidic conditions and markedly suppress tumor cell proliferation via triggering cell apoptosis and inhibiting protein synthesis. Most importantly, treatment by intravenous injection into subcutaneous SKOV3 solid tumors in a mouse model showed that TpG was much more effective than gelonin in curtailing tumor growth rates with negligible toxicity. Collectively, our present work suggests that the tumor acidity-targeted delivery manner endowed by pHLIP offers a new avenue for efficient delivery of other bioactive substances to acidic diseased tissues. A pH-responsive gelonin delivery platform — TpG was molecularly engineered. TpG exhibited good thermal stability and excellent serum stability. TpG enabled an efficient intracellular translocation of gelonin at pH 6.5. TpG exerted pronounced anti-proliferative effect via inducing massive apoptosis. TpG significantly delayed tumor growth with favorable in vivo biosafety profile.
Collapse
|
9
|
9-Aminoacridine Inhibits Ribosome Biogenesis by Targeting Both Transcription and Processing of Ribosomal RNA. Int J Mol Sci 2022; 23:ijms23031260. [PMID: 35163183 PMCID: PMC8836032 DOI: 10.3390/ijms23031260] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/13/2022] Open
Abstract
Aminoacridines, used for decades as antiseptic and antiparasitic agents, are prospective candidates for therapeutic repurposing and new drug development. Although the mechanisms behind their biological effects are not fully elucidated, they are most often attributed to the acridines’ ability to intercalate into DNA. Here, we characterized the effects of 9-aminoacridine (9AA) on pre-rRNA metabolism in cultured mammalian cells. Our results demonstrate that 9AA inhibits both transcription of the ribosomal RNA precursors (pre-rRNA) and processing of the already synthesized pre-rRNAs, thereby rapidly abolishing ribosome biogenesis. Using a fluorescent intercalator displacement assay, we further show that 9AA can bind to RNA in vitro, which likely contributes to its ability to inhibit post-transcriptional steps in pre-rRNA maturation. These findings extend the arsenal of small-molecule compounds that can be used to block ribosome biogenesis in mammalian cells and have implications for the pharmacological development of new ribosome biogenesis inhibitors.
Collapse
|
10
|
Messling JE, Agger K, Andersen KL, Kromer K, Kuepper HM, Lund AH, Helin K. Targeting RIOK2 ATPase activity leads to decreased protein synthesis and cell death in acute myeloid leukemia. Blood 2022; 139:245-255. [PMID: 34359076 PMCID: PMC8759535 DOI: 10.1182/blood.2021012629] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 07/29/2021] [Indexed: 01/16/2023] Open
Abstract
Novel therapies for the treatment of acute myeloid leukemia (AML) are urgently needed, because current treatments do not cure most patients with AML. We report a domain-focused, kinome-wide CRISPR-Cas9 screening that identified protein kinase targets for the treatment of AML, which led to the identification of Rio-kinase 2 (RIOK2) as a potential novel target. Loss of RIOK2 led to a decrease in protein synthesis and to ribosomal instability followed by apoptosis in leukemic cells, but not in fibroblasts. Moreover, the ATPase function of RIOK2 was necessary for cell survival. When a small-molecule inhibitor was used, pharmacological inhibition of RIOK2 similarly led to loss of protein synthesis and apoptosis and affected leukemic cell growth in vivo. Our results provide proof of concept for targeting RIOK2 as a potential treatment of patients with AML.
Collapse
Affiliation(s)
- Jan-Erik Messling
- Biotech Research and Innovation Centre and
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; and
| | - Karl Agger
- Biotech Research and Innovation Centre and
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; and
| | | | - Kristina Kromer
- Biotech Research and Innovation Centre and
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; and
| | - Hanna M Kuepper
- Biotech Research and Innovation Centre and
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; and
| | | | - Kristian Helin
- Biotech Research and Innovation Centre and
- The Novo Nordisk Foundation Center for Stem Cell Biology (DanStem), University of Copenhagen, Copenhagen, Denmark; and
- Cell Biology Program and
- Center for Epigenetics Research, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
11
|
Glioma Cells Acquire Stem-like Characters by Extrinsic Ribosome Stimuli. Cells 2021; 10:cells10112970. [PMID: 34831193 PMCID: PMC8616507 DOI: 10.3390/cells10112970] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 10/21/2021] [Accepted: 10/27/2021] [Indexed: 12/17/2022] Open
Abstract
Although glioblastoma (GBM) stem-like cells (GSCs), which retain chemo-radio resistance and recurrence, are key prognostic factors in GBM patients, the molecular mechanisms of GSC development are largely unknown. Recently, several studies revealed that extrinsic ribosome incorporation into somatic cells resulted in stem cell properties and served as a key trigger and factor for the cell reprogramming process. In this study, we aimed to investigate the mechanisms underlying GSCs development by focusing on extrinsic ribosome incorporation into GBM cells. Ribosome-induced cancer cell spheroid (RICCS) formation was significantly upregulated by ribosome incorporation. RICCS showed the stem-like cell characters (number of cell spheroid, stem cell markers, and ability for trans differentiation towards adipocytes and osteocytes). In RICCS, the phosphorylation and protein expression of ribosomal protein S6 (RPS6), an intrinsic ribosomal protein, and STAT3 phosphorylation were upregulated, and involved in the regulation of cell spheroid formation. Consistent with those results, glioma-derived extrinsic ribosome also promoted GBM-RICCS formation through intrinsic RPS6 phosphorylation. Moreover, in glioma patients, RPS6 phosphorylation was dominantly observed in high-grade glioma tissues, and predominantly upregulated in GSCs niches, such as the perinecrosis niche and perivascular niche. Those results indicate the potential biological and clinical significance of extrinsic ribosomal proteins in GSC development.
Collapse
|
12
|
Kasim M, Gencturk E, Ulgen KO. Real-Time Single-Cell Monitoring of Drug Effects Using Droplet-Based Microfluidic Technology: A Proof-of-Concept Study. OMICS-A JOURNAL OF INTEGRATIVE BIOLOGY 2021; 25:641-651. [PMID: 34582730 DOI: 10.1089/omi.2021.0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Drugs that act on ribosome biogenesis and cell proliferation play important roles in treatment of human diseases. Moreover, measurement of drug effects at a single-cell level would create vast opportunities for pharmaceutical innovation. We present in this study an original proof-of-concept study of single-cell measurement of drug effects with a focus on inhibition of ribosome biogenesis and cell proliferation, and using yeast (Saccharomyces cerevisiae) as a model eukaryotic organism. We employed a droplet-based microfluidic technology and nucleolar protein-tagged strain of the yeast for real-time monitoring of the cells. We report a comprehensive account of the ways in which interrelated pathways are impacted by drug treatment in a single-cell level. Self-organizing maps, transcription factor, and Gene Ontology enrichment analyses were utilized to these ends. This article makes a contribution to advance single-cell measurement of drug effects. We anticipate the microfluidic technology platform presented herein is well poised for future applications in personalized/precision medicine research as well as in industrial settings for drug discovery and clinical development. In addition, the study offers new insights on ribosome biogenesis and cell proliferation that should prove useful in cancer research and other complex human diseases impacted by these key cellular processes.
Collapse
Affiliation(s)
- Muge Kasim
- Department of Chemical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Elif Gencturk
- Department of Chemical Engineering, Boğaziçi University, Istanbul, Turkey
| | - Kutlu O Ulgen
- Department of Chemical Engineering, Boğaziçi University, Istanbul, Turkey
| |
Collapse
|
13
|
Singh A, Schermann G, Reislöhner S, Kellner N, Hurt E, Brunner M. Global Transcriptome Characterization and Assembly of the Thermophilic Ascomycete Chaetomium thermophilum. Genes (Basel) 2021; 12:1549. [PMID: 34680944 PMCID: PMC8535861 DOI: 10.3390/genes12101549] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 09/24/2021] [Accepted: 09/26/2021] [Indexed: 12/30/2022] Open
Abstract
A correct genome annotation is fundamental for research in the field of molecular and structural biology. The annotation of the reference genome of Chaetomium thermophilum has been reported previously, but it is essentially limited to open reading frames (ORFs) of protein coding genes and contains only a few noncoding transcripts. In this study, we identified and annotated full-length transcripts of C. thermophilum by deep RNA sequencing. We annotated 7044 coding genes and 4567 noncoding genes. Astonishingly, 23% of the coding genes are alternatively spliced. We identified 679 novel coding genes as well as 2878 novel noncoding genes and corrected the structural organization of more than 50% of the previously annotated genes. Furthermore, we substantially extended the Gene Ontology (GO) and Enzyme Commission (EC) lists, which provide comprehensive search tools for potential industrial applications and basic research. The identified novel transcripts and improved annotation will help to understand the gene regulatory landscape in C. thermophilum. The analysis pipeline developed here can be used to build transcriptome assemblies and identify coding and noncoding RNAs of other species.
Collapse
Affiliation(s)
| | | | | | | | | | - Michael Brunner
- Heidelberg University Biochemistry Center (BZH), Im Neuenheimer Feld 328, D-69120 Heidelberg, Germany; (A.S.); (G.S.); (S.R.); (N.K.); (E.H.)
| |
Collapse
|
14
|
Challa S, Khulpateea BR, Nandu T, Camacho CV, Ryu KW, Chen H, Peng Y, Lea JS, Kraus WL. Ribosome ADP-ribosylation inhibits translation and maintains proteostasis in cancers. Cell 2021; 184:4531-4546.e26. [PMID: 34314702 PMCID: PMC8380725 DOI: 10.1016/j.cell.2021.07.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 05/11/2021] [Accepted: 07/02/2021] [Indexed: 10/20/2022]
Abstract
Defects in translation lead to changes in the expression of proteins that can serve as drivers of cancer formation. Here, we show that cytosolic NAD+ synthesis plays an essential role in ovarian cancer by regulating translation and maintaining protein homeostasis. Expression of NMNAT-2, a cytosolic NAD+ synthase, is highly upregulated in ovarian cancers. NMNAT-2 supports the catalytic activity of the mono(ADP-ribosyl) transferase (MART) PARP-16, which mono(ADP-ribosyl)ates (MARylates) ribosomal proteins. Depletion of NMNAT-2 or PARP-16 leads to inhibition of MARylation, increased polysome association and enhanced translation of specific mRNAs, aggregation of their translated protein products, and reduced growth of ovarian cancer cells. Furthermore, MARylation of the ribosomal proteins, such as RPL24 and RPS6, inhibits polysome assembly by stabilizing eIF6 binding to ribosomes. Collectively, our results demonstrate that ribosome MARylation promotes protein homeostasis in cancers by fine-tuning the levels of protein synthesis and preventing toxic protein aggregation.
Collapse
Affiliation(s)
- Sridevi Challa
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Beman R Khulpateea
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Tulip Nandu
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Cristel V Camacho
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Keun W Ryu
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Hao Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yan Peng
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jayanthi S Lea
- Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - W Lee Kraus
- Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
15
|
Kiss A, Csikos C, Regdon Z, Polgár Z, Virág L, Hegedűs C. NMNAT1 Is a Survival Factor in Actinomycin D-Induced Osteosarcoma Cell Death. Int J Mol Sci 2021; 22:8869. [PMID: 34445574 PMCID: PMC8396190 DOI: 10.3390/ijms22168869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 08/12/2021] [Accepted: 08/16/2021] [Indexed: 01/25/2023] Open
Abstract
Osteosarcoma is a frequent and extremely aggressive type of pediatric cancer. New therapeutic approaches are needed to improve the overall survival of osteosarcoma patients. Our previous results suggest that NMNAT1, a key enzyme in nuclear NAD+ synthesis, facilitates the survival of cisplatin-treated osteosarcoma cells. A high-throughput cytotoxicity screening was performed to identify novel pathways or compounds linked to the cancer-promoting role of NMNAT1. Nine compounds caused higher toxicity in the NMNAT1 KO U2OS cells compared to their wild type counterparts, and actinomycin D (ActD) was the most potent. ActD-treatment of NMNAT1 KO cells increased caspase activity and secondary necrosis. The reduced NAD+ content in NMNAT1 KO cells was further decreased by ActD, which partially inhibited NAD+-dependent enzymes, including the DNA nick sensor enzyme PARP1 and the NAD+-dependent deacetylase SIRT1. Impaired PARP1 activity increased DNA damage in ActD-treated NMNAT1 knockout cells, while SIRT1 impairment increased acetylation of the p53 protein, causing the upregulation of pro-apoptotic proteins (NOXA, BAX). Proliferation was decreased through both PARP- and SIRT-dependent pathways. On the one hand, PARP inhibitors sensitized wild type but not NMNAT1 KO cells to ActD-induced anti-clonogenic effects; on the other hand, over-acetylated p53 induced the expression of the anti-proliferative p21 protein leading to cell cycle arrest. Based on our results, NMNAT1 acts as a survival factor in ActD-treated osteosarcoma cells. By inhibiting both PARP1- and SIRT1-dependent cellular pathways, NMNAT1 inhibition can be a promising new tool in osteosarcoma chemotherapy.
Collapse
Affiliation(s)
- Alexandra Kiss
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.K.); (C.C.); (Z.R.); (Z.P.)
- Doctoral School of Molecular Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Csaba Csikos
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.K.); (C.C.); (Z.R.); (Z.P.)
| | - Zsolt Regdon
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.K.); (C.C.); (Z.R.); (Z.P.)
| | - Zsuzsanna Polgár
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.K.); (C.C.); (Z.R.); (Z.P.)
| | - László Virág
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.K.); (C.C.); (Z.R.); (Z.P.)
- MTA-DE Cell Biology and Signaling Research Group, H-4032 Debrecen, Hungary
| | - Csaba Hegedűs
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary; (A.K.); (C.C.); (Z.R.); (Z.P.)
| |
Collapse
|
16
|
Bai M, Cui M, Li M, Yao X, Wu Y, Zheng L, Sun L, Song Q, Wang S, Liu L, Yu C, Huang Y. Discovery of a novel HDACi structure that inhibits the proliferation of ovarian cancer cells in vivo and in vitro. Int J Biol Sci 2021; 17:3493-3507. [PMID: 34512161 PMCID: PMC8416734 DOI: 10.7150/ijbs.62339] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022] Open
Abstract
Histone deacetylases (HDACs) exhibit increased expression in cancer and promote oncogenesis via the acetylation of or interactions with key transcriptional regulators. HDAC inhibitors (HDACis) decrease HDAC activity to selectively inhibit the occurrence and development of tumors. Our study screened and obtained a new HDACi structure. In vitro experiments have showed that among the leads, Z31216525 significantly inhibited the proliferation and induced the apoptosis of epithelial ovarian cancer (EOC) cells. In vivo experiments demonstrated that compared to the control, Z31216525 significantly inhibited tumor growth and showed very low toxicity. Further mechanistic studies revealed that Z31216525 may exert an antitumor effect by inhibiting the expression of the c-Myc gene. Collectively, our studies identified a novel HDACi that is expected to become a new potential therapeutic drug for EOC and has important value for the design of new HDACi structures.
Collapse
Affiliation(s)
- Miao Bai
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Mengqi Cui
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Mingyue Li
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Xinlei Yao
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Yulun Wu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Lihua Zheng
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Luguo Sun
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Qiuhang Song
- Hebei Key Laboratory of Chinese Medicine Research on Cardio-Cerebrovascular Disease, Hebei University of Chinese Medicine, Shijiazhuang, China
| | - Shuyue Wang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Lei Liu
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| | - Chunlei Yu
- Research Center of Agriculture and Medicine gene Engineering of Ministry of Education, Northeast Normal University, Changchun, 130024, China
| | - Yanxin Huang
- National Engineering Laboratory for Druggable Gene and Protein Screening, Northeast Normal University, Changchun, 130024, China
| |
Collapse
|
17
|
Lin YH, Wang H, Fiore A, Förster M, Tung LT, Belle JI, Robert F, Pelletier J, Langlais D, Nijnik A. Loss of MYSM1 inhibits the oncogenic activity of cMYC in B cell lymphoma. J Cell Mol Med 2021; 25:7089-7094. [PMID: 34114734 PMCID: PMC8278115 DOI: 10.1111/jcmm.16554] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 03/15/2021] [Accepted: 03/26/2021] [Indexed: 12/18/2022] Open
Abstract
MYSM1 is a chromatin‐binding protein, widely investigated for its functions in haematopoiesis in human and mouse; however, its role in haematologic malignancies remains unexplored. Here, we investigate the cross‐talk between MYSM1 and oncogenic cMYC in the transcriptional regulation of genes encoding ribosomal proteins, and the implications of these mechanisms for cMYC‐driven carcinogenesis. We demonstrate that in cMYC‐driven B cell lymphoma in mouse models, MYSM1‐loss represses ribosomal protein gene expression and protein synthesis. Importantly, the loss of MYSM1 also strongly inhibits cMYC oncogenic activity and protects against B cell lymphoma onset and progression in the mouse models. This advances the understanding of the molecular and transcriptional mechanisms of lymphomagenesis, and suggests MYSM1 as a possible drug target for cMYC‐driven malignancies.
Collapse
Affiliation(s)
- Yun Hsiao Lin
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - HanChen Wang
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Amanda Fiore
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - Michael Förster
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - Lin Tze Tung
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - Jad I Belle
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| | - Francis Robert
- Department of Biochemistry, McGill University, Montreal, QC, Canada.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, QC, Canada.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC, Canada
| | - David Langlais
- McGill University Research Centre on Complex Traits, McGill University, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada.,McGill University Genome Centre, McGill University, Montreal, QC, Canada.,Department of Microbiology and Immunology, McGill University, Montreal, QC, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, Montreal, QC, Canada.,McGill University Research Centre on Complex Traits, McGill University, QC, Canada
| |
Collapse
|
18
|
Ladelfa MF, Peche LY, Amato GE, Escalada MC, Zampieri S, Pascucci FA, Benevento AF, Do Porto DF, Dardis A, Schneider C, Monte M. Expression of the tumor-expressed protein MageB2 enhances rRNA transcription. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2021; 1868:119015. [PMID: 33741433 DOI: 10.1016/j.bbamcr.2021.119015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 02/22/2021] [Accepted: 03/12/2021] [Indexed: 11/17/2022]
Abstract
An essential requirement for cells to sustain a high proliferating rate is to be paired with enhanced protein synthesis through the production of ribosomes. For this reason, part of the growth-factor signaling pathways, are devoted to activate ribosome biogenesis. Enhanced production of ribosomes is a hallmark in cancer cells, which is boosted by different mechanisms. Here we report that the nucleolar tumor-protein MageB2, whose expression is associated with cell proliferation, also participates in ribosome biogenesis. Studies carried out in both siRNA-mediated MageB2 silenced cells and CRISPR/CAS9-mediated MageB2 knockout (KO) cells showed that its expression is linked to rRNA transcription increase independently of the cell proliferation status. Mechanistically, MageB2 interacts with phospho-UBF, a protein which causes the recruitment of RNA Pol I pre-initiation complex required for rRNA transcription. In addition, cells expressing MageB2 displays enhanced phospho-UBF occupancy at the rDNA gene promoter. Proteomic studies performed in MageB2 KO cells revealed impairment in ribosomal protein (RPs) content. Functionally, enhancement in rRNA production in MageB2 expressing cells, was directly associated with an increased dynamic in protein synthesis. Altogether our results unveil a novel function for a tumor-expressed protein from the MAGE-I family. Findings reported here suggest that nucleolar MageB2 might play a role in enhancing ribosome biogenesis as part of its repertoire to support cancer cell proliferation.
Collapse
Affiliation(s)
- María Fátima Ladelfa
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Leticia Yamila Peche
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, Area Science Park, Trieste, Italy
| | - Gastón Ezequiel Amato
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Micaela Carolina Escalada
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Stefania Zampieri
- Centro di Coordinamento Regionale per le Malattie Rare, Ospedale Universitario Santa Maria Della Misericordia, Udine, Italy
| | - Franco Andrés Pascucci
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Andres Fernandez Benevento
- Plataforma de Bioinformática Argentina, Instituto de Cálculo, Pabellón 2, Ciudad Universitaria, Facultad de Ciencias Exactas y Naturales, UBA, Buenos Aires, Argentina
| | - Dario Fernandez Do Porto
- Plataforma de Bioinformática Argentina, Instituto de Cálculo, Pabellón 2, Ciudad Universitaria, Facultad de Ciencias Exactas y Naturales, UBA, Buenos Aires, Argentina
| | - Andrea Dardis
- Centro di Coordinamento Regionale per le Malattie Rare, Ospedale Universitario Santa Maria Della Misericordia, Udine, Italy
| | - Claudio Schneider
- Laboratorio Nazionale del Consorzio Interuniversitario per le Biotecnologie, Area Science Park, Trieste, Italy; Dipartimento di Scienze e Tecnologie Biomediche, Università di Udine, p.le Kolbe 4, Udine, Italy
| | - Martin Monte
- Lab. Oncología Molecular, Departamento de Química Biológica, IQUIBICEN-UBA/CONICET, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Buenos Aires, Argentina.
| |
Collapse
|
19
|
Liu Y, Xu W, Xu X, Tan Z, Xu J, Ma L, Du P, Yang Y. Loss of BRMS2 induces cell growth inhibition and translation capacity reduction in colorectal cancer cells. Am J Cancer Res 2021; 11:930-944. [PMID: 33791164 PMCID: PMC7994161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 01/12/2021] [Indexed: 06/12/2023] Open
Abstract
A variety of chemotherapeutic drugs targeting ribosome processing have been developed and applied to cancer treatment mainly based on the impaired ribosome biogenesis checkpoint (IRBC). The IMP U3 small nucleolar ribonucleoprotein 3 (IMP3, BRMS2) has been identified as a participant in pre-rRNA processing for nearly twenty years. However, the roles of BRMS2 in cancers still unknown. In this research, a tissue microarray (TMA) with 151 paired tissues showed the aberrant overexpression of BRMS2 in CRC tissues which was associated with the worse prognosis. To clarify the function of BRMS2 in CRC cells, an inducible knockdown system was introduced in vitro and in vivo and the cell growth was drastically suppressed. Mechanistically, we found depletion of BRMS2 markedly decreased the protein translation rates which can limit cell growth. Furthermore, to confirm whether the IRBC played a role, multiple approaches including detection of the p53 pathway, depletion of BRMS2 in p53-mutated SW620 cells, and co-depletion of RPL11 were taken. To our surprise, IRBC was not activated. That indicated BRMS2 may play a unique role in ribosome biosynthesis and IRBC. Taken together, our results demonstrated the oncogenic function of BRMS2 in CRC cells and supported its potential as a therapeutic target.
Collapse
Affiliation(s)
- Yaofu Liu
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical CollegeSuzhou, China
| | - Weimin Xu
- Department of Colorectal Surgery, Xin-Hua Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Xin Xu
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical CollegeSuzhou, China
| | - Zhengzhi Tan
- Department of Computer Science, University of Alabama at BirminghamBirmingham, USA
| | - Jing Xu
- International School for Advanced Studies (SISSA)Trieste, Italy
| | - Lei Ma
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical CollegeSuzhou, China
| | - Peng Du
- Department of Colorectal Surgery, Xin-Hua Hospital, Shanghai Jiaotong University School of MedicineShanghai, China
| | - Yili Yang
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical CollegeSuzhou, China
- China Regional Research Center, International Centre for Genetic Engineering and BiotechnologyTaizhou, Jiangsu, China
| |
Collapse
|
20
|
Zhang Y, Zhang J, Chen X, Yang Z. Polymeric immunoglobulin receptor (PIGR) exerts oncogenic functions via activating ribosome pathway in hepatocellular carcinoma. Int J Med Sci 2021; 18:364-371. [PMID: 33390805 PMCID: PMC7757154 DOI: 10.7150/ijms.49790] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 11/03/2020] [Indexed: 02/05/2023] Open
Abstract
Objective: This report aimed to investigate the potential mechanism of polymeric immunoglobulin receptor (PIGR) in promoting cancer development in hepatocellular carcinoma (HCC). Methods: PIGR expression was investigated in Gene Expression Omnibus (GEO), Oncomine, The Cancer Genome Atlas (TCGA) and The Human Protein Atlas (HPA) databases. Relationships between PIGR and HCC survival and clinico-pathological features were conducted in TCGA. RNAseq of PIGR overexpression and knockdown samples in Bel-7404 cells were performed for identifying potential mechanisms. Results: PIGR was significantly overexpressed in tumors compared to nontumors and in HCC serum peripheral blood mononuclear cells (PBMC) than in healthy individuals (all p < 0.05). In TCGA, PIGR was highly altered in 14% HCC patients. PIGR upregulation was significantly associated with poor disease-free survival (p < 0.05). More patients recurred/progressed in PIGR altered group compared to unaltered group (p < 0.01). PIGR was significantly higher in HCC patients with incomplete cirrhosis (p < 0.001) and established cirrhosis (p < 0.05). Fewer patients had N0 lymph node stage in PIGR altered group than those in the unaltered group (p < 0.05). PIGR RNAseq revealed that ribosome signaling was the common pathway in PIGR overexpression and PIGR knockdown samples. RNAseq analysis indicated that RPL10, RPL10A, RPL12, RPL19, RPL36, RPL38, RPL41, RPL6, RPL8, RPS12, RPS14, RPS15A, RPS2, RPS27A and RPSA were significantly upregulated in PIGR overexpression group and downregulated in PIGR underexpression group (all p < 0.05). Conclusions: Aberrant PIGR was associated with HCC recurrence, and PIGR stimulated ribosome pathway might be a potential mechanism.
Collapse
MESH Headings
- Biomarkers, Tumor/blood
- Biomarkers, Tumor/genetics
- Carcinogenesis/genetics
- Carcinoma, Hepatocellular/blood
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Cell Line, Tumor
- Datasets as Topic
- Disease Progression
- Disease-Free Survival
- Gene Expression Regulation, Neoplastic
- Gene Knockdown Techniques
- Humans
- Liver/pathology
- Liver Neoplasms/blood
- Liver Neoplasms/genetics
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Neoplasm Recurrence, Local/epidemiology
- Neoplasm Recurrence, Local/genetics
- RNA-Seq
- Receptors, Polymeric Immunoglobulin/blood
- Receptors, Polymeric Immunoglobulin/genetics
- Ribosomal Proteins/metabolism
- Ribosomes/metabolism
- Signal Transduction/genetics
- Up-Regulation
Collapse
Affiliation(s)
- Yuan Zhang
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Jijie Zhang
- Department of Oncology, The People's Hospital of Danyang, Affiliated Danyang Hospital of Nantong University, Jiangsu 212300, China
| | - Xiaorong Chen
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| | - Zongguo Yang
- Department of Integrative Medicine, Shanghai Public Health Clinical Center, Fudan University, Shanghai 201508, China
| |
Collapse
|
21
|
Nait Slimane S, Marcel V, Fenouil T, Catez F, Saurin JC, Bouvet P, Diaz JJ, Mertani HC. Ribosome Biogenesis Alterations in Colorectal Cancer. Cells 2020; 9:E2361. [PMID: 33120992 PMCID: PMC7693311 DOI: 10.3390/cells9112361] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2020] [Revised: 10/23/2020] [Accepted: 10/25/2020] [Indexed: 12/24/2022] Open
Abstract
Many studies have focused on understanding the regulation and functions of aberrant protein synthesis in colorectal cancer (CRC), leaving the ribosome, its main effector, relatively underappreciated in CRC. The production of functional ribosomes is initiated in the nucleolus, requires coordinated ribosomal RNA (rRNA) processing and ribosomal protein (RP) assembly, and is frequently hyperactivated to support the needs in protein synthesis essential to withstand unremitting cancer cell growth. This elevated ribosome production in cancer cells includes a strong alteration of ribosome biogenesis homeostasis that represents one of the hallmarks of cancer cells. None of the ribosome production steps escape this cancer-specific dysregulation. This review summarizes the early and late steps of ribosome biogenesis dysregulations described in CRC cell lines, intestinal organoids, CRC stem cells and mouse models, and their possible clinical implications. We highlight how this cancer-related ribosome biogenesis, both at quantitative and qualitative levels, can lead to the synthesis of ribosomes favoring the translation of mRNAs encoding hyperproliferative and survival factors. We also discuss whether cancer-related ribosome biogenesis is a mere consequence of cancer progression or is a causal factor in CRC, and how altered ribosome biogenesis pathways can represent effective targets to kill CRC cells. The association between exacerbated CRC cell growth and alteration of specific steps of ribosome biogenesis is highlighted as a key driver of tumorigenesis, providing promising perspectives for the implementation of predictive biomarkers and the development of new therapeutic drugs.
Collapse
Affiliation(s)
- Sophie Nait Slimane
- Cancer Initiation and Tumor Cell Identity, Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1052, CNRS UMR5286 Centre Léon Bérard, 69008 Lyon, France; (S.N.S.); (V.M.); (F.C.); (P.B.)
| | - Virginie Marcel
- Cancer Initiation and Tumor Cell Identity, Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1052, CNRS UMR5286 Centre Léon Bérard, 69008 Lyon, France; (S.N.S.); (V.M.); (F.C.); (P.B.)
| | - Tanguy Fenouil
- Institute of Pathology EST, Hospices Civils de Lyon, Site-Est Groupement Hospitalier- Est, 69677 Bron, France;
| | - Frédéric Catez
- Cancer Initiation and Tumor Cell Identity, Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1052, CNRS UMR5286 Centre Léon Bérard, 69008 Lyon, France; (S.N.S.); (V.M.); (F.C.); (P.B.)
| | - Jean-Christophe Saurin
- Gastroenterology and Genetic Department, Edouard Herriot Hospital, Hospices Civils de Lyon, 69008 Lyon, France;
| | - Philippe Bouvet
- Cancer Initiation and Tumor Cell Identity, Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1052, CNRS UMR5286 Centre Léon Bérard, 69008 Lyon, France; (S.N.S.); (V.M.); (F.C.); (P.B.)
| | - Jean-Jacques Diaz
- Cancer Initiation and Tumor Cell Identity, Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1052, CNRS UMR5286 Centre Léon Bérard, 69008 Lyon, France; (S.N.S.); (V.M.); (F.C.); (P.B.)
| | - Hichem C. Mertani
- Cancer Initiation and Tumor Cell Identity, Cancer Research Center of Lyon, Université de Lyon, Université Claude Bernard Lyon 1, Inserm U1052, CNRS UMR5286 Centre Léon Bérard, 69008 Lyon, France; (S.N.S.); (V.M.); (F.C.); (P.B.)
| |
Collapse
|
22
|
Lopez Sanchez MIG, Cipullo M, Gopalakrishna S, Khawaja A, Rorbach J. Methylation of Ribosomal RNA: A Mitochondrial Perspective. Front Genet 2020; 11:761. [PMID: 32765591 PMCID: PMC7379855 DOI: 10.3389/fgene.2020.00761] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/26/2020] [Indexed: 01/02/2023] Open
Abstract
Ribosomal RNA (rRNA) from all organisms undergoes post-transcriptional modifications that increase the diversity of its composition and activity. In mitochondria, specialized mitochondrial ribosomes (mitoribosomes) are responsible for the synthesis of 13 oxidative phosphorylation proteins encoded by the mitochondrial genome. Mitoribosomal RNA is also modified, with 10 modifications thus far identified and all corresponding modifying enzymes described. This form of epigenetic regulation of mitochondrial gene expression affects mitoribosome biogenesis and function. Here, we provide an overview on rRNA methylation and highlight critical work that is beginning to elucidate its role in mitochondrial gene expression. Given the similarities between bacterial and mitochondrial ribosomes, we focus on studies involving Escherichia coli and human models. Furthermore, we highlight the use of state-of-the-art technologies, such as cryoEM in the study of rRNA methylation and its biological relevance. Understanding the mechanisms and functional relevance of this process represents an exciting frontier in the RNA biology and mitochondrial fields.
Collapse
Affiliation(s)
- M Isabel G Lopez Sanchez
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden.,Centre for Eye Research Australia, Melbourne, VIC, Australia
| | - Miriam Cipullo
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden.,Max Planck Institute for Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Shreekara Gopalakrishna
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden.,Max Planck Institute for Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Anas Khawaja
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden.,Max Planck Institute for Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| | - Joanna Rorbach
- Division of Molecular Metabolism, Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Solna, Sweden.,Max Planck Institute for Biology of Ageing - Karolinska Institutet Laboratory, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
23
|
Belle JI, Wang H, Fiore A, Petrov JC, Lin YH, Feng CH, Nguyen TTM, Tung J, Campeau PM, Behrends U, Brunet T, Leszinski GS, Gros P, Langlais D, Nijnik A. MYSM1 maintains ribosomal protein gene expression in hematopoietic stem cells to prevent hematopoietic dysfunction. JCI Insight 2020; 5:125690. [PMID: 32641579 DOI: 10.1172/jci.insight.125690] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 05/21/2020] [Indexed: 01/09/2023] Open
Abstract
Ribosomopathies are congenital disorders caused by mutations in the genes encoding ribosomal and other functionally related proteins. They are characterized by anemia, other hematopoietic and developmental abnormalities, and p53 activation. Ribosome assembly requires coordinated expression of many ribosomal protein (RP) genes; however, the regulation of RP gene expression, especially in hematopoietic stem cells (HSCs), remains poorly understood. MYSM1 is a transcriptional regulator essential for HSC function and hematopoiesis. We established that HSC dysfunction in Mysm1 deficiency is driven by p53; however, the mechanisms of p53 activation remained unclear. Here, we describe the transcriptome of Mysm1-deficient mouse HSCs and identify MYSM1 genome-wide DNA binding sites. We establish a direct role for MYSM1 in RP gene expression and show a reduction in protein synthesis in Mysm1-/- HSCs. Loss of p53 in mice fully rescues Mysm1-/- anemia phenotype but not RP gene expression, indicating that RP gene dysregulation is a direct outcome of Mysm1 deficiency and an upstream mediator of Mysm1-/- phenotypes through p53 activation. We characterize a patient with a homozygous nonsense MYSM1 gene variant, and we demonstrate reduced protein synthesis and increased p53 levels in patient hematopoietic cells. Our work provides insights into the specialized mechanisms regulating RP gene expression in HSCs and establishes a common etiology of MYSM1 deficiency and ribosomopathy syndromes.
Collapse
Affiliation(s)
- Jad I Belle
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - HanChen Wang
- Department of Physiology.,McGill University Research Centre on Complex Traits, and.,Department of Human Genetics, McGill University, Quebec, Canada
| | - Amanda Fiore
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Jessica C Petrov
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Yun Hsiao Lin
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Chu-Han Feng
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Thi Tuyet Mai Nguyen
- Centre Hospitalier Universitaire St. Justine Research Center, University of Montreal, Quebec, Canada
| | - Jacky Tung
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| | - Philippe M Campeau
- Centre Hospitalier Universitaire St. Justine Research Center, University of Montreal, Quebec, Canada
| | | | - Theresa Brunet
- Institute of Human Genetics, Technische Universität München (TUM), Munich, Germany
| | - Gloria Sarah Leszinski
- Institute of Human Genetics, Technische Universität München (TUM), Munich, Germany.,Institute of Human Genetics, Helmholtz Zentrum München, Neuherberg, Germany
| | - Philippe Gros
- McGill University Research Centre on Complex Traits, and.,Department of Biochemistry and.,The Rosalind and Morris Goodman Cancer Research Centre, McGill University, Quebec, Canada
| | - David Langlais
- McGill University Research Centre on Complex Traits, and.,Department of Human Genetics, McGill University, Quebec, Canada.,McGill University Genome Centre, Montreal, Quebec, Canada
| | - Anastasia Nijnik
- Department of Physiology.,McGill University Research Centre on Complex Traits, and
| |
Collapse
|
24
|
Shirakawa Y, Hide T, Yamaoka M, Ito Y, Ito N, Ohta K, Shinojima N, Mukasa A, Saito H, Jono H. Ribosomal protein S6 promotes stem-like characters in glioma cells. Cancer Sci 2020; 111:2041-2051. [PMID: 32246865 PMCID: PMC7293102 DOI: 10.1111/cas.14399] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/18/2020] [Accepted: 03/20/2020] [Indexed: 12/11/2022] Open
Abstract
Glioblastoma multiforme (GBM), a lethal brain tumor developing in the white matter of the adult brain, contains a small population of GBM stem cells (GSCs), which potentially cause chemotherapeutic resistance and tumor recurrence. However, the mechanisms underlying the pathogenesis and maintenance of GSCs remain largely unknown. A recent study reported that incorporation of ribosomes and ribosomal proteins into somatic cells promoted lineage trans-differentiation toward multipotency. This study aimed to investigate the mechanism underlying stemness acquisition in GBM cells by focusing on 40S ribosomal protein S6 (RPS6). RPS6 was significantly upregulated in high-grade glioma and localized at perivascular, perinecrotic, and border niches in GBM tissues. siRNA-mediated RPS6 knock-down significantly suppressed the characteristics of GSCs, including their tumorsphere potential and GSC marker expression; STAT3 was downregulated in GBM cells. RPS6 overexpression enhanced the tumorsphere potential of GSCs and these effects were attenuated by STAT3 inhibitor (AG490). Moreover, RPS6 expression was significantly correlated with SOX2 expression in different glioma grades. Immunohistochemistry data herein indicated that RPS6 was predominant in GSC niches, concurrent with the data from IVY GAP databases. Furthermore, RPS6 and other ribosomal proteins were upregulated in GSC-predominant areas in this database. The present results indicate that, in GSC niches, ribosomal proteins play crucial roles in the development and maintenance of GSCs and are clinically associated with chemoradioresistance and GBM recurrence.
Collapse
Affiliation(s)
- Yuki Shirakawa
- Department of Clinical Pharmaceutical SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| | - Takuichiro Hide
- Department of NeurosurgeryKitasato University School of MedicineSagamiharaJapan
| | - Michiko Yamaoka
- Department of Clinical Pharmaceutical SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| | - Yuki Ito
- Department of Clinical Pharmaceutical SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
| | - Naofumi Ito
- Department of Developmental NeurobiologyGraduate School of Life SciencesKumamoto UniversityKumamotoJapan
| | - Kunimasa Ohta
- Department of Developmental NeurobiologyGraduate School of Life SciencesKumamoto UniversityKumamotoJapan
| | - Naoki Shinojima
- Department of NeurosurgeryFaculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Akitake Mukasa
- Department of NeurosurgeryFaculty of Life SciencesKumamoto UniversityKumamotoJapan
| | - Hideyuki Saito
- Department of Clinical Pharmaceutical SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
- Department of PharmacyKumamoto University HospitalKumamoto CityJapan
| | - Hirofumi Jono
- Department of Clinical Pharmaceutical SciencesGraduate School of Pharmaceutical SciencesKumamoto UniversityKumamotoJapan
- Department of PharmacyKumamoto University HospitalKumamoto CityJapan
| |
Collapse
|
25
|
Ishaq M, Ojha R, Sharma AP, Singh SK. Autophagy in cancer: Recent advances and future directions. Semin Cancer Biol 2020; 66:171-181. [PMID: 32201367 DOI: 10.1016/j.semcancer.2020.03.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 02/10/2020] [Accepted: 03/16/2020] [Indexed: 02/06/2023]
Abstract
Autophagy is being explored as a potential therapeutic target for enhancing the cytotoxic effects of chemotherapeutic regimens in various malignancies. Autophagy plays a very important role in cancer pathogenesis. Here, we discuss the updates on the modulation of autophagy via dynamic interactions with different organelles and the exploitation of selective autophagy for exploring therapeutic strategies. We further discuss the role of autophagy inhibitors in cancer preclinical and clinical trials, novel autophagy inhibitors, and challenges likely to be faced by clinicians while inducting autophagy modulators in clinical practice.
Collapse
Affiliation(s)
- Mohd Ishaq
- School of Medicine, Department of Pathology, Stanford University, CA, USA.
| | - Rani Ojha
- School of Medicine, Department of Pathology, Stanford University, CA, USA.
| | - Aditya P Sharma
- Department of Urology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Shrawan K Singh
- Department of Urology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
26
|
Napoli M, Flores ER. The p53 family reaches the final frontier: the variegated regulation of the dark matter of the genome by the p53 family in cancer. RNA Biol 2020; 17:1636-1647. [PMID: 31910062 PMCID: PMC7567494 DOI: 10.1080/15476286.2019.1710054] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
The tumour suppressor p53 and its paralogues, p63 and p73, are essential to maintain cellular homoeostasis and the integrity of the cell's genetic material, thus meriting the title of 'guardians of the genome'. The p53 family members are transcription factors and fulfill their activities by controlling the expression of protein-coding and non-coding genes. Here, we review how the latter group transcended from the 'dark matter' of the transcriptome, providing unexpected and intriguing anti-cancer therapeutic strategies.
Collapse
Affiliation(s)
- Marco Napoli
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL, USA
| | - Elsa R Flores
- Department of Molecular Oncology, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL, USA.,Cancer Biology and Evolution Program, H. Lee Moffitt Cancer Center and Research Institute , Tampa, FL, USA
| |
Collapse
|
27
|
Prattes M, Lo YH, Bergler H, Stanley RE. Shaping the Nascent Ribosome: AAA-ATPases in Eukaryotic Ribosome Biogenesis. Biomolecules 2019; 9:E715. [PMID: 31703473 PMCID: PMC6920918 DOI: 10.3390/biom9110715] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 02/08/2023] Open
Abstract
AAA-ATPases are molecular engines evolutionarily optimized for the remodeling of proteins and macromolecular assemblies. Three AAA-ATPases are currently known to be involved in the remodeling of the eukaryotic ribosome, a megadalton range ribonucleoprotein complex responsible for the translation of mRNAs into proteins. The correct assembly of the ribosome is performed by a plethora of additional and transiently acting pre-ribosome maturation factors that act in a timely and spatially orchestrated manner. Minimal disorder of the assembly cascade prohibits the formation of functional ribosomes and results in defects in proliferation and growth. Rix7, Rea1, and Drg1, which are well conserved across eukaryotes, are involved in different maturation steps of pre-60S ribosomal particles. These AAA-ATPases provide energy for the efficient removal of specific assembly factors from pre-60S particles after they have fulfilled their function in the maturation cascade. Recent structural and functional insights have provided the first glimpse into the molecular mechanism of target recognition and remodeling by Rix7, Rea1, and Drg1. Here we summarize current knowledge on the AAA-ATPases involved in eukaryotic ribosome biogenesis. We highlight the latest insights into their mechanism of mechano-chemical complex remodeling driven by advanced cryo-EM structures and the use of highly specific AAA inhibitors.
Collapse
Affiliation(s)
- Michael Prattes
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010 Graz, Austria;
| | - Yu-Hua Lo
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, Durham, NC 27709, USA;
| | - Helmut Bergler
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010 Graz, Austria;
| | - Robin E. Stanley
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, 111 T. W. Alexander Drive, Research Triangle Park, Durham, NC 27709, USA;
| |
Collapse
|
28
|
Awad D, Prattes M, Kofler L, Rössler I, Loibl M, Pertl M, Zisser G, Wolinski H, Pertschy B, Bergler H. Inhibiting eukaryotic ribosome biogenesis. BMC Biol 2019; 17:46. [PMID: 31182083 PMCID: PMC6558755 DOI: 10.1186/s12915-019-0664-2] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Ribosome biogenesis is a central process in every growing cell. In eukaryotes, it requires more than 250 non-ribosomal assembly factors, most of which are essential. Despite this large repertoire of potential targets, only very few chemical inhibitors of ribosome biogenesis are known so far. Such inhibitors are valuable tools to study this highly dynamic process and elucidate mechanistic details of individual maturation steps. Moreover, ribosome biogenesis is of particular importance for fast proliferating cells, suggesting its inhibition could be a valid strategy for treatment of tumors or infections. RESULTS We systematically screened ~ 1000 substances for inhibitory effects on ribosome biogenesis using a microscopy-based screen scoring ribosomal subunit export defects. We identified 128 compounds inhibiting maturation of either the small or the large ribosomal subunit or both. Northern blot analysis demonstrates that these inhibitors cause a broad spectrum of different rRNA processing defects. CONCLUSIONS Our findings show that the individual inhibitors affect a wide range of different maturation steps within the ribosome biogenesis pathway. Our results provide for the first time a comprehensive set of inhibitors to study ribosome biogenesis by chemical inhibition of individual maturation steps and establish the process as promising druggable pathway for chemical intervention.
Collapse
Affiliation(s)
- Dominik Awad
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
- Present address: Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Prattes
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Lisa Kofler
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Ingrid Rössler
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Mathias Loibl
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Melanie Pertl
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Gertrude Zisser
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Heimo Wolinski
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria
| | - Brigitte Pertschy
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria.
| | - Helmut Bergler
- Institute of Molecular Biosciences, University of Graz, Humboldtstrasse 50/EG, A-8010, Graz, Austria.
| |
Collapse
|
29
|
Lessard F, Brakier-Gingras L, Ferbeyre G. Ribosomal Proteins Control Tumor Suppressor Pathways in Response to Nucleolar Stress. Bioessays 2019; 41:e1800183. [PMID: 30706966 DOI: 10.1002/bies.201800183] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 12/18/2018] [Indexed: 01/05/2023]
Abstract
Ribosome biogenesis includes the making and processing of ribosomal RNAs, the biosynthesis of ribosomal proteins from their mRNAs in the cytosol and their transport to the nucleolus to assemble pre-ribosomal particles. Several stresses including cellular senescence reduce nucleolar rRNA synthesis and maturation increasing the availability of ribosome-free ribosomal proteins. Several ribosomal proteins can activate the p53 tumor suppressor pathway but cells without p53 can still arrest their proliferation in response to an imbalance between ribosomal proteins and mature rRNA production. Recent results on senescence-associated ribogenesis defects (SARD) show that the ribosomal protein S14 (RPS14 or uS11) can act as a CDK4/6 inhibitor linking ribosome biogenesis defects to the main engine of cell cycle progression. This work offers new insights into the regulation of the cell cycle and suggests novel avenues to design anticancer drugs.
Collapse
Affiliation(s)
- Frédéric Lessard
- Department of Biochemistry and Molecular Medicine, Université de Montréal, C.P. 6128, Succ. Centre-Ville, Montréal, Québec H3C 3J7, Canada
| | - Léa Brakier-Gingras
- Department of Biochemistry and Molecular Medicine, Université de Montréal, C.P. 6128, Succ. Centre-Ville, Montréal, Québec H3C 3J7, Canada
| | - Gerardo Ferbeyre
- Department of Biochemistry and Molecular Medicine, Université de Montréal, C.P. 6128, Succ. Centre-Ville, Montréal, Québec H3C 3J7, Canada.,CRCHUM, 900 Saint-Denis - bureau R10.432, Montréal, Québec H2X 0A9, Canada
| |
Collapse
|
30
|
El Hassouni B, Sarkisjan D, Vos JC, Giovannetti E, Peters GJ. Targeting the Ribosome Biogenesis Key Molecule Fibrillarin to Avoid Chemoresistance. Curr Med Chem 2019; 26:6020-6032. [PMID: 30501594 DOI: 10.2174/0929867326666181203133332] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 11/07/2018] [Accepted: 11/07/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Inherent or acquired chemo resistance in cancer patients has been a perpetual limitation in cancer treatment. Expanding knowledge on essential cellular processes opens a new window for therapeutic targeting. Ribosome biogenesis is a process that shows potential due to its fundamental role in cell development and contribution to tumorigenesis as a result of its upregulation. Inhibiting components of ribosome biogenesis has been explored and has shown interesting results. Yet, an important key component, methyltransferase Fibrillarin (FBL), which influences both the abundance and composition of ribosomes, has not been exploited thus far. METHODS In this literature review, we describe relevant aspects of ribosome biogenesis in cancer to emphasize the potential of FBL as a therapeutic target, in order to lower the genotoxic effects of anti-cancer treatment. RESULTS Remarkably, the amplification of the 19q13 cytogenetic band, including the gene coding for FBL, correlated to cell viability and resistance in pancreatic cells as well as to a trend toward a shorter survival in pancreatic cancer patients. Targeting ribosome biogenesis, more specifically compared to the secondary effects of chemotherapeutics such as 5-fluorouracil or oxaliplatin, has been achieved by compound CX-5461. The cell dependent activity of this Pol I inhibitor has been reported in ovarian cancer, melanoma and leukemia models with active or mutated p53 status, presenting a promising mechanism to evade p53 resistance. CONCLUSION Targeting critical ribosome biogenesis components in order to decrease the genotoxic activity in cancer cell looks promising. Hence, we believe that targeting key protein rRNA methyltransferase FBL shows great potential, due to its pivotal role in ribosome biogenesis, its correlation to an improved survival rate at low expression in breast cancer patients and its association with p53.
Collapse
Affiliation(s)
- Btissame El Hassouni
- Department of Medical Oncology, VU University Medical Center- Cancer Center Amsterdam, De Boelelaan 1118, 1081 HV Amsterdam, Netherlands
| | - Dzjemma Sarkisjan
- Department of Medical Oncology, VU University Medical Center- Cancer Center Amsterdam, De Boelelaan 1118, 1081 HV Amsterdam, Netherlands
| | - J Chris Vos
- AIMMS-Division of Molecular Toxicology, Department of Chemistry and Pharmaceutical Sciences, VU University Amsterdam, O|2 building, De Boelelaan 1108, 1081 HZ Amsterdam, Netherlands
| | - Elisa Giovannetti
- Department of Medical Oncology, VU University Medical Center- Cancer Center Amsterdam, De Boelelaan 1118, 1081 HV Amsterdam, Netherlands
- Cancer Pharmacology Lab, AIRC Start-Up Unit, University of Pisa, Pisa, Italy
| | - Godefridus J Peters
- Department of Medical Oncology, VU University Medical Center- Cancer Center Amsterdam, De Boelelaan 1118, 1081 HV Amsterdam, Netherlands
| |
Collapse
|
31
|
Abstract
Ribosomes, which synthesize the proteins of a cell, comprise ribosomal RNA and ribosomal proteins, which coassemble hierarchically during a process termed ribosome biogenesis. Historically, biochemical and molecular biology approaches have revealed how preribosomal particles form and mature in consecutive steps, starting in the nucleolus and terminating after nuclear export into the cytoplasm. However, only recently, due to the revolution in cryo-electron microscopy, could pseudoatomic structures of different preribosomal particles be obtained. Together with in vitro maturation assays, these findings shed light on how nascent ribosomes progress stepwise along a dynamic biogenesis pathway. Preribosomes assemble gradually, chaperoned by a myriad of assembly factors and small nucleolar RNAs, before they reach maturity and enter translation. This information will lead to a better understanding of how ribosome synthesis is linked to other cellular pathways in humans and how it can cause diseases, including cancer, if disturbed.
Collapse
Affiliation(s)
- Jochen Baßler
- Biochemistry Center, University of Heidelberg, 69120 Heidelberg, Germany; ,
| | - Ed Hurt
- Biochemistry Center, University of Heidelberg, 69120 Heidelberg, Germany; ,
| |
Collapse
|
32
|
Vadivel Gnanasundram S, Fåhraeus R. Translation Stress Regulates Ribosome Synthesis and Cell Proliferation. Int J Mol Sci 2018; 19:ijms19123757. [PMID: 30486342 PMCID: PMC6321632 DOI: 10.3390/ijms19123757] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 11/23/2018] [Accepted: 11/24/2018] [Indexed: 12/11/2022] Open
Abstract
Ribosome and protein synthesis are major metabolic events that control cellular growth and proliferation. Impairment in ribosome biogenesis pathways and mRNA translation is associated with pathologies such as cancer and developmental disorders. Processes that control global protein synthesis are tightly regulated at different levels by numerous factors and linked with multiple cellular signaling pathways. Several of these merge on the growth promoting factor c-Myc, which induces ribosome biogenesis by stimulating Pol I, Pol II, and Pol III transcription. However, how cells sense and respond to mRNA translation stress is not well understood. It was more recently shown that mRNA translation stress activates c-Myc, through a specific induction of E2F1 synthesis via a PI3Kδ-dependent pathway. This review focuses on how this novel feedback pathway stimulates cellular growth and proliferation pathways to synchronize protein synthesis with ribosome biogenesis. It also describes for the first time the oncogenic activity of the mRNA, and not the encoded protein.
Collapse
Affiliation(s)
| | - Robin Fåhraeus
- Inserm UMRS1162, Institut de Génétique Moléculaire, Université Paris 7, Hôpital St. Louis, F-75010 Paris, France.
- RECAMO, Masaryk Memorial Cancer Institute, Zluty kopec 7, 65653 Brno, Czech Republic.
- Department of Medical Biosciences, Building 6M, Umeå University, 901 85 Umeå, Sweden.
- ICCVS, University of Gdańsk, Science, ul. Wita Stwosza 63, 80-308 Gdańsk, Poland.
| |
Collapse
|
33
|
Fan S, Liang Z, Gao Z, Pan Z, Han S, Liu X, Zhao C, Yang W, Pan Z, Feng W. Identification of the key genes and pathways in prostate cancer. Oncol Lett 2018; 16:6663-6669. [PMID: 30405806 PMCID: PMC6202544 DOI: 10.3892/ol.2018.9491] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 09/17/2018] [Indexed: 12/13/2022] Open
Abstract
Prostate cancer (PCa) is one of the most common malignancies in men globally. The aim of the present study was to identify the key genes and pathways involved in the occurrence of PCa. Gene expression profile (GSE55945) was downloaded from Gene Expression Omnibus, and the differentially expressed genes (DEGs) were identified. Subsequently, Gene ontology analysis, KEGG pathway analysis and protein-protein interaction (PPI) analysis of DEGs were performed. Finally, the identified key genes were confirmed by immunohistochemistry. The GO analysis results showed that the DEGs were mainly participated in cell cycle, cell division, cell development and cell junction. The KEGG pathway analysis showed that the DEGs were mainly enriched in proteoglycans in cancer, endocytosis, focal adhesion and hippo signaling pathway. The PPI analysis results showed that RPS21, FOXO1, BIRC5, POLR2H, RPL22L1 and NPM1 were the key genes involved in the occurrence of PCa, and the Module analysis indicated that the occurrence of PCa was associated with cell cycle, oocyte meiosis and ribosome biogenesis. IHC result showed that the expression of RPS21, BIRC5, POLR2H, RPL22L1 and NPM1 were significantly upregulated in PCa, while the expression of FOXO1 was significantly downregulated in PCa, matching with the bioinformatics analysis. Taken together, several key genes and pathways were identified involved in PCa, which might provide the potential biomarker for prognosis, diagnosis and drug targets.
Collapse
Affiliation(s)
- Shutong Fan
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zumu Liang
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhiqin Gao
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhiwei Pan
- Department of Internal Medicine, Laizhou Development Zone Hospital, Yantai, Shandong 261400, P.R. China
| | - Shaojie Han
- Animal Epidemic Prevention and Epidemic Control Center, Changle County Bureau of Animal Health and Production, Weifang, Shandong 262400, P.R. China
| | - Xiaoying Liu
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Chunling Zhao
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Weiwei Yang
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Zhifang Pan
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Weiguo Feng
- College of Bioscience and Technology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| |
Collapse
|
34
|
Derenzini E, Rossi A, Treré D. Treating hematological malignancies with drugs inhibiting ribosome biogenesis: when and why. J Hematol Oncol 2018; 11:75. [PMID: 29855342 PMCID: PMC5984324 DOI: 10.1186/s13045-018-0609-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Accepted: 04/26/2018] [Indexed: 01/05/2023] Open
Abstract
It is well known that chemotherapy can cure only some cancers in advanced stage, mostly those with an intact p53 pathway. Hematological cancers such as lymphoma and certain forms of leukemia are paradigmatic examples of such scenario. Recent evidence indicates that the efficacy of many of the alkylating and intercalating agents, antimetabolites, topoisomerase, and kinase inhibitors used in cancer therapy is largely due to p53 stabilization and activation consequent to the inhibition of ribosome biogenesis. In this context, innovative drugs specifically hindering ribosome biogenesis showed preclinical activity and are currently in early clinical development in hematological malignancies. The mechanism of p53 stabilization after ribosome biogenesis inhibition is a multistep process, depending on specific factors that can be altered in tumor cells, which can affect the antitumor efficacy of ribosome biogenesis inhibitors (RiBi). In the present review, the basic mechanisms underlying the anticancer activity of RiBi are discussed based on the evidence deriving from available preclinical and clinical studies, with the purpose of defining when and why the treatment with drugs inhibiting ribosomal biogenesis could be highly effective in hematological malignancies.
Collapse
Affiliation(s)
- Enrico Derenzini
- European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy.
| | - Alessandra Rossi
- European Institute of Oncology, Via Ripamonti 435, 20141, Milan, Italy
| | - Davide Treré
- DIMES, Università di Bologna, Via Massarenti 9, Bologna, Italy.
| |
Collapse
|
35
|
Bram Ednersson S, Stenson M, Stern M, Enblad G, Fagman H, Nilsson-Ehle H, Hasselblom S, Andersson PO. Expression of ribosomal and actin network proteins and immunochemotherapy resistance in diffuse large B cell lymphoma patients. Br J Haematol 2018; 181:770-781. [PMID: 29767447 DOI: 10.1111/bjh.15259] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Accepted: 03/05/2018] [Indexed: 10/25/2022]
Abstract
Diffuse large B cell lymphoma (DLBCL) patients with early relapse or refractory disease have a very poor outcome. Immunochemotherapy resistance will probably, also in the era of targeted drugs, remain the major cause of treatment failure. We used proteomic mass spectrometry to analyse the global protein expression of micro-dissected formalin-fixed paraffin-embedded tumour tissues from 97 DLBCL patients: 44 with primary refractory disease or relapse within 1 year from diagnosis (REF/REL), and 53 who were progression-free more than 5 years after diagnosis (CURED). We identified 2127 proteins: 442 were found in all patients and 102 were differentially expressed. Sixty-five proteins were overexpressed in REF/REL patients, of which 46 were ribosomal proteins (RPs) compared with 2 of the 37 overexpressed proteins in CURED patients (P = 7·6 × 10-10 ). Twenty of 37 overexpressed proteins in CURED patients were associated with actin regulation, compared with 1 of 65 in REF/REL patients (P = 1·4 × 10-9 ). Immunohistochemical staining showed higher expression of RPS5 and RPL17 in REF/REL patients while MARCKS-like protein, belonging to the actin network, was more highly expressed in CURED patients. Even though functional studies aimed at individual proteins and protein interactions to evaluate potential clinical effect are needed, our findings suggest new mechanisms behind immunochemotherapy resistance in DLBCL.
Collapse
Affiliation(s)
- Susanne Bram Ednersson
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Martin Stenson
- Section of Haematology, Department of Medicine, Kungälvs Hospital, Kungälv, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Mimmie Stern
- Section of Haematology, Department of Medicine, South Älvsborg Hospital, Borås, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Gunilla Enblad
- Department of Immunology, Genetics and Pathology/Experimental and Clinical Oncology, Uppsala University, Uppsala, Sweden
| | - Henrik Fagman
- Department of Pathology, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Herman Nilsson-Ehle
- Section of Haematology and Coagulation, Sahlgrenska University Hospital, Gothenburg, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| | - Sverker Hasselblom
- Department of Research, Development and Education, Region Halland, Halmstad, Sweden
| | - Per-Ola Andersson
- Section of Haematology, Department of Medicine, South Älvsborg Hospital, Borås, Sweden.,Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
36
|
Gelgor A, Gam Ze Letova C, Yegorov Y, Kalt I, Sarid R. Nucleolar stress enhances lytic reactivation of the Kaposi's sarcoma-associated herpesvirus. Oncotarget 2018; 9:13822-13833. [PMID: 29568397 PMCID: PMC5862618 DOI: 10.18632/oncotarget.24497] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2017] [Accepted: 02/01/2018] [Indexed: 02/07/2023] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is a human tumorigenic virus exhibiting two forms of infection, latent and lytic. Latent infection is abortive and allows the virus to establish lifelong infection, while lytic infection is productive, and is needed for virus dissemination within the host and between hosts. Latent infection may reactivate and switch towards the lytic cycle. This switch is a critical step in the maintenance of long-term infection and for the development of KSHV-related neoplasms. In this study, we examined the effect of nucleolar stress, manifested by failure in ribosome biogenesis or function and often coupled with p53 activation, on lytic reactivation of KSHV. To this end, we induced nucleolar stress by treatment with Actinomycin D, CX-5461 or BMH-21. Treatment with these compounds alone did not induce the lytic cycle. However, enhancement of the lytic cycle by these compounds was evident when combined with expression of the viral protein K-Rta. Further experiments employing combined treatments with Nutlin-3, knock-down of p53 and isogenic p53+/+ and p53-/- cells indicated that the enhancement of lytic reactivation by nucleolar stress does not depend on p53. Thus, our study identifies nucleolar stress as a novel regulator of KSHV infection, which synergizes with K-Rta expression to increase lytic reactivation. This suggests that certain therapeutic interventions, which induce nucleolar stress, may affect the outcome of KSHV infection.
Collapse
Affiliation(s)
- Anastasia Gelgor
- The Mina and Everard Goodman Faculty of Life Sciences and Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan, Israel
| | - Chen Gam Ze Letova
- The Mina and Everard Goodman Faculty of Life Sciences and Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan, Israel
| | - Yana Yegorov
- The Mina and Everard Goodman Faculty of Life Sciences and Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan, Israel
| | - Inna Kalt
- The Mina and Everard Goodman Faculty of Life Sciences and Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan, Israel
| | - Ronit Sarid
- The Mina and Everard Goodman Faculty of Life Sciences and Advanced Materials and Nanotechnology Institute, Bar Ilan University, Ramat-Gan, Israel
| |
Collapse
|
37
|
Peng Q, Lin K, Chang T, Zou L, Xing P, Shen Y, Zhu Y. Identification of genomic expression differences between right-sided and left-sided colon cancer based on bioinformatics analysis. Onco Targets Ther 2018; 11:609-618. [PMID: 29430186 PMCID: PMC5797455 DOI: 10.2147/ott.s154207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Introduction More and more findings have demonstrated that right-sided colon cancers (RCC) and left-sided colon cancers (LCC) are distinct clinical and biological entities and suggest that they should be treated as different diseases. However, the reasons why RCC and LCC harbor different clinical and biological features remain unclear. Materials and methods To identify the genomic expression differences between RCC and LCC and uncover the mechanisms underlying these differences, we chose the gene expression profiles of GSE14333 from the Gene Expression Omnibus (GEO) database as an object of study. Then, a systematic and integrative bioinformatics analysis was performed to research the possible mechanism of the differentially expressed (DE) genes from the Gene Expression Omnibus dataset including gene ontology (GO) analysis, pathway enrichment analysis, protein-protein interaction (PPI) network construction, and module analysis. Totally, we extracted 3,793 DE genes from samples of colon cancer including 1,961 genes upregulated in RCC and 1,832 genes upregulated in LCC from the selected dataset. Results The results of GO and pathway enrichment analysis indicated that RCC and LCC could predispose to different pathways regulated by different genes. Based on the PPI network, PCNA, TP53, HSP90AA1, CSNK2A1, UBB, LRRK2, ABL1, PRKACA, CAV1, and JUN were identified as the key hub genes. Also, significant modules were screened from the PPI network. Conclusion In conclusion, the present study indicated that the identified genes and pathways may promote new insights into the underlying molecular mechanisms contributing to the difference between RCC and LCC and might be used as specific therapeutic targets and prognostic markers for the personalized treatment of RCC and LCC.
Collapse
Affiliation(s)
- Qiliang Peng
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou.,Suzhou Key Laboratory for Radiation Oncology, Suzhou
| | - Kaisu Lin
- Department of Oncology, Nantong Rich Hospital, Nantong
| | - Tao Chang
- Department of General Surgery, The Sixth People's Hospital of Kunshan, Kunshan, People's Republic of China
| | - Li Zou
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou.,Suzhou Key Laboratory for Radiation Oncology, Suzhou
| | - Pengfei Xing
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou.,Suzhou Key Laboratory for Radiation Oncology, Suzhou
| | - Yuntian Shen
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou.,Suzhou Key Laboratory for Radiation Oncology, Suzhou
| | - Yaqun Zhu
- Department of Radiotherapy & Oncology, The Second Affiliated Hospital of Soochow University, Suzhou.,Institute of Radiotherapy & Oncology, Soochow University, Suzhou.,Suzhou Key Laboratory for Radiation Oncology, Suzhou
| |
Collapse
|
38
|
Abstract
MicroRNA (miRNA) is the noncoding gene: therefore, the miRNA gene inheritably controls protein gene expression through transcriptional and post-transcriptional levels. Aberrant expression of miRNA genes causes various human diseases, especially cancers. Although cancer is a complex disease, cancer/miRNA implication has yet been grasped from the perspective of miRNA profile in bed side. Since miRNA is the mobile genetic element, the clinical verification of miRNA in microvesicle of blood is too much straggle to predict potential cancer/miRNA associations without bioinformatical computing. Further, experimental investigation of miRNA/cancer pathways is expensive and time-consuming. While the accumulated data (big data) of miRNA profiles has been on line as the databases in cancers, using the database algorithms for miRNA target prediction have reduced required time for conventional experiments and have cut the cost. Computational prediction of miRNA/target mRNA has shown numerous significant outcomes that are unobtainable only by experimental approaches. However, ID of miRNA in the annotation is an arbitrary number and the ID is not related with miRNA its functions. Therefore, it has not been physicochemically shown why multiple miRNAs in blood or tissues are useful for diagnosis and porgnosis of human diseases or why function of single miRNA in cancer is rendered to oncomir or tumopr suppressor. In addition, it is less cleared why environmental factors, such as temperature, radiation, therapeutic anti-cancer immune or chemical agents can alter the expression of miRNAs in the cell. The ceRNA theory would not be enough for the investigation of such subjects. Given miRNA/target prediction tools, to elucidate such issues with computer simulation we have previously introduced the quantum miRNA/miRNA interaction as a new scoring using big database. The quantum score was implicated in miRNA synergisms in cancer and participated in the miRNA/target interaction on human diseases. On the other hand, ribosomal RNA (rRNA) is the dominant RNA species of the cells. It is well known that ribosomopathies, such as Diamond-Blackfan anemia, dyskeratiosis congenital, Shwachman-Diamond syndrome, 5q-myelodysplastic syndrome, Treacher Collins syndrome, cartilage-hair hypoplasia, North American Indian childhood cirrhosis, isolated congenital asplenia, Bowen-Conradi syndrome and cancer are caused by altered expression of ribosomal proteins or rRNA genes. We have proposed the hypothesis that the interaction among miRNAs from rRNA and/or other cellular miRNAs would be involved into cancer as the ribosomopathy. Subsequently, we found rRNA-derived miRNAs (rmiRNAs) by using the sequence homology search (miPS) with miRNA database (miRBase). Further, the pathway related with cancer between rmiRNA/target protein gene was predicted by miRNA entangling target sorting (METS) algorithm. In this chapter, we describe about the usage of in silico miRNA identification program, miRNA/target prediction search through the database and quantum language of miRNA by the METS, and the ontology analysis. In particular, the METS algorithm according to the quantum value would be useful simulator to discover a new therapeutic target aganist cancer. It may also partly contribute to the elucidation of complex mechanisms and development of agents of anti-cancer.
Collapse
Affiliation(s)
- Yoichi Robertus Fujii
- Retroviral Genetics Group, Pharmaco-MicroRNA Genomics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya, Aichi, Japan.
| |
Collapse
|
39
|
Kalinina NO, Makarova S, Makhotenko A, Love AJ, Taliansky M. The Multiple Functions of the Nucleolus in Plant Development, Disease and Stress Responses. FRONTIERS IN PLANT SCIENCE 2018; 9:132. [PMID: 29479362 PMCID: PMC5811523 DOI: 10.3389/fpls.2018.00132] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Accepted: 01/23/2018] [Indexed: 05/18/2023]
Abstract
The nucleolus is the most conspicuous domain in the eukaryotic cell nucleus, whose main function is ribosomal RNA (rRNA) synthesis and ribosome biogenesis. However, there is growing evidence that the nucleolus is also implicated in many other aspects of cell biology, such as regulation of cell cycle, growth and development, senescence, telomerase activity, gene silencing, responses to biotic and abiotic stresses. In the first part of the review, we briefly assess the traditional roles of the plant nucleolus in rRNA synthesis and ribosome biogenesis as well as possible functions in other RNA regulatory pathways such as splicing, nonsense-mediated mRNA decay and RNA silencing. In the second part of the review we summarize recent progress and discuss already known and new hypothetical roles of the nucleolus in plant growth and development. In addition, this part will highlight studies showing new nucleolar functions involved in responses to pathogen attack and abiotic stress. Cross-talk between the nucleolus and Cajal bodies is also discussed in the context of their association with poly(ADP ribose)polymerase (PARP), which is known to play a crucial role in various physiological processes including growth, development and responses to biotic and abiotic stresses.
Collapse
Affiliation(s)
- Natalia O. Kalinina
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- Natalia O. Kalinina
| | - Svetlana Makarova
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Antonida Makhotenko
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Michael Taliansky
- Branch of the Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
- The James Hutton Institute, Dundee, United Kingdom
- *Correspondence: Michael Taliansky
| |
Collapse
|
40
|
Fraile JM, Campos-Iglesias D, Rodríguez F, Astudillo A, Vilarrasa-Blasi R, Verdaguer-Dot N, Prado MA, Paulo JA, Gygi SP, Martín-Subero JI, Freije JMP, López-Otín C. Loss of the deubiquitinase USP36 destabilizes the RNA helicase DHX33 and causes preimplantation lethality in mice. J Biol Chem 2017; 293:2183-2194. [PMID: 29273634 DOI: 10.1074/jbc.m117.788430] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 12/19/2017] [Indexed: 12/22/2022] Open
Abstract
Deubiquitinases are proteases with a wide functional diversity that profoundly impact multiple biological processes. Among them, the ubiquitin-specific protease 36 (USP36) has been implicated in the regulation of nucleolar activity. However, its functional relevance in vivo has not yet been fully described. Here, we report the generation of an Usp36-deficient mouse model to examine the function of this enzyme. We show that Usp36 depletion is lethal in preimplantation mouse embryos, where it blocks the transition from morula to blastocyst during embryonic development. USP36 reduces the ubiquitination levels and increases the stability of the DEAH-box RNA helicase DHX33, which is critically involved in ribosomal RNA synthesis and mRNA translation. In agreement with this finding, O-propargyl-puromycin incorporation experiments, Northern blot, and electron microscopy analyses demonstrated the role of USP36 in ribosomal RNA and protein synthesis. Finally, we show that USP36 down-regulation alters cell proliferation in human cancer cells by inducing both apoptosis and cell cycle arrest, and that reducing DHX33 levels through short hairpin RNA interference has the same effect. Collectively, these results support that Usp36 is essential for cell and organism viability because of its role in ribosomal RNA processing and protein synthesis, which is mediated, at least in part, by regulating DHX33 stability.
Collapse
Affiliation(s)
- Julia M Fraile
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006-Oviedo, Spain
| | - Diana Campos-Iglesias
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006-Oviedo, Spain
| | - Francisco Rodríguez
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006-Oviedo, Spain
| | - Aurora Astudillo
- the Servicio de Anatomía Patológica, Hospital Universitario Central de Asturias, 33006-Oviedo, Spain
| | - Roser Vilarrasa-Blasi
- the Departamento de Anatomía Patológica, Farmacología y Microbiología, Universitat de Barcelona, IDIBAPS, 08036-Barcelona, Spain
| | - Nuria Verdaguer-Dot
- the Departamento de Anatomía Patológica, Farmacología y Microbiología, Universitat de Barcelona, IDIBAPS, 08036-Barcelona, Spain
| | - Miguel A Prado
- the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Joao A Paulo
- the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Steven P Gygi
- the Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - José I Martín-Subero
- the Departamento de Anatomía Patológica, Farmacología y Microbiología, Universitat de Barcelona, IDIBAPS, 08036-Barcelona, Spain
| | - José M P Freije
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006-Oviedo, Spain, .,the Centro de Investigación Biomédica en Red de Cáncer, Spain
| | - Carlos López-Otín
- From the Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología (IUOPA), Universidad de Oviedo, 33006-Oviedo, Spain, .,the Centro de Investigación Biomédica en Red de Cáncer, Spain
| |
Collapse
|
41
|
Erol A, Acikgoz E, Guven U, Duzagac F, Turkkani A, Colcimen N, Oktem G. Ribosome biogenesis mediates antitumor activity of flavopiridol in CD44 +/CD24 - breast cancer stem cells. Oncol Lett 2017; 14:6433-6440. [PMID: 29422957 PMCID: PMC5770605 DOI: 10.3892/ol.2017.7029] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2016] [Accepted: 07/13/2017] [Indexed: 01/16/2023] Open
Abstract
Flavopiridol is a synthetically produced flavonoid that potently inhibits the proliferation of human tumor cell lines. Flavopiridol exerts strong antitumor activity via several mechanisms, including the induction of cell cycle arrest and apoptosis, and the modulation of transcriptional regulation. The aim of the present study was to determine the effect of flavopiridol on a subpopulation of cluster of differentiation (CD)44+/CD24− human breast cancer MCF7 stem cells. The CD44+/CD24− cells were isolated from the MCF7 cell line by fluorescence-activated cell sorting and treated with 100, 300, 500, 750 and 1,000 nM flavopiridol for 24, 48 and 72 h. Cell viability and proliferation assays were performed to determine the inhibitory effect of flavopiridol. Gene expression profiling was analyzed using Illumina Human HT-12 v4 Expression BeadChip microarray. According to the results, the half maximal inhibitory concentration (IC50) value of flavopiridol was 500 nM in monolayer cells. Flavopiridol induced growth inhibition and cytotoxicity in breast cancer stem cells (BCSCs) at the IC50 dose. The present study revealed several differentially regulated genes between flavopiridol-treated and untreated cells. The result of the pathway analysis revealed that flavopiridol serves an important role in translation, the ribosome biogenesis pathway, oxidative phosphorylation, the electron transport chain pathway, carbon metabolism and cell cycle. A notable result from the present study is that ribosome-associated gene expression is significantly affected by flavopiridol treatment. The data of the present study indicate that flavopiridol exhibits antitumor activity against CD44+/CD24− MCF7 BCSCs through different mechanisms, mainly by inhibiting translation and the ribosome biogenesis pathway, and could be an effective chemotherapeutic molecule to target and kill BCSCs.
Collapse
Affiliation(s)
- Ayse Erol
- Department of Medical Pharmacology, School of Medicine, Ege University, 35100 Izmir, Turkey
| | - Eda Acikgoz
- Department of Histology and Embryology, School of Medicine, Yuzuncu Yil University, 65000 Van, Turkey.,Department of Histology and Embryology, School of Medicine, TOBB University of Economics and Technology, 06560 Ankara, Turkey
| | - Ummu Guven
- Department of Stem Cell, Institute of Health Sciences, Ege University, 35100 Izmir, Turkey
| | - Fahriye Duzagac
- Department of Stem Cell, Institute of Health Sciences, Ege University, 35100 Izmir, Turkey
| | - Ayten Turkkani
- Department of Histology and Embryology, School of Medicine, TOBB University of Economics and Technology, 06560 Ankara, Turkey
| | - Nese Colcimen
- Department of Histology and Embryology, School of Medicine, Yuzuncu Yil University, 65000 Van, Turkey
| | - Gulperi Oktem
- Department of Histology and Embryology, School of Medicine, TOBB University of Economics and Technology, 06560 Ankara, Turkey.,Department of Stem Cell, Institute of Health Sciences, Ege University, 35100 Izmir, Turkey
| |
Collapse
|
42
|
Zhang YH, Huang T, Chen L, Xu Y, Hu Y, Hu LD, Cai Y, Kong X. Identifying and analyzing different cancer subtypes using RNA-seq data of blood platelets. Oncotarget 2017; 8:87494-87511. [PMID: 29152097 PMCID: PMC5675649 DOI: 10.18632/oncotarget.20903] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 08/16/2017] [Indexed: 12/11/2022] Open
Abstract
Detection and diagnosis of cancer are especially important for early prevention and effective treatments. Traditional methods of cancer detection are usually time-consuming and expensive. Liquid biopsy, a newly proposed noninvasive detection approach, can promote the accuracy and decrease the cost of detection according to a personalized expression profile. However, few studies have been performed to analyze this type of data, which can promote more effective methods for detection of different cancer subtypes. In this study, we applied some reliable machine learning algorithms to analyze data retrieved from patients who had one of six cancer subtypes (breast cancer, colorectal cancer, glioblastoma, hepatobiliary cancer, lung cancer and pancreatic cancer) as well as healthy persons. Quantitative gene expression profiles were used to encode each sample. Then, they were analyzed by the maximum relevance minimum redundancy method. Two feature lists were obtained in which genes were ranked rigorously. The incremental feature selection method was applied to the mRMR feature list to extract the optimal feature subset, which can be used in the support vector machine algorithm to determine the best performance for the detection of cancer subtypes and healthy controls. The ten-fold cross-validation for the constructed optimal classification model yielded an overall accuracy of 0.751. On the other hand, we extracted the top eighteen features (genes), including TTN, RHOH, RPS20, TRBC2, in another feature list, the MaxRel feature list, and performed a detailed analysis of them. The results indicated that these genes could be important biomarkers for discriminating different cancer subtypes and healthy controls.
Collapse
Affiliation(s)
- Yu-Hang Zhang
- Department of General Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, People's Republic of China.,Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Tao Huang
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Lei Chen
- College of Information Engineering, Shanghai Maritime University, Shanghai 201306, People's Republic of China
| | - YaoChen Xu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Yu Hu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Lan-Dian Hu
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| | - Yudong Cai
- School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Xiangyin Kong
- Institute of Health Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai 200031, People's Republic of China
| |
Collapse
|
43
|
Moraleva A, Magoulas C, Polzikov M, Hacot S, Mertani HC, Diaz JJ, Zatsepina O. Involvement of the specific nucleolar protein SURF6 in regulation of proliferation and ribosome biogenesis in mouse NIH/3T3 fibroblasts. Cell Cycle 2017; 16:1979-1991. [PMID: 28873013 DOI: 10.1080/15384101.2017.1371880] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The nucleolar proteins which link cell proliferation to ribosome biogenesis are regarded to be potentially oncogenic. Here, in order to examine the involvement of an evolutionary conserved nucleolar protein SURF6/Rrp14 in proliferation and ribosome biogenesis in mammalian cells, we established stably transfected mouse NIH/3T3 fibroblasts capable of conditional overexpression of the protein. Cell proliferation was monitored in real-time, and various cell cycle parameters were quantified based on flow cytometry, Br-dU-labeling and conventional microscopy data. We show that overexpression of SURF6 accelerates cell proliferation and promotes transition through all cell cycle phases. The most prominent SURF6 pro-proliferative effects include a significant reduction of the population doubling time, from 19.8 ± 0.7 to 16.2 ± 0.5 hours (t-test, p < 0.001), and of the length of cell division cycle, from 17.6 ± 0.6 to 14.0 ± 0.4 hours (t-test, p < 0.001). The later was due to the shortening of all cell cycle phases but the length of G1 period was reduced most, from 5.7 ± 0.4 to 3.8 ± 0.3 hours, or by ∼30%, (t-test, p < 0.05). By Northern blots and qRT-PCR, we further showed that the acceleration of cell proliferation was concomitant with an accumulation of rRNA species along both ribosomal subunit maturation pathways. It is evident, therefore, that like the yeast homologue Rrp14, mammalian SURF6 is involved in various steps of rRNA processing during ribosome biogenesis. We concluded that SURF6 is a novel positive regulator of proliferation and G1/S transition in mammals, implicating that SURF6 is a potential oncogenic protein, which can be further studied as a putative target in anti-cancer therapy.
Collapse
Affiliation(s)
- Anastasiia Moraleva
- a Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russian Federation
| | - Charalambos Magoulas
- b Centre for Investigative and Diagnostic Oncology, Department of Natural Sciences, School of Science and Technology , Middlesex University , London , United Kingdom
| | - Mikhail Polzikov
- a Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russian Federation
| | - Sabine Hacot
- c Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard , Université Claude Bernard Lyon I, Université de Lyon , Lyon , France
| | - Hichem C Mertani
- c Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard , Université Claude Bernard Lyon I, Université de Lyon , Lyon , France
| | - Jean-Jacques Diaz
- c Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard , Université Claude Bernard Lyon I, Université de Lyon , Lyon , France
| | - Olga Zatsepina
- a Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences , Moscow , Russian Federation.,c Centre de Recherche en Cancérologie de Lyon, UMR INSERM 1052-CNRS 5286, Centre Léon Bérard , Université Claude Bernard Lyon I, Université de Lyon , Lyon , France
| |
Collapse
|
44
|
Yang X, Luo MJ, Yeung ACM, Lewis PJ, Chan PKS, Ip M, Ma C. First-In-Class Inhibitor of Ribosomal RNA Synthesis with Antimicrobial Activity against Staphylococcus aureus. Biochemistry 2017; 56:5049-5052. [PMID: 28782938 DOI: 10.1021/acs.biochem.7b00349] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
We report the discovery of the first bacterial ribosomal RNA (rRNA) synthesis inhibitor that has specific antimicrobial activity against methicillin-resistant Staphylococcus aureus (MRSA). A pharmacophore model was constructed on the basis of the protein-protein interaction between essential bacterial rRNA transcription factors NusB and NusE and employed for an in silico screen to identify potential leads. One compound, (E)-2-{[(3-ethynylphenyl)imino]methyl}-4-nitrophenol (MC4), demonstrated antimicrobial activity against a panel of S. aureus strains, including MRSA, without significant toxicity to mammalian cells. MC4 resulted in a decrease in the rRNA level in bacteria, and the target specificity of MC4 was confirmed at the molecular level. Results obtained from this work validated the bacterial rRNA transcription machinery as a novel antimicrobial target. This approach may be extended to other factors in rRNA transcription, and MC4 could be applied as a chemical probe to dissect the relationship among MRSA infection, MRSA growth rate, and rRNA synthesis, in addition to its therapeutic potential.
Collapse
Affiliation(s)
- Xiao Yang
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin, Hong Kong
| | - Ming Jing Luo
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin, Hong Kong
| | - Apple C M Yeung
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin, Hong Kong
| | - Peter J Lewis
- School of Environmental and Life Sciences, University of Newcastle , Callaghan, NSW 2308, Australia
| | - Paul K S Chan
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin, Hong Kong.,Stanley Ho Centre for Emerging Infectious Diseases, The Chinese University of Hong Kong , Shatin, Hong Kong
| | - Margaret Ip
- Department of Microbiology, The Chinese University of Hong Kong, Prince of Wales Hospital , Shatin, Hong Kong
| | - Cong Ma
- Department of Applied Biology and Chemical Technology and State Key Laboratory of Chirosciences, The Hong Kong Polytechnic University , Hung Hom, Hong Kong.,The Hong Kong Polytechnic University Shenzhen Research Institute , Shenzhen, China
| |
Collapse
|
45
|
Sapio RT, Nezdyur AN, Krevetski M, Anikin L, Manna VJ, Minkovsky N, Pestov DG. Inhibition of post-transcriptional steps in ribosome biogenesis confers cytoprotection against chemotherapeutic agents in a p53-dependent manner. Sci Rep 2017; 7:9041. [PMID: 28831158 PMCID: PMC5567254 DOI: 10.1038/s41598-017-09002-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2017] [Accepted: 07/17/2017] [Indexed: 12/11/2022] Open
Abstract
The p53-mediated nucleolar stress response associated with inhibition of ribosomal RNA transcription was previously shown to potentiate killing of tumor cells. Here, we asked whether targeting of ribosome biogenesis can be used as the basis for selective p53-dependent cytoprotection of nonmalignant cells. Temporary functional inactivation of the 60S ribosome assembly factor Bop1 in a 3T3 cell model markedly increased cell recovery after exposure to camptothecin or methotrexate. This was due, at least in part, to reversible pausing of the cell cycle preventing S phase associated DNA damage. Similar cytoprotective effects were observed after transient shRNA-mediated silencing of Rps19, but not several other tested ribosomal proteins, indicating distinct cellular responses to the inhibition of different steps in ribosome biogenesis. By temporarily inactivating Bop1 function, we further demonstrate selective killing of p53-deficient cells with camptothecin while sparing isogenic p53-positive cells. Thus, combining cytotoxic treatments with inhibition of select post-transcriptional steps of ribosome biogenesis holds potential for therapeutic targeting of cells that have lost p53.
Collapse
Affiliation(s)
- Russell T Sapio
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA.,Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA
| | - Anastasiya N Nezdyur
- Department of Chemistry and Biochemistry, Rowan University, Glassboro, NJ, 08028, USA
| | - Matthew Krevetski
- Department of Biological Sciences, Rowan University, Glassboro, NJ, 08028, USA
| | - Leonid Anikin
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA.,Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA
| | - Vincent J Manna
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA.,Graduate School of Biomedical Sciences, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA
| | - Natalie Minkovsky
- Department of Biological Sciences, Rowan University, Glassboro, NJ, 08028, USA
| | - Dimitri G Pestov
- Department of Cell Biology and Neuroscience, Rowan University School of Osteopathic Medicine, Stratford, NJ, 08084, USA.
| |
Collapse
|
46
|
Yuan F, Zhang Y, Ma L, Cheng Q, Li G, Tong T. Enhanced NOLC1 promotes cell senescence and represses hepatocellular carcinoma cell proliferation by disturbing the organization of nucleolus. Aging Cell 2017; 16:726-737. [PMID: 28493459 PMCID: PMC5506443 DOI: 10.1111/acel.12602] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/20/2017] [Indexed: 01/11/2023] Open
Abstract
The nucleolus is a key organelle that is responsible for the synthesis of rRNA and assembly of ribosomal subunits, which is also the center of metabolic control because of the critical role of ribosomes in protein synthesis. Perturbations of rRNA biogenesis are closely related to cell senescence and tumor progression; however, the underlying molecular mechanisms are not well understood. Here, we report that cellular senescence‐inhibited gene (CSIG) knockdown up‐regulated NOLC1 by stabilizing the 5′UTR of NOLC1 mRNA, and elevated NOLC1 induced the retention of NOG1 in the nucleolus, which is responsible for rRNA processing. Besides, the expression of NOLC1 was negatively correlated with CSIG in the aged mouse tissue and replicative senescent 2BS cells, and the down‐regulation of NOLC1 could rescue CSIG knockdown‐induced 2BS senescence. Additionally, NOLC1 expression was decreased in human hepatocellular carcinoma (HCC) tissue, and the ectopic expression of NOLC1 repressed the proliferation of HCC cells and tumor growth in a HCC xenograft model.
Collapse
Affiliation(s)
- Fuwen Yuan
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| | - Yu Zhang
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| | - Liwei Ma
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| | - Qian Cheng
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
- Department of Hepatobilliary Surgery; Beijing, Key Surgical Basic Research Laboratory of Liver Cirrhosis and Liver Cancer; Peking University People's Hospital; Beijing 100044 China
| | - Guodong Li
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| | - Tanjun Tong
- Peking University Research Center on Aging; Department of Biochemistry and Molecular Biology; Peking University Health Science Center, Beijing, Key Laboratory of Protein Posttranslational Modifications and Cell Function; Beijing 100191 China
| |
Collapse
|
47
|
Justilien V, Lewis KC, Murray NR, Fields AP. Oncogenic Ect2 signaling regulates rRNA synthesis in NSCLC. Small GTPases 2017; 10:388-394. [PMID: 28657426 DOI: 10.1080/21541248.2017.1335274] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
The Rho GTPase family members Rac1, Cdc42 and RhoA play key contributory roles in the transformed phenotype of human cancers. Epithelial Cell Transforming Sequence 2 (Ect2), a guanine nucleotide exchange factor (GEF) for these Rho GTPases, has also been implicated in a variety of human cancers. We have shown that Ect2 is frequently overexpressed in both major forms of non-small cell lung cancer (NSCLC), lung adenocarcinoma (LADC) and lung squamous cell carcinoma (LSCC), which together make up approximately 70% of all lung cancer diagnoses. Furthermore, we have found that Ect2 is required for multiple aspects of the transformed phenotype of NSCLC cells including transformed growth and invasion in vitro and tumorigenesis in vivo. More recently, we showed that a major mechanism by which Ect2 drives KRAS-mediated LADC transformation is by regulating rRNA (rRNA) synthesis. However, it remains unclear whether Ect2 plays a similar role in ribosome biogenesis in LSCC. Here we demonstrate that Ect2 expression correlates positively with expression of ribosome biogenesis genes and with pre-ribosomal 45S RNA abundance in primary LSCC tumors. Furthermore, we demonstrate that Ect2 functionally regulates rRNA synthesis in LSCC cells. Based on these data, we propose that inhibition of Ect2-mediated nucleolar signaling holds promise as a potential therapeutic strategy for improved treatment of both LADC and LSCC.
Collapse
Affiliation(s)
- Verline Justilien
- a Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center , Jacksonville , FL , USA
| | - Kayla C Lewis
- a Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center , Jacksonville , FL , USA
| | - Nicole R Murray
- a Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center , Jacksonville , FL , USA
| | - Alan P Fields
- a Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center , Jacksonville , FL , USA
| |
Collapse
|
48
|
Principles of 60S ribosomal subunit assembly emerging from recent studies in yeast. Biochem J 2017; 474:195-214. [PMID: 28062837 DOI: 10.1042/bcj20160516] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 11/22/2016] [Accepted: 11/24/2016] [Indexed: 12/31/2022]
Abstract
Ribosome biogenesis requires the intertwined processes of folding, modification, and processing of ribosomal RNA, together with binding of ribosomal proteins. In eukaryotic cells, ribosome assembly begins in the nucleolus, continues in the nucleoplasm, and is not completed until after nascent particles are exported to the cytoplasm. The efficiency and fidelity of ribosome biogenesis are facilitated by >200 assembly factors and ∼76 different small nucleolar RNAs. The pathway is driven forward by numerous remodeling events to rearrange the ribonucleoprotein architecture of pre-ribosomes. Here, we describe principles of ribosome assembly that have emerged from recent studies of biogenesis of the large ribosomal subunit in the yeast Saccharomyces cerevisiae We describe tools that have empowered investigations of ribosome biogenesis, and then summarize recent discoveries about each of the consecutive steps of subunit assembly.
Collapse
|
49
|
Diwakarla C, Hannan K, Hein N, Yip D. Advanced pancreatic ductal adenocarcinoma - Complexities of treatment and emerging therapeutic options. World J Gastroenterol 2017; 23:2276-2285. [PMID: 28428707 PMCID: PMC5385394 DOI: 10.3748/wjg.v23.i13.2276] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Revised: 01/18/2017] [Accepted: 03/15/2017] [Indexed: 02/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma is a devastating disease with a poor prognosis regardless of stage. To date the mainstay of therapy for advanced disease has been chemotherapy with little incremental improvements in outcome. Despite extensive research investigating new treatment options the current practices continue to utilise fluorouracil or gemcitabine containing combinations. The need for novel therapeutic approaches is mandated by the ongoing poor survival rates associated with this disease. One such approach may include manipulation of ribosome biogenesis and the nucleolar stress response, which has recently been applied to haematological malignancies such as lymphoma and prostate cancer with promising results. This review will focus on the current therapeutic options for pancreatic ductal adenocarcinoma and the complexities associated with developing novel treatments, with a particular emphasis on the role of the nucleolus as a treatment strategy.
Collapse
|
50
|
Derenzini M, Montanaro L, Trerè D. Ribosome biogenesis and cancer. Acta Histochem 2017; 119:190-197. [PMID: 28168996 DOI: 10.1016/j.acthis.2017.01.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2017] [Accepted: 01/27/2017] [Indexed: 12/21/2022]
Abstract
There is growing evidence indicating that the human pathological conditions characterized by an up-regulated ribosome biogenesis are at an increased risk of cancer onset. At the basis of this relationship is the close interconnection between the ribosome biogenesis and cell proliferation. Cell proliferation-stimulating factors also stimulate ribosome production, while the ribosome biogenesis rate controls the cell cycle progression. The major tumour suppressor, the p53 protein, plays an important balancing role between the ribosome biogenesis rate and the cell progression through the cell cycle phases. The perturbation of ribosome biogenesis stabilizes and activates p53, with a consequent cell cycle arrest and/or apoptotic cell death, whereas an up-regulated ribosome production down-regulates p53 expression and activity, thus facilitating neoplastic transformation. In the present review we describe the interconnection between ribosome biogenesis and cell proliferation, while highlighting the mechanisms by which quantitative changes in ribosome biogenesis may induce cancer.
Collapse
Affiliation(s)
- Massimo Derenzini
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40138, Italy.
| | - Lorenzo Montanaro
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40138, Italy.
| | - Davide Trerè
- Department of Experimental, Diagnostic and Specialty Medicine, University of Bologna, Bologna 40138, Italy.
| |
Collapse
|