1
|
Phelps PE, Ha SM, Khankan RR, Mekonnen MA, Juarez G, Ingraham Dixie KL, Chen YW, Yang X. Olfactory ensheathing cells from adult female rats are hybrid glia that promote neural repair. eLife 2025; 13:RP95629. [PMID: 40297980 PMCID: PMC12040321 DOI: 10.7554/elife.95629] [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] [Indexed: 04/30/2025] Open
Abstract
Olfactory ensheathing cells (OECs) are unique glial cells found in both central and peripheral nervous systems where they support continuous axonal outgrowth of olfactory sensory neurons to their targets. Previously, we reported that following severe spinal cord injury, OECs transplanted near the injury site modify the inhibitory glial scar and facilitate axon regeneration past the scar border and into the lesion. To better understand the mechanisms underlying the reparative properties of OECs, we used single-cell RNA-sequencing of OECs from adult rats to study their gene expression programs. Our analyses revealed five diverse OEC subtypes, each expressing novel marker genes and pathways indicative of progenitor, axonal regeneration, secreted molecules, or microglia-like functions. We found substantial overlap of OEC genes with those of Schwann cells, but also with microglia, astrocytes, and oligodendrocytes. We confirmed established markers on cultured OECs, and localized select top genes of OEC subtypes in olfactory bulb tissue. We also show that OECs secrete Reelin and Connective tissue growth factor, extracellular matrix molecules which are important for neural repair and axonal outgrowth. Our results support that OECs are a unique hybrid glia, some with progenitor characteristics, and that their gene expression patterns indicate functions related to wound healing, injury repair, and axonal regeneration.
Collapse
Affiliation(s)
- Patricia E Phelps
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Sung Min Ha
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Rana R Khankan
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Mahlet A Mekonnen
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Giovanni Juarez
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | | | - Yen-Wei Chen
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| | - Xia Yang
- Department of Integrative Biology and Physiology, UCLALos AngelesUnited States
| |
Collapse
|
2
|
Guérin TM, Uhlmann F. Cohesinopathies: Is enhancer disruption to blame? Curr Biol 2025; 35:R214-R216. [PMID: 40132551 DOI: 10.1016/j.cub.2025.01.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/27/2025]
Abstract
Cohesinopathies are debilitating human developmental disorders. New research pins down mechanisms by which the chromosomal cohesin complex controls the intricate transcriptional program that underlies healthy human growth and development.
Collapse
Affiliation(s)
- Thomas M Guérin
- Université Paris Cité and Université Paris-Saclay, CEA, INSERM, 92260 Fontenay-aux-Roses, France
| | - Frank Uhlmann
- Chromosome Segregation Laboratory, The Francis Crick Institute, London NW1 1AT, UK.
| |
Collapse
|
3
|
Otero AM, Connolly MG, Gonzalez-Ricon RJ, Wang SS, Allen JM, Antonson AM. Influenza A virus during pregnancy disrupts maternal intestinal immunity and fetal cortical development in a dose- and time-dependent manner. Mol Psychiatry 2025; 30:13-28. [PMID: 38961232 PMCID: PMC11649561 DOI: 10.1038/s41380-024-02648-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 06/19/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024]
Abstract
Epidemiological studies link exposure to viral infection during pregnancy, including influenza A virus (IAV) infection, with increased incidence of neurodevelopmental disorders (NDDs) in offspring. Models of maternal immune activation (MIA) using viral mimetics demonstrate that activation of maternal intestinal T helper 17 (TH17) cells, which produce effector cytokine interleukin (IL)-17, leads to aberrant fetal brain development, such as neocortical malformations. Fetal microglia and border-associated macrophages (BAMs) also serve as potential cellular mediators of MIA-induced cortical abnormalities. However, neither the inflammation-induced TH17 cell pathway nor fetal brain-resident macrophages have been thoroughly examined in models of live viral infection during pregnancy. Here, we inoculated pregnant mice with two infectious doses of IAV and evaluated peak innate and adaptive immune responses in the dam and fetus. While respiratory IAV infection led to dose-dependent maternal colonic shortening and microbial dysregulation, there was no elevation in intestinal TH17 cells nor IL-17. Systemically, IAV resulted in consistent dose- and time-dependent increases in IL-6 and IFN-γ. Fetal cortical abnormalities and global changes in fetal brain transcripts were observable in the high-but not the moderate-dose IAV group. Profiling of fetal microglia and BAMs revealed dose- and time-dependent differences in the numbers of meningeal but not choroid plexus BAMs, while microglial numbers and proliferative capacity of Iba1+ cells remained constant. Fetal brain-resident macrophages increased phagocytic CD68 expression, also in a dose- and time-dependent fashion. Taken together, our findings indicate that certain features of MIA are conserved between mimetic and live virus models, while others are not. Overall, we provide consistent evidence of an infection severity threshold for downstream maternal inflammation and fetal cortical abnormalities, which recapitulates a key feature of the epidemiological data and further underscores the importance of using live pathogens in NDD modeling to better evaluate the complete immune response and to improve translation to the clinic.
Collapse
Affiliation(s)
- Ashley M Otero
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Meghan G Connolly
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | | | - Selena S Wang
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jacob M Allen
- Department of Kinesiology and Community Health, University of Illinois Urbana-Champaign, Urbana, IL, USA
| | - Adrienne M Antonson
- Neuroscience Program, University of Illinois Urbana-Champaign, Urbana, IL, USA.
- Department of Animal Sciences, University of Illinois Urbana-Champaign, Urbana, IL, USA.
| |
Collapse
|
4
|
Garikapati K, Young IC, Hong S, Rai P, Jain C, Briegel KJ. Blocking LBH expression causes replication stress and sensitizes triple-negative breast cancer cells to ATR inhibitor treatment. Oncogene 2024; 43:851-865. [PMID: 38297083 DOI: 10.1038/s41388-024-02951-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 01/16/2024] [Accepted: 01/18/2024] [Indexed: 02/02/2024]
Abstract
Triple-negative (ER-PR-HER2-) breast cancers (TNBC) are highly aggressive and difficult to treat. TNBC exhibit high genomic instability, which enables them to adapt and become resistant to chemo/radiation therapy, leading to rapid disease relapse and mortality. The pro-survival factors that safeguard genome integrity in TNBC cells are poorly understood. LBH is an essential mammary stem cell-specific transcription regulator in the WNT pathway that is aberrantly overexpressed in TNBC, correlating with poor prognosis. Herein, we demonstrate a novel role for LBH in promoting TNBC cell survival. Depletion of LBH in multiple TNBC cell models triggered apoptotic cell death both in vitro and in vivo and led to S-G2M cell cycle delays. Mechanistically, LBH loss causes replication stress due to DNA replication fork stalling, leading to ssDNA breaks, ɣH2AX and 53BP1 nuclear foci formation, and activation of the ATR/CHK1 DNA damage response. Notably, ATR inhibition in combination with LBH downmodulation had a synergistic effect, boosting TNBC cell killing and blocking in vivo tumor growth. Our findings demonstrate, for the first time, that LBH protects the genome integrity of cancer cells by preventing replicative stress. Importantly, they uncover new synthetic lethal vulnerabilities in TNBC that could be exploited for future multi-modal precision medicine.
Collapse
Affiliation(s)
- Koteswararao Garikapati
- DeWitt Daugherty Department of Surgery, Molecular Oncology Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - In-Chi Young
- DeWitt Daugherty Department of Surgery, Molecular Oncology Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Sunhwa Hong
- DeWitt Daugherty Department of Surgery, Molecular Oncology Program, University of Miami Miller School of Medicine, Miami, FL, USA
- Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Priyamvada Rai
- Department of Radiation Oncology and Tumor Biology Program at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Chaitanya Jain
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Karoline J Briegel
- DeWitt Daugherty Department of Surgery, Molecular Oncology Program, University of Miami Miller School of Medicine, Miami, FL, USA.
- Braman Family Breast Cancer Institute at the Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
5
|
Qiu D, Cai H, Liang J, Wang Z, Song F, Jiang Y, Tan R, Hou J. Identification of CKS2 as a novel prognostic biomarker and potential therapeutic target for oral squamous cell carcinoma. Transl Cancer Res 2023; 12:2276-2293. [PMID: 37859732 PMCID: PMC10583009 DOI: 10.21037/tcr-23-511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 08/15/2023] [Indexed: 10/21/2023]
Abstract
Background The cyclin-dependent kinase subunit 2 (CKS2) is recognized to have a substantial impact on the pathogenesis and advancement of several malignant neoplasms. Nevertheless, its biological function and prognostic significance in oral squamous cell carcinoma (OSCC) have yet to be thoroughly investigated. Our primary objective was to clarify the contribution of CKS2 in the progression and prognosis of OSCC. Methods We first conducted a thorough examination of online databases to investigate the expression of CKS2, and subsequently corroborated our discoveries by analyzing clinical specimens that we collected. According to the clinicopathological data, we then explored the prognostic significance of CKS2. Furthermore, we predicted the role of CKS2 in OSCC progression by employing weighted gene co-expression network analysis (WGCNA) in conjunction with functional enrichment analysis. We conducted functional experiments in vitro to confirm our speculations. Additionally, we explored other potential functions of CKS2 in immune infiltration, tumor mutation burden (TMB), and drug sensitivity. Finally, we established and validated a nomogram that effectively integrated CKS2-related genes and other relevant clinical factors. Results Our findings indicated a significant upregulation of CKS2 expression in OSCC tissues compared to normal groups, which was positively associated with poor clinical outcomes. We also predicted and validated the role of CKS2 in promoting proliferation by regulating the cell cycle. Additionally, its upregulation was significantly correlated to enhanced immune cell infiltration, high TMB, and increased sensitivity of anti-tumor agents. Following verification, the nomogram was conducted to quantify an individual's survival probability. Conclusions In general, our study indicates that CKS2 is a novel prognostic biomarker and potential therapeutic target in OSCC.
Collapse
Affiliation(s)
- Danqi Qiu
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Hongshi Cai
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jianfeng Liang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ziyi Wang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Fan Song
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yaoqi Jiang
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Rukeng Tan
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jingsong Hou
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
6
|
Cabana-Domínguez J, Llonga N, Arribas L, Alemany S, Vilar-Ribó L, Demontis D, Fadeuilhe C, Corrales M, Richarte V, Børglum AD, Ramos-Quiroga JA, Soler Artigas M, Ribasés M. Transcriptomic risk scores for attention deficit/hyperactivity disorder. Mol Psychiatry 2023; 28:3493-3502. [PMID: 37537283 PMCID: PMC10618083 DOI: 10.1038/s41380-023-02200-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/17/2023] [Accepted: 07/21/2023] [Indexed: 08/05/2023]
Abstract
Attention deficit/hyperactivity disorder (ADHD) is a highly heritable neurodevelopmental disorder. We performed a transcriptome-wide association study (TWAS) using the latest genome-wide association study (GWAS) meta-analysis, in 38,691 individuals with ADHD and 186,843 controls, and 14 gene-expression reference panels across multiple brain tissues and whole blood. Based on TWAS results, we selected subsets of genes and constructed transcriptomic risk scores (TRSs) for the disorder in peripheral blood mononuclear cells of individuals with ADHD and controls. We found evidence of association between ADHD and TRSs constructed using expression profiles from multiple brain areas, with individuals with ADHD carrying a higher burden of TRSs than controls. TRSs were uncorrelated with the polygenic risk score (PRS) for ADHD and, in combination with PRS, improved significantly the proportion of variance explained over the PRS-only model. These results support the complementary predictive potential of genetic and transcriptomic profiles in blood and underscore the potential utility of gene expression for risk prediction and deeper insight in molecular mechanisms underlying ADHD.
Collapse
Affiliation(s)
- Judit Cabana-Domínguez
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain.
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
| | - Natalia Llonga
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Lorena Arribas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Silvia Alemany
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | - Laura Vilar-Ribó
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
| | - Ditte Demontis
- Department of Biomedicine/Human Genetics, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
- The Novo Nordisk Foundation Center for Genomic Mechanisms of Disease, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Christian Fadeuilhe
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Montse Corrales
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Vanesa Richarte
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Anders D Børglum
- Department of Biomedicine/Human Genetics, Aarhus University, Aarhus, Denmark
- The Lundbeck Foundation Initiative for Integrative Psychiatric Research, iPSYCH, Aarhus, Denmark
- Center for Genomics and Personalized Medicine, Aarhus, Denmark
| | - Josep Antoni Ramos-Quiroga
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain
- Department of Psychiatry and Forensic Medicine, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - María Soler Artigas
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain.
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
| | - Marta Ribasés
- Psychiatric Genetics Unit, Group of Psychiatry, Mental Health and Addiction, Vall d'Hebron Research Institute (VHIR), Universitat Autònoma de Barcelona, Barcelona, Spain.
- Department of Mental Health, Hospital Universitari Vall d'Hebron, Barcelona, Spain.
- Biomedical Network Research Centre on Mental Health (CIBERSAM), Madrid, Spain.
- Department of Genetics, Microbiology, and Statistics, Faculty of Biology, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
7
|
Wan D, He L, Guo C, Zhong Z, Yan X, Cao J, Xu Q, Zhang H, Duan B. m6A-related lncRNAs predict prognosis and indicate cell cycle in gastric cancer. Front Genet 2023; 14:1140218. [PMID: 37408779 PMCID: PMC10319253 DOI: 10.3389/fgene.2023.1140218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2023] [Accepted: 04/11/2023] [Indexed: 07/07/2023] Open
Abstract
Background: N6-methyladenosine (m6A) modification is a common epigenetic methylation modification of RNA, which plays an important role in gastric carcinogenesis and progression by regulating long non-coding RNA (lncRNA). This study is aimed to investigate the potential prognostic signatures of m6A -related lncRNAs in STAD. Methods: The m6A-related lncRNAs with the most significant impact on gastric cancer prognosis in the TCGA database were identified by bioinformatics and machine learning methods. The m6A-related lncRNA prognostic model (m6A-LPS) and nomogram was constructed by Cox regression analysis with the minimum absolute contraction and selection operator (LASSO) algorithm. The functional enrichment analysis of m6A-related lncRNAs was also investigated. The miRTarBase, miRDB and TargetScan databases were utilized to establish a prognosis-related network of competing endogenous RNA (ceRNA) by bioinformatics methods. The correlation of AL391152.1 expressions and cell cycle were experimentally testified by qRT-PCR and flow cytometry. Results: In total, 697 lncRNAs that were identified as m6A-related lncRNAs in GC samples. The survival analysis showed that 18 lncRNAs demonstrated prognostic values. A risk model with 11 lncRNAs was established by Lasso Cox regression, and can predict the prognosis of GC patients. Cox regression analysis and ROC curve indicated that this lncRNA prediction model was an independent risk factor for survival rates. Functional enrichment analysis and ceRNA network revealed that the nomogram was notably associated with cell cycle. qRT-PCR and flow cytometry revealed that downregulation of GC m6A-related lncRNA AL391152.1 could decrease cyclins expression in SGC7901 cells. Conclusion: A m6A-related lncRNAs prognostic model was established in this study, which can be applied to predict prognosis and cell cycle in gastric cancer.
Collapse
Affiliation(s)
- Dong Wan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Lingnan He
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Cheng Guo
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zishao Zhong
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Xiaohan Yan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Jia Cao
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qinwei Xu
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haibin Zhang
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Bensong Duan
- Endoscopy Center, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
8
|
Grey W, Atkinson S, Rix B, Casado P, Ariza-McNaughton L, Hawley C, Sopoena ML, Bridge KS, Kent D, Cutillas PR, Bonnet D. The CKS1/CKS2 Proteostasis Axis Is Crucial to Maintain Hematopoietic Stem Cell Function. Hemasphere 2023; 7:e853. [PMID: 36874381 PMCID: PMC9977483 DOI: 10.1097/hs9.0000000000000853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Accepted: 01/30/2023] [Indexed: 03/04/2023] Open
Abstract
Long-term hematopoietic stem cells are rare, highly quiescent stem cells of the hematopoietic system with life-long self-renewal potential and the ability to transplant and reconstitute the entire hematopoietic system of conditioned recipients. Most of our understanding of these rare cells has relied on cell surface identification, epigenetic, and transcriptomic analyses. Our knowledge of protein synthesis, folding, modification, and degradation-broadly termed protein homeostasis or "proteostasis"-in these cells is still in its infancy, with very little known about how the functional state of the proteome is maintained in hematopoietic stem cells. We investigated the requirement of the small phospho-binding adaptor proteins, the cyclin-dependent kinase subunits (CKS1 and CKS2), for maintaining ordered hematopoiesis and long-term hematopoietic stem cell reconstitution. CKS1 and CKS2 are best known for their roles in p27 degradation and cell cycle regulation, and by studying the transcriptome and proteome of Cks1 -/- and Cks2 -/- mice, we demonstrate regulation of key signaling pathways that govern hematopoietic stem cell biology including AKT, FOXO1, and NFκB, together balancing protein homeostasis and restraining reactive oxygen species to ensure healthy hematopoietic stem cell function.
Collapse
Affiliation(s)
- William Grey
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Samantha Atkinson
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| | - Beatrice Rix
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Pedro Casado
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Bart’s Cancer Institute, London, United Kingdom
| | | | - Cathy Hawley
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Miriam L. Sopoena
- Bioinformatics Core, The Francis Crick Institute, London, United Kingdom
| | - Katherine S. Bridge
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - David Kent
- York Biomedical Research Institute, Department of Biology, University of York, United Kingdom
| | - Pedro R. Cutillas
- Cell Signalling and Proteomics Group, Centre for Genomics and Computational Biology, Bart’s Cancer Institute, London, United Kingdom
| | - Dominique Bonnet
- Hematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, United Kingdom
| |
Collapse
|
9
|
Grey W, Rio-Machin A, Casado-Izquierdo P, Grönroos E, Ali S, Miettinen JJ, Bewicke-Copley F, Parsons A, Heckman CA, Swanton C, Cutillas P, Gribben J, Fitzgibbon J, Bonnet D. CKS1 inhibition depletes leukemic stem cells and protects healthy hematopoietic stem cells in acute myeloid leukemia. Sci Transl Med 2022; 14:eabn3248. [PMID: 35731890 PMCID: PMC7612983 DOI: 10.1126/scitranslmed.abn3248] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Acute myeloid leukemia (AML) is an aggressive hematological disorder comprising a hierarchy of quiescent leukemic stem cells (LSCs) and proliferating blasts with limited self-renewal ability. AML has a dismal prognosis, with extremely low 2-year survival rates in the poorest cytogenetic risk patients, primarily due to the failure of intensive chemotherapy protocols to deplete LSCs and toxicity of therapy toward healthy hematopoietic cells. We studied the role of cyclin-dependent kinase regulatory subunit 1 (CKS1)-dependent protein degradation in primary human AML and healthy hematopoiesis xenograft models in vivo. Using a small-molecule inhibitor (CKS1i), we demonstrate a dual role for CKS1-dependent protein degradation in reducing patient-derived AML blasts in vivo and, importantly, depleting LSCs, whereas inhibition of CKS1 has the opposite effect on normal hematopoiesis, protecting normal hematopoietic stem cells from chemotherapeutic toxicity. Proteomic analysis of responses to CKS1i in our patient-derived xenograft mouse model demonstrate that inhibition of CKS1 in AML leads to hyperactivation of RAC1 and accumulation of lethal reactive oxygen species, whereas healthy hematopoietic cells enter quiescence in response to CKS1i, protecting hematopoietic stem cells. Together, these findings demonstrate that CKS1-dependent proteostasis is a key vulnerability in malignant stem cell biology.
Collapse
Affiliation(s)
- William Grey
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, U.K
| | - Ana Rio-Machin
- Centre for Genomics and Computational Biology, Bart’s Cancer Institute, London, U.K
| | - Pedro Casado-Izquierdo
- Cell signalling and proteomics group, Centre for Genomics and Computational Biology, Barts Cancer Institute, London, U.K
| | - Eva Grönroos
- Cancer evolution and genome instability laboratory, The Francis Crick Institute, London, U.K
| | - Sara Ali
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, U.K
| | - Juho J. Miettinen
- Institute for Molecular Medicine Finland – FINN, HiLIFE – Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | | | - Alun Parsons
- Institute for Molecular Medicine Finland – FINN, HiLIFE – Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Caroline A. Heckman
- Institute for Molecular Medicine Finland – FINN, HiLIFE – Helsinki Institute of Life Science, iCAN Digital Precision Cancer Medicine Flagship, University of Helsinki, Helsinki, Finland
| | - Charles Swanton
- Cancer evolution and genome instability laboratory, The Francis Crick Institute, London, U.K
| | - Pedro Cutillas
- Cell signalling and proteomics group, Centre for Genomics and Computational Biology, Barts Cancer Institute, London, U.K
| | - John Gribben
- Centre for Haemato-Oncology, Bart’s Cancer Institute, London, U.K
| | - Jude Fitzgibbon
- Centre for Genomics and Computational Biology, Bart’s Cancer Institute, London, U.K
| | - Dominique Bonnet
- Haematopoietic Stem Cell Laboratory, The Francis Crick Institute, London, U.K
| |
Collapse
|
10
|
Yang Y, Wu J, Yu X, Wu Q, Cao H, Dai X, Chen H. SLC34A2 promotes cancer proliferation and cell cycle progression by targeting TMPRSS3 in colorectal cancer. Pathol Res Pract 2021; 229:153706. [PMID: 34929599 DOI: 10.1016/j.prp.2021.153706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 09/26/2021] [Accepted: 11/20/2021] [Indexed: 10/19/2022]
Abstract
Solute carrier family 34 member 2 (SLC34A2), a family member of sodium-driven phosphate cotransporters, has been reported to facilitate cell proliferation and tumor growth. However, the functional mechanism by which SLC34A2 promotes cell growth and cell cycle progression remains poorly understood. Here, we reported that SLC34A2 was overexpressed in CRC by analysis of TCGA and GEO datasets. A total of 45 differentially expressed genes (DEGs) were identified from comparing SLC34A2-high or -low groups and functional enrichment analysis of these DEGs demonstrated that cell cycle pathway was enriched. Interestingly, we found a positive correlation between TMPRSS3 (transmembrane serine protease 3) and SLC34A2, which was confirmed by RT-qPCR and western blotting. Furthermore, TMPRSS3 was also upregulated in CRC tumor tissues compared to normal tissues. Patients with high TMPRSS3 expression had poor prognosis. Functionally, TMPRSS3 deficiency inhibited cell proliferation and colony formation in CRC cells. TMPRSS3 overexpression in SLC34A2-deficient cells antagonized siSLC34A2-mediated cell cycle inhibition by promoting cyclin E, cyclin A protein expression. Based on these results, our study suggests that SLC34A2 promotes cancer proliferation and cell cycle progression by targeting TMPRSS3 in colorectal cancer cells.
Collapse
Affiliation(s)
- Yi Yang
- Department of Oncological Surgery, Kunshan Traditional Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan City, Jiangsu Province 215300, China
| | - Jiang Wu
- Department of Oncological Surgery, Kunshan Traditional Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan City, Jiangsu Province 215300, China
| | - Xiaofeng Yu
- Department of Oncological Surgery, Kunshan Traditional Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan City, Jiangsu Province 215300, China
| | - Qing Wu
- Department of Oncological Surgery, Kunshan Traditional Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan City, Jiangsu Province 215300, China
| | - Huihua Cao
- Department of Oncological Surgery, Kunshan Traditional Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan City, Jiangsu Province 215300, China
| | - Xinyi Dai
- Department of Spleen and Stomach Disease Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing City, Jiangsu Province 210092, China
| | - Haijun Chen
- Department of Oncological Surgery, Kunshan Traditional Hospital Affiliated to Nanjing University of Chinese Medicine, Kunshan City, Jiangsu Province 215300, China.
| |
Collapse
|
11
|
Hwang DJ, Choi DH, Kwon KC, Kim EH, Kim TK, Koo JH, Cho JY. Exercise Reverses Amyloid Beta-Peptide-mediated Cognitive Deficits in Alzheimer's Disease Mice Expressing Mutant Presenilin-2. Med Sci Sports Exerc 2021; 54:551-565. [PMID: 34816813 DOI: 10.1249/mss.0000000000002834] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
PURPOSE The molecular mechanisms by which physical exercise produces beneficial effects on pathological features and behavioral symptoms of Alzheimer's disease (AD) are not well understood. Herein, we examined whether regular moderate exercise could improve cognitive function and produce transcriptomic responses in the brain. METHODS Four groups of mice were studied: non-transgenic control (Non-Tg), mice expressing the human presenilin-2 wild type (Tg-PS2w), mice expressing the human presenilin-2 with the N141I mutation (Tg-PS2m), and Tg-PS2m that were subjected to treadmill exercise (TE) at a speed of 10 m/min for 50 min/day, 5 days/week, for 6 weeks (Tg-PS2m/Ex). RESULTS Tg-PS2m/Ex mice exhibited increased preference in exploring a novel object than Tg-PS2m in the novel object recognition test (NORT), whereas differences observed in the water maze test and passive avoidance test were not significant. Western blot and histological analyses using amyloid oligomer (A11) and Aβ (6E10) antibody indicated that amyloid oligomer-reactive bands and plaque deposition in the hippocampus were reduced, though not significantly, after TE. Transcriptomic (RNA-sequencing) analysis and subsequent protein analysis revealed that the cell cycle regulatory gene, Cdc28 protein kinase regulatory subunit 2 (Cks2), was decreased, and the cell cycle- and apoptotic cell death-related factors, including cyclin D1, proliferating cell nuclear antigen, and cleaved caspase-3 were increased in the hippocampus of Tg-PS2m, whereas TE reversed their altered expression. CONCLUSION These results support the hypothesis that the pathological features and behavioral symptoms of AD caused by accumulation of amyloid beta-peptide in hippocampus, causing aberrant cell cycle re-entry and apoptosis, can be reversed by regular exercise.
Collapse
Affiliation(s)
- Dong-Joo Hwang
- Exercise Biochemistry Laboratory, Korea National Sport University, Seoul, Republic of Korea Department of Physical Education, Dongguk University, Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
12
|
Wan Z, Wang L, Yang D, Li P, Liu Q, Wang B. CKS2 Promotes the Growth in Non-Small-Cell Lung Cancer by Downregulating Cyclin-Dependent Kinase Inhibitor. Pathobiology 2021; 89:13-22. [PMID: 34333494 DOI: 10.1159/000517755] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/07/2021] [Indexed: 12/24/2022] Open
Abstract
INTRODUCTION/OBJECTIVE This study aimed to explore the expression of cyclin-dependent kinase subunit 2 (CKS2) in tissues and cells in non-small-cell lung cancer (NSCLC) and the function mechanism of CKS2 in NSCLC cell growth and tumorigensis. METHODS After transfecting NCI-H2170 cells with short-hair RNA (shRNA), an shCKS2 gene-silencing model was established. The cells were divided into a shRNA group and shNC group. For overexpression cell lines, we used the same method to establish the NCI-H2170-CKS2 cell lines. Cell Count Kit-8 assay and colony formation assay were used to determine cell viability and cell growth, respectively. Propidium iodide staining was used to determine cell cycle progression. The mRNA expression of CKS2 and protein expression of CKS2, p21, p53, and PTEN were determined by RT-qPCR and Western blotting, respectively. The expression of CKS2, p53, and Ki67 in tissues was determined by immunohistochemical stain. The in vivo tumorigenesis assays were used to determine the ability of CKS2 in tumor growth. RESULTS The results of RT-qPCR and Western blotting assay revealed that CKS2 upregulated expression in NSCLC tissues and cells. The results of the CCK-8 assay revealed that the shRNA group exhibited significantly lower cell viability and foci formation than the empty plasmid group, while CKS2 overexpression induces cell growth and cell cycle progression. The result of nude mice suggested that CKS2 knockdown expression suppressed tumorigenesis in the in vivo animal model. CONCLUSIONS Our study suggests that CKS2 could be a biomarker in the progression and prognosis of NSCLC.
Collapse
Affiliation(s)
- Zongren Wan
- Department of Pulmonary and Critical Care Medicine, Huai'an First People's Hospital, Huai'an, China
| | - Lixin Wang
- Department of Pulmonary and Critical Care Medicine, Huai'an First People's Hospital, Huai'an, China
| | - Dan Yang
- Department of Pulmonary and Critical Care Medicine, Huai'an First People's Hospital, Huai'an, China
| | - Pengling Li
- Department of Pulmonary and Critical Care Medicine, Huai'an First People's Hospital, Huai'an, China
| | - Qing Liu
- Department of Pulmonary and Critical Care Medicine, Huai'an First People's Hospital, Huai'an, China
| | - Baolan Wang
- Department of Pulmonary and Critical Care Medicine, Huai'an First People's Hospital, Huai'an, China
| |
Collapse
|
13
|
Zhang W, Wang R, Giesy JP, Zhang S, Wei S, Wang P. Proteomic analysis using isobaric tags for relative and absolute quantification technology reveals mechanisms of toxic effects of tris (1,3-dichloro-2-propyl) phosphate on RAW264.7 macrophage cells. J Appl Toxicol 2021; 42:190-202. [PMID: 34036598 DOI: 10.1002/jat.4201] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 05/08/2021] [Accepted: 05/10/2021] [Indexed: 01/29/2023]
Abstract
Tris (1,3-dichloro-2-propyl) phosphate (TDCIPP) is one of the most commonly used organophosphorus flame retardants. Immuno-toxicity induced by TDCIPP is becoming of increasing concern. However, effects of TDCIPP on immune cells and mechanisms resulting in those effects are poorly understood. In this study, it was determined, for the first time, by use of isobaric tags for relative and absolute quantification (iTRAQ) based proteomic techniques expression of global proteins in RAW264.7 cells exposed to 10 μM TDCIPP. A total of 180 significantly differentially expressed proteins (DEPs) were identified. Of these, 127 were up-regulated and 53 were down-regulated. The DEPs associated with toxic effects of TDCIPP were then screened by use of Gene Ontology and the Kyoto Encyclopedia of Genes and Genomes for enrichment analysis. Results showed that these DEPs were involved in a number of pathways including apoptosis, DNA damage, cell cycle arrest, immune-toxicity, and signaling pathways, such as the Toll-like receptor, PPAR and p53 signaling pathways. The complex regulatory relationships between different DEPs, which might play an important role in cell death were also observed in the form of a protein-protein interaction network. Meanwhile, mitochondrial membrane potential (MMP) in RAW264.7 cells after TDCIPP treatment was also analyzed, the collapse of the MMP was speculated to play an important role in TDCIPP induced apoptosis. Moreover, some of the important regulator proteins discovered in this study, such as Chk1, Aurora A, would provide novel insight into the molecular mechanisms involved in toxic responses to TDCIPP.
Collapse
Affiliation(s)
- Wei Zhang
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Ruiguo Wang
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - John P Giesy
- Department of Veterinary Biomedical Sciences and Toxicology Centre, University of Saskatchewan, Saskatoon, Saskatchewan, Canada.,Department of Zoology and Center for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA.,Department of Environmental Sciences, Baylor University, Waco, Texas, USA.,State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing, China
| | - Su Zhang
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Shulin Wei
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Peilong Wang
- Institute of Quality Standard and Testing Technology for Agro-Products, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
14
|
Bury M, Le Calvé B, Ferbeyre G, Blank V, Lessard F. New Insights into CDK Regulators: Novel Opportunities for Cancer Therapy. Trends Cell Biol 2021; 31:331-344. [PMID: 33676803 DOI: 10.1016/j.tcb.2021.01.010] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 01/28/2021] [Accepted: 01/29/2021] [Indexed: 12/13/2022]
Abstract
Cyclins and their catalytic partners, the cyclin-dependent kinases (CDKs), control the transition between different phases of the cell cycle. CDK/cyclin activity is regulated by CDK inhibitors (CKIs), currently comprising the CDK-interacting protein/kinase inhibitory protein (CIP/KIP) family and the inhibitor of kinase (INK) family. Recent studies have identified a third group of CKIs, called ribosomal protein-inhibiting CDKs (RPICs). RPICs were discovered in the context of cellular senescence, a stable cell cycle arrest with tumor-suppressing abilities. RPICs accumulate in the nonribosomal fraction of senescent cells due to a decrease in rRNA biogenesis. Accordingly, RPICs are often downregulated in human cancers together with other ribosomal proteins, the tumor-suppressor functions of which are still under study. In this review, we discuss unique therapies that have been developed to target CDK activity in the context of cancer treatment or senescence-associated pathologies, providing novel tools for precision medicine.
Collapse
Affiliation(s)
- Marina Bury
- De Duve Institute, UCLouvain, 1200 Brussels, Belgium
| | | | - Gerardo Ferbeyre
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, H3C 3J7, Canada.
| | - Volker Blank
- Lady Davis Institute for Medical Research, Departments of Medicine and Physiology, McGill University, Montreal, QC, H3T 1E2, Canada.
| | - Frédéric Lessard
- Department of Biochemistry and Molecular Medicine, University of Montreal, Montreal, QC, H3C 3J7, Canada.
| |
Collapse
|
15
|
Liu Y, Yu W, Ren P, Zhang T. Upregulation of centromere protein M promotes tumorigenesis: A potential predictive target for cancer in humans. Mol Med Rep 2020; 22:3922-3934. [PMID: 33000180 PMCID: PMC7533490 DOI: 10.3892/mmr.2020.11461] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 07/20/2020] [Indexed: 01/04/2023] Open
Abstract
Centromere protein M (CENPM), a protein required for chromosome separation, is involved in in mitosis. However, little has been reported about the roles of CENPM in various types of cancer. The present study identified that the mRNA expression levels of CENPM were significantly upregulated in 14 types of human cancer and identified a positive association between CENPM mRNA expression and patient mortality using the Oncomine, Gene Expression Profiling Interactive Analysis, Human Protein Atlas and Kaplan‑Meier Plotter databases. A protein interaction network constructed with CENPM‑interacting genes obtained from the cBioPortal demonstrated that nine genes participating in the cell cycle served key roles in the function of CENPM. Cell cycle analysis, reverse transcription‑quantitative polymerase chain reaction, a Cell Counting Kit‑8‑based proliferation assay and a terminal deoxynucleotidyl transferase dUTP nick end labelling assay further revealed the tumorigenic and carcinogenic roles of CENPM in vitro. In addition, it was identified that the mRNA expression levels of five of the nine identified genes were significantly associated with CENPM in MCF7 cells and that CENPM was rarely mutated among various types of human cancer. In conclusion, the data from the present study revealed that CENPM exerted its pro‑tumorigenic function by regulating cell cycle‑associated protein expression and suggested that CENPM could be used as a prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Ying Liu
- Key Laboratory of Endemic and Ethnic Diseases of The Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Wenfeng Yu
- Key Laboratory of Endemic and Ethnic Diseases of The Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Peng Ren
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 556000, P.R. China
| | - Ting Zhang
- Key Laboratory of Endemic and Ethnic Diseases of The Ministry of Education, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
16
|
Bibbo' S, Lamolinara A, Capone E, Purgato S, Tsakaneli A, Panella V, Sallese M, Rossi C, Ciufici P, Nieddu V, De Laurenzi V, Iezzi M, Perini G, Sala G, Sala A. Repurposing a psychoactive drug for children with cancer: p27 Kip1-dependent inhibition of metastatic neuroblastomas by Prozac. Oncogenesis 2020; 9:3. [PMID: 31900399 PMCID: PMC6949307 DOI: 10.1038/s41389-019-0186-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2019] [Revised: 12/02/2019] [Accepted: 12/03/2019] [Indexed: 12/31/2022] Open
Abstract
The MYC family of transcription factors is a major driver of human cancer and potential therapeutic target. However, no clinically viable drugs have been yet developed that are able to directly tackle MYC oncoproteins. In our laboratory, we are exploring alternative approaches aiming to disturb signalling downstream of MYC. MYCN is frequently activated in neuroblastoma, a paediatric solid malignancy that, in its metastatic form, has a very poor prognosis. An important pathway regulated by MYC is the CKS1/SKP2/p27kip1 axis. In this study, we have repurposed the anti-psychotic drug Prozac to disrupt CKS1/SKP2/p27Kip1 signalling and assess its potential as an anti-neuroblastoma agent in vitro and in vivo. Using DNA editing technology, we show that stabilisation of p27Kip1 operated by Prozac in MYC-activated cells is essential for the anti-neuroblastoma activity of the drug. Furthermore, dosing mice with a concentration of Prozac equivalent to that used in long-term clinical trials in children with psychiatric disorders caused a significant reduction of metastatic disease in two models of high-risk neuroblastoma. The favourable toxicity profile of Prozac suggests that long-term treatments might be implemented in children with MYC/CKS1high neuroblastomas.
Collapse
Affiliation(s)
- Sandra Bibbo'
- Dipartimento di Scienze Psicologiche, della Salute e del Territorio, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Alessia Lamolinara
- Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Dipartimento di Medicina e Scienze dell'Invecchiamento, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Emily Capone
- Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Dipartmento di Scienze Mediche, Orali e Biotecnologiche, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Stefania Purgato
- Dipartimento di Farmacia e Biotecnologia, Universita' di Bologna, 40126, Bologna, Italy
| | - Alexia Tsakaneli
- Institute of Environment, Health and Societies, College of Health and Life Sciences, Brunel University London, UB8 3PH, Uxbridge, UK
| | - Valeria Panella
- Dipartimento di Scienze Psicologiche, della Salute e del Territorio, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Michele Sallese
- Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Dipartmento di Scienze Mediche, Orali e Biotecnologiche, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Cosmo Rossi
- Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Paolo Ciufici
- Dipartimento di Scienze Psicologiche, della Salute e del Territorio, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Valentina Nieddu
- Dipartimento di Scienze Psicologiche, della Salute e del Territorio, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Unit of Gynecological Oncology Research, European Institute of Oncology, Via G. Ripamonti 435, 20141, Milano, Italy
| | - Vincenzo De Laurenzi
- Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Dipartmento di Scienze Mediche, Orali e Biotecnologiche, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Manuela Iezzi
- Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Dipartimento di Medicina e Scienze dell'Invecchiamento, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Giovanni Perini
- Dipartimento di Farmacia e Biotecnologia, Universita' di Bologna, 40126, Bologna, Italy
| | - Gianluca Sala
- Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy.,Dipartmento di Scienze Mediche, Orali e Biotecnologiche, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy
| | - Arturo Sala
- Dipartimento di Scienze Psicologiche, della Salute e del Territorio, Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy. .,Centro di Studi e Tecnologie Avanzate (CAST), Università "G D'Annunzio" Chieti-Pescara, 66100, Chieti, Italy. .,Institute of Environment, Health and Societies, College of Health and Life Sciences, Brunel University London, UB8 3PH, Uxbridge, UK.
| |
Collapse
|
17
|
Post AEM, Bussink J, Sweep FCGJ, Span PN. Changes in DNA Damage Repair Gene Expression and Cell Cycle Gene Expression Do Not Explain Radioresistance in Tamoxifen-Resistant Breast Cancer. Oncol Res 2019; 28:33-40. [PMID: 31046897 PMCID: PMC7851527 DOI: 10.3727/096504019x15555794826018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Tamoxifen-induced radioresistance, reported in vitro, might pose a problem for patients who receive neoadjuvant tamoxifen treatment and subsequently receive radiotherapy after surgery. Previous studies suggested that DNA damage repair or cell cycle genes are involved, and could therefore be targeted to preclude the occurrence of cross-resistance. We aimed to characterize the observed cross-resistance by investigating gene expression of DNA damage repair genes and cell cycle genes in estrogen receptor-positive MCF-7 breast cancer cells that were cultured to tamoxifen resistance. RNA sequencing was performed, and expression of genes characteristic for several DNA damage repair pathways was investigated, as well as expression of genes involved in different phases of the cell cycle. The association of differentially expressed genes with outcome after radiotherapy was assessed in silico in a large breast cancer cohort. None of the DNA damage repair pathways showed differential gene expression in tamoxifen-resistant cells compared to wild-type cells. Two DNA damage repair genes were more than two times upregulated (NEIL1 and EME2), and three DNA damage repair genes were more than two times downregulated (PCNA, BRIP1, and BARD1). However, these were not associated with outcome after radiotherapy in the TCGA breast cancer cohort. Genes involved in G1, G1/S, G2, and G2/M phases were lower expressed in tamoxifen-resistant cells compared to wild-type cells. Individual genes that were more than two times upregulated (MAPK13) or downregulated (E2F2, CKS2, GINS2, PCNA, MCM5, and EIF5A2) were not associated with response to radiotherapy in the patient cohort investigated. We assessed the expression of DNA damage repair genes and cell cycle genes in tamoxifen-resistant breast cancer cells. Though several genes in both pathways were differentially expressed, these could not explain the cross-resistance for irradiation in these cells, since no association to response to radiotherapy in the TCGA breast cancer cohort was found.
Collapse
Affiliation(s)
- Annemarie E M Post
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| | - Johan Bussink
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| | - Fred C G J Sweep
- Radboud University Medical Center, Department of Laboratory MedicineNijmegenThe Netherlands
| | - Paul N Span
- Radboud University Medical Center, Department of Radiation Oncology, Radiotherapy and OncoImmunology LaboratoryNijmegenThe Netherlands
| |
Collapse
|
18
|
Grey W, Ivey A, Milne TA, Haferlach T, Grimwade D, Uhlmann F, Voisset E, Yu V. The Cks1/Cks2 axis fine-tunes Mll1 expression and is crucial for MLL-rearranged leukaemia cell viability. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2018; 1865:105-116. [PMID: 28939057 PMCID: PMC5701546 DOI: 10.1016/j.bbamcr.2017.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 09/09/2017] [Accepted: 09/17/2017] [Indexed: 12/25/2022]
Abstract
The Cdc28 protein kinase subunits, Cks1 and Cks2, play dual roles in Cdk-substrate specificity and Cdk-independent protein degradation, in concert with the E3 ubiquitin ligase complexes SCFSkp2 and APCCdc20. Notable targets controlled by Cks include p27 and Cyclin A. Here, we demonstrate that Cks1 and Cks2 proteins interact with both the MllN and MllC subunits of Mll1 (Mixed-lineage leukaemia 1), and together, the Cks proteins define Mll1 levels throughout the cell cycle. Overexpression of CKS1B and CKS2 is observed in multiple human cancers, including various MLL-rearranged (MLLr) AML subtypes. To explore the importance of MLL-Fusion Protein regulation by CKS1/2, we used small molecule inhibitors (MLN4924 and C1) to modulate their protein degradation functions. These inhibitors specifically reduced the proliferation of MLLr cell lines compared to primary controls. Altogether, this study uncovers a novel regulatory pathway for MLL1, which may open a new therapeutic approach to MLLr leukaemia.
Collapse
Affiliation(s)
- William Grey
- Department of Medical and Molecular Genetics, King's College London, Faculty of Life Sciences and Medicine, London, UK.
| | - Adam Ivey
- Department of Medical and Molecular Genetics, King's College London, Faculty of Life Sciences and Medicine, London, UK
| | - Thomas A Milne
- MRC Molecular Haematology Unit, Weatherall Institute of Molecular Medicine, NIHR Oxford Biomedical Research Centre Programme, University of Oxford, UK
| | | | - David Grimwade
- Department of Medical and Molecular Genetics, King's College London, Faculty of Life Sciences and Medicine, London, UK
| | - Frank Uhlmann
- Chromosome Segregation Laboratory, The Francis Crick Institute, London, UK
| | - Edwige Voisset
- Department of Medical and Molecular Genetics, King's College London, Faculty of Life Sciences and Medicine, London, UK.
| | - Veronica Yu
- Department of Medical and Molecular Genetics, King's College London, Faculty of Life Sciences and Medicine, London, UK
| |
Collapse
|
19
|
Kukalev A, Ng YM, Ju L, Saidi A, Lane S, Mondragon A, Dormann D, Walker SE, Grey W, Ho PWL, Stephens DN, Carr AM, Lamsa K, Tse E, Yu VPCC. Deficiency of Cks1 Leads to Learning and Long-Term Memory Defects and p27 Dependent Formation of Neuronal Cofilin Aggregates. Cereb Cortex 2017; 27:11-23. [PMID: 28365778 PMCID: PMC5939225 DOI: 10.1093/cercor/bhw354] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2016] [Revised: 09/23/2016] [Indexed: 01/11/2023] Open
Abstract
In mitotic cells, the cyclin-dependent kinase (CDK) subunit protein CKS1 regulates S phase entry by mediating degradation of the CDK inhibitor p27. Although mature neurons lack mitotic CDKs, we found that CKS1 was actively expressed in post-mitotic neurons of the adult hippocampus. Interestingly, Cks1 knockout (Cks1-/-) mice exhibited poor long-term memory, and diminished maintenance of long-term potentiation in the hippocampal circuits. Furthermore, there was neuronal accumulation of cofilin-actin rods or cofilin aggregates, which are associated with defective dendritic spine maturation and synaptic loss. We further demonstrated that it was the increased p27 level that activated cofilin by suppressing the RhoA kinase-mediated inhibitory phosphorylation of cofilin, resulting in the formation of cofilin aggregates in the Cks1-/- neuronal cells. Consistent with reports that the peptidyl-prolyl-isomerase PIN1 competes with CKS1 for p27 binding, we found that inhibition of PIN1 diminished the formation of cofilin aggregates through decreasing p27 levels, thereby activating RhoA and increasing cofilin phosphorylation. Our results revealed that CKS1 is involved in normal glutamatergic synapse development and dendritic spine maturation in adult hippocampus through modulating p27 stability.
Collapse
Affiliation(s)
- Alexander Kukalev
- Eukaryotic Chromatin Dynamics Group
,
MRC Clinical Sciences Centre
,
Imperial College Hammersmith Campus
,
London W12 0NN
,
UK
- Department of Medical and Molecular Genetics
,
King's College London School of Medicine
,
Guy's Hospital
,
Great Maze Pond
,
London SE1 9RT
,
UK
- Current address:
Epigenetic Regulation and Chromatin Architecture Group
,
Berlin Institute for Medical Systems Biology, Max-Delbrück Centre for Molecular Medicine
,
Robert-Rössle Strasse
,
Berlin-Buch 13125
,
Germany
| | - Yiu-Ming Ng
- Department of Medical and Molecular Genetics
,
King's College London School of Medicine
,
Guy's Hospital
,
Great Maze Pond
,
London SE1 9RT
,
UK
- Division of Haematology
,
Department of Medicine
,
The University of Hong Kong
,
Hong Kong
| | - Limei Ju
- Genome Damage and Stability Centre
,
School of Life Sciences
,
University of Sussex
,
Falmer, Sussex BN1 9RQ
,
UK
| | - Amal Saidi
- Genome Damage and Stability Centre
,
School of Life Sciences
,
University of Sussex
,
Falmer, Sussex BN1 9RQ
,
UK
| | - Sophie Lane
- Eukaryotic Chromatin Dynamics Group
,
MRC Clinical Sciences Centre
,
Imperial College Hammersmith Campus
,
London W12 0NN
,
UK
| | - Angeles Mondragon
- Eukaryotic Chromatin Dynamics Group
,
MRC Clinical Sciences Centre
,
Imperial College Hammersmith Campus
,
London W12 0NN
,
UK
| | - Dirk Dormann
- Microscopy Facility
,
MRC Clinical Sciences Centre
,
Imperial College Hammersmith Campus
,
London W12 0NN
,
UK
| | - Sophie E. Walker
- School of Psychology
,
University of Sussex
,
Sussex, Brighton BN1 9QG
,
UK
| | - William Grey
- Department of Medical and Molecular Genetics
,
King's College London School of Medicine
,
Guy's Hospital
,
Great Maze Pond
,
London SE1 9RT
,
UK
| | - Philip Wing-Lok Ho
- Division of Neurology
,
Department of Medicine
,
University of Hong Kong
,
Hong Kong
| | - David N. Stephens
- School of Psychology
,
University of Sussex
,
Sussex, Brighton BN1 9QG
,
UK
| | - Antony M. Carr
- Genome Damage and Stability Centre
,
School of Life Sciences
,
University of Sussex
,
Falmer, Sussex BN1 9RQ
,
UK
| | - Karri Lamsa
- Department of Pharmacology
,
Oxford University
,
Oxford OX1 3QT
,
UK
- Current address:
Department of Physiology, Anatomy and Neuroscience
,
University of Szeged
,
Közép fasor 52
,
Szeged H-6726,Hungary
| | - Eric Tse
- Division of Haematology
,
Department of Medicine
,
The University of Hong Kong
,
Hong Kong
| | - Veronica P. C. C. Yu
- Eukaryotic Chromatin Dynamics Group
,
MRC Clinical Sciences Centre
,
Imperial College Hammersmith Campus
,
London W12 0NN
,
UK
- Department of Medical and Molecular Genetics
,
King's College London School of Medicine
,
Guy's Hospital
,
Great Maze Pond
,
London SE1 9RT
,
UK
| |
Collapse
|
20
|
Characterization of cyclin-dependent kinases and Cdc2/Cdc28 kinase subunits in Trichomonas vaginalis. Parasitology 2016; 144:571-582. [PMID: 27928981 DOI: 10.1017/s0031182016002195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Cyclin-dependent kinases (CDKs) have important roles in regulating key checkpoints between stages of the cell cycle. Their activity is tightly regulated through a variety of mechanisms, including through binding with cyclin proteins and the Cdc2/Cdc28 kinase subunit (CKS), and their phosphorylation at specific amino acids. Studies of the components involved in cell cycle control in parasitic protozoa are limited. Trichomonas vaginalis is the causative agent of trichomoniasis in humans and is therefore important in public health; however, some of the basic biological processes used by this organism have not been defined. Here, we characterized proteins potentially involved in cell cycle regulation in T. vaginalis. Three genes encoding protein kinases were identified in the T. vaginalis genome, and the corresponding recombinant proteins (TvCRK1, TvCRK2, TvCRK5) were studied. These proteins displayed similar sequence features to CDKs. Two genes encoding CKSs were also identified, and the corresponding recombinant proteins were found to interact with TvCRK1 and TvCRK2 by a yeast two-hybrid system. One putative cyclin B protein from T. vaginalis was found to bind to and activate the kinase activities of TvCRK1 and TvCRK5, but not TvCRK2. This work is the first characterization of proteins involved in cell cycle control in T. vaginalis.
Collapse
|
21
|
Hua K, Jin J, Zhang H, Zhao B, Wu C, Xu H, Fang L. MicroRNA-7 inhibits proliferation, migration and invasion of thyroid papillary cancer cells via targeting CKS2. Int J Oncol 2016; 49:1531-1540. [DOI: 10.3892/ijo.2016.3660] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/05/2016] [Indexed: 11/05/2022] Open
|
22
|
Hafez MM, Alhoshani AR, Al-Hosaini KA, Alsharari SD, Al Rejaie SS, Sayed-Ahmed MM, Al-Shabanah OA. SKP2/P27Kip1 pathway is associated with Advanced Ovarian Cancer in Saudi Patients. Asian Pac J Cancer Prev 2016; 16:5807-15. [PMID: 26320455 DOI: 10.7314/apjcp.2015.16.14.5807] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Ovarian cancer is the most common gynecological malignancy and constitutes the fifth leading cause of female cancer death. Some biological parameters have prognostic roles in patients with advanced ovarian cancer and their expression may contribute to tumor progression. The aim of this study was to investigate the potential prognostic value of SKP2, genes P27Kip1, K-ras, c-Myc, COX2 and HER2 genes expression in ovarian cancer. MATERIALS AND METHODS This study was performed on two hundred formalin fixed paraffin embedded ovarian cancer and normal adjacent tissues (NAT). Gene expression levels were assessed using real time PCR and Western blotting. RESULTS Elevated expression levels of SKP2, K-ras, c-Myc, HER2 and COX2 genes were observed in 61.5% (123/200), 92.5% (185/200), 74% (148/200), 96 % (192/200), 90% (180/200) and 78.5% (157/200) of cancer tissues, respectively. High expression of SKP2 and down-regulation of P27 was associated with advanced stages of cancer. CONCLUSIONS The association between high expression of c-Myc and SKP2 with low expression of P27 suggested that the Skp2-P27 pathway may play an important role in ovarian carcinogenesis. Reduced expression of P27 is associated with advanced stage of cancer and can be used as a biological marker in clinical routine assessment and management of women with advanced ovarian cancer.
Collapse
Affiliation(s)
- Mohamed M Hafez
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University Riyadh, Kingdom of Saudi Arabia E-mail :
| | | | | | | | | | | | | |
Collapse
|
23
|
Zheng J, Li Q, Wang W, Wang Y, Fu X, Wang W, Fan L, Yan W. Apoptosis-related protein-1 acts as a tumor suppressor in cholangiocarcinoma cells by inducing cell cycle arrest via downregulation of cyclin-dependent kinase subunits. Oncol Rep 2015; 35:809-16. [PMID: 26572808 DOI: 10.3892/or.2015.4422] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 10/13/2015] [Indexed: 11/05/2022] Open
Abstract
Cholangiocarcinoma, a malignancy arising from the biliary tract, is associated with high mortality due to the late diagnosis and lack of effective therapeutic approaches. Our knowledge of the molecular alterations during the carcinogenesis of cholangiocarcinoma is limited. Previous study suggests that apoptosis-related protein-1 (Apr-1) is involved in cancer cell proliferation and survival. In the present study, we first detected the expression pattern of Apr-1 in human cholangiocarcinoma tissues and the effects of forced Apr-1 expression on cell proliferation and cell cycle progression. Cell cycle gene array analysis was used to identify downstream molecules that were regulated by Apr-1, and their expression levels were further evaluated in human cholangiocarcinoma tissues. We showed that Apr-1 expression was downregulated in human cholangiocarcinoma tissues. Forced expression of Apr-1 inhibited cell proliferation of cholangiocarcinoma cell line QBC939 and induced G2/M phase arrest. Downregulation of cell cycle-related genes cyclin-dependent kinase (Cdk) 2, and cyclin-dependent kinase subunits (Cks) 1 and 2 was involved in Apr-1-induced cell cycle arrest. Furthermore, we found that Cdk2 and Cks1/2 expression levels were elevated in human cholangiocarcinoma tissues. Taken together, our data showed that Apr-1 plays a crucial role in cell proliferation by controlling cell cycle progression, implying a tumor-suppressor function of Apr-1 in cholangiocarcinoma carcinogenesis. Thus, the present study provides a rationale to further study the underlying mechanisms of Apr-1 downregulation in cholangiocarcinoma for exploring potential diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Jianyong Zheng
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Qinlong Li
- State Key Laboratory of Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Weihua Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Yingmei Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Xin Fu
- State Key Laboratory of Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wenyong Wang
- State Key Laboratory of Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Linni Fan
- State Key Laboratory of Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Wei Yan
- State Key Laboratory of Cancer Biology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
24
|
Yu MH, Luo Y, Qin SL, Wang ZS, Mu YF, Zhong M. Up-regulated CKS2 promotes tumor progression and predicts a poor prognosis in human colorectal cancer. Am J Cancer Res 2015; 5:2708-2718. [PMID: 26609478 PMCID: PMC4633900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2015] [Accepted: 07/20/2015] [Indexed: 06/05/2023] Open
Abstract
Cyclin-dependent kinases regulatory subunit 2 (CKS2) is a cyclin-dependent kinase-interacting protein, which is essential for cell cycle regulation. Elevated expression of CKS2 has been demonstrated in multiple types of human malignancies. However, the clinical significance, oncogenic functions and related mechanisms of CKS2 in colorectal cancer (CRC) remain largely unexplored. In this study, data from Oncomine database revealed that CKS2 is significantly up-regulated in CRC tissues compared with their normal counterparts. Immunohistochemical analysis of a CRC tissue microarray demonstrated that elevated CKS2 expression is closely associated with enhanced TNM stage, larger tumor size and a poor prognosis in patients with CRC. Multivariate Cox regression analysis revealed that CKS2 and TNM stage are two independent prognostic factors for CRC. Suppression of CKS2 expression resulted in decreased cell viability, increased cell apoptosis, cell cycle arrest and reduced expression of cyclins in Caco-2 and SW620 cells. Furthermore, gain and loss of function studies demonstrated that CKS2 promotes cell invasion in CRC cells through regulating claudin1. Taken together, our study reveal that CKS2 is promising prognostic indicator and contributes to tumor progression in CRC, and support that CKS2-related signaling may represent a novel target for CRC therapy.
Collapse
Affiliation(s)
- Min-Hao Yu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, P. R. China
| | - Yang Luo
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, P. R. China
| | - Shao-Lan Qin
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, P. R. China
| | - Zheng-Shi Wang
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, P. R. China
| | - Yi-Fei Mu
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, P. R. China
| | - Ming Zhong
- Department of Gastrointestinal Surgery, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127, P. R. China
| |
Collapse
|
25
|
Wang J, Xu L, Liu Y, Chen J, Jiang H, Yang S, Tan H. Expression of cyclin kinase subunit 2 in human breast cancer and its prognostic significance. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2014; 7:8593-8601. [PMID: 25674223 PMCID: PMC4313994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Accepted: 12/01/2014] [Indexed: 06/04/2023]
Abstract
Cyclin kinase subunit 2 (CKS2) protein is a small cyclin-dependent kinase-interacting protein, which is essential for the first metaphase/anaphase transition of mammalian meiosis. CKS2 is up-regulated in various malignancies, suggesting that CKS2 maybe an oncogene. However, data on its expression pattern and clinical relevance in breast cancer are unknown. The aim of this study is to investigate CKS2 expression and its prognostic significance in breast cancer. The CKS2 expression was examined at mRNA and protein levels by real-time quantitative polymerase chain reaction (RT-PCR) and Western blotting analysis in paired breast cancer tissues and the adjacent normal tissues. The expression of CKS2 protein in 126 specimens of breast cancer was determined by immunohistochemistry assay. The relations between CKS2 expression and clinicopathological features were analyzed. The result show the expression of CKS2 mRNA and protein was higher in breast cancer than the adjacent normal tissues. Compared with adjacent normal breast tissues, Overexpression of CKS2 was detected in 56.3% (71/126) patients. Overexpression of CKS2 was significantly associated with large tumor size (P = 0.035), poor cellular differentiation (P = 0.016), lack expression of progesterone receptor (P = 0.006), and decreased overall survival (P = 0.001). In multivariate analysis, CKS2 expression was an independent prognostic factor for overall survival (Hazard ratio [HR] = 3.404, 95% confidence interval [CI] 1.482-7.818; P = 0.004). CKS2 is up-regulated in breast cancer and associated with large tumor size, lack expression of progesterone receptor, poor tumor differentiation and survival. CKS2 may serve as a good prognostic indicator for patients with breast cancer.
Collapse
Affiliation(s)
- Jiani Wang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, China
| | - Lihua Xu
- Department of Oncology and Hematology, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Yu Liu
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, China
| | - Jianning Chen
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, China
| | - Hua Jiang
- Breast Cancer Center, The Third Affiliated Hospital of Sun Yat-Sen UniversityGuangzhou, China
| | - Shaojiang Yang
- Department of Oncology and Hematology, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| | - Huo Tan
- Department of Oncology and Hematology, The First Affiliated Hospital of Guangzhou Medical UniversityGuangzhou, China
| |
Collapse
|
26
|
Van de Casteele M, Leuckx G, Cai Y, Yuchi Y, Coppens V, De Groef S, Van Gassen N, Baeyens L, Heremans Y, Wright CVE, Heimberg H. Partial duct ligation: β-cell proliferation and beyond. Diabetes 2014; 63:2567-77. [PMID: 25060885 DOI: 10.2337/db13-0831] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Affiliation(s)
| | - Gunter Leuckx
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Ying Cai
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yixing Yuchi
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Violette Coppens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Sofie De Groef
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Naomi Van Gassen
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Luc Baeyens
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Yves Heremans
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| | - Christopher V E Wright
- Vanderbilt University Program in Developmental Biology, Department of Cell and Developmental Biology, Vanderbilt University Medical Center, Nashville, TN
| | - Harry Heimberg
- Diabetes Research Center, Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
27
|
Zhang W, Cheng Y, Li Y, Chen Z, Jin P, Chen D. A feed-forward mechanism involving Drosophila fragile X mental retardation protein triggers a replication stress-induced DNA damage response. Hum Mol Genet 2014; 23:5188-96. [PMID: 24833720 DOI: 10.1093/hmg/ddu241] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Fragile X syndrome, a common form of inherited mental retardation, is caused by loss of the fragile X mental retardation protein (FMRP). As a selective RNA-binding protein, FMRP is localized predominately in cytoplasm, where it regulates translational control. However, there is a small portion of FMRP present in the nucleus, and its function there has been elusive. Here, we show that Drosophila dFMR1 in nucleus is required for replication stress-induced H2Av phosphorylation in the DNA damage response (DDR). Replication stress could induce the expression of dFmr1 and promote the nuclear accumulation of dFMR1. We show that, upon the stimulation of replication stress, dFMR1 is associated with chromatin in a domain-specific manner, which is essential for its ability to induce the phosphorylation of H2Av. These results together reveal an unexpected nuclear role of FMRP in DDR and uncover a feed-forward mechanism by which dFmr1 and early DDR induced by replication stress reciprocally regulate each other, thereby synergistically triggering activity of the DDR signaling cascade.
Collapse
Affiliation(s)
- Wenxin Zhang
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Ying Cheng
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Yujing Li
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Zhenping Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| | - Peng Jin
- Department of Human Genetics, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Dahua Chen
- State Key Laboratory of Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, PR China
| |
Collapse
|
28
|
Reinhardt HC, Yaffe MB. Phospho-Ser/Thr-binding domains: navigating the cell cycle and DNA damage response. Nat Rev Mol Cell Biol 2013; 14:563-80. [PMID: 23969844 DOI: 10.1038/nrm3640] [Citation(s) in RCA: 216] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Coordinated progression through the cell cycle is a complex challenge for eukaryotic cells. Following genotoxic stress, diverse molecular signals must be integrated to establish checkpoints specific for each cell cycle stage, allowing time for various types of DNA repair. Phospho-Ser/Thr-binding domains have emerged as crucial regulators of cell cycle progression and DNA damage signalling. Such domains include 14-3-3 proteins, WW domains, Polo-box domains (in PLK1), WD40 repeats (including those in the E3 ligase SCF(βTrCP)), BRCT domains (including those in BRCA1) and FHA domains (such as in CHK2 and MDC1). Progress has been made in our understanding of the motif (or motifs) that these phospho-Ser/Thr-binding domains connect with on their targets and how these interactions influence the cell cycle and DNA damage response.
Collapse
Affiliation(s)
- H Christian Reinhardt
- David H. Koch Institute for Integrative Cancer Research, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | |
Collapse
|
29
|
Tompkins VS, Han SS, Olivier A, Syrbu S, Bair T, Button A, Jacobus L, Wang Z, Lifton S, Raychaudhuri P, Morse HC, Weiner G, Link B, Smith BJ, Janz S. Identification of candidate B-lymphoma genes by cross-species gene expression profiling. PLoS One 2013; 8:e76889. [PMID: 24130802 PMCID: PMC3793908 DOI: 10.1371/journal.pone.0076889] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 08/29/2013] [Indexed: 01/08/2023] Open
Abstract
Comparative genome-wide expression profiling of malignant tumor counterparts across the human-mouse species barrier has a successful track record as a gene discovery tool in liver, breast, lung, prostate and other cancers, but has been largely neglected in studies on neoplasms of mature B-lymphocytes such as diffuse large B cell lymphoma (DLBCL) and Burkitt lymphoma (BL). We used global gene expression profiles of DLBCL-like tumors that arose spontaneously in Myc-transgenic C57BL/6 mice as a phylogenetically conserved filter for analyzing the human DLBCL transcriptome. The human and mouse lymphomas were found to have 60 concordantly deregulated genes in common, including 8 genes that Cox hazard regression analysis associated with overall survival in a published landmark dataset of DLBCL. Genetic network analysis of the 60 genes followed by biological validation studies indicate FOXM1 as a candidate DLBCL and BL gene, supporting a number of studies contending that FOXM1 is a therapeutic target in mature B cell tumors. Our findings demonstrate the value of the "mouse filter" for genomic studies of human B-lineage neoplasms for which a vast knowledge base already exists.
Collapse
Affiliation(s)
- Van S. Tompkins
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Seong-Su Han
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Alicia Olivier
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Sergei Syrbu
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Thomas Bair
- Bioinformatics, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Anna Button
- Department of Biostatistics, University of Iowa College of Public Health, Iowa City, Iowa, United States of America
| | - Laura Jacobus
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Zebin Wang
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
| | - Samuel Lifton
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Department of Statistics & Actuarial Science, University of Iowa College of Liberal Arts & Sciences, Iowa City, Iowa, United States of America
| | - Pradip Raychaudhuri
- Department of Biochemistry and Molecular Genetics, University of Illinois at Chicago College of Medicine, Chicago, Illinois, United States of America
| | - Herbert C. Morse
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - George Weiner
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Brian Link
- Department of Internal Medicine, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Brian J. Smith
- Department of Biostatistics, University of Iowa College of Public Health, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| | - Siegfried Janz
- Department of Pathology, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
- Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, Iowa, United States of America
| |
Collapse
|
30
|
Guo X, Yang C, Qian X, Lei T, Li Y, Shen H, Fu L, Xu B. Estrogen receptor α regulates ATM Expression through miRNAs in breast cancer. Clin Cancer Res 2013; 19:4994-5002. [PMID: 23857602 DOI: 10.1158/1078-0432.ccr-12-3700] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE Estrogen receptor α (ERα) is an essential element regulating mammary gland development and it contributes to breast cancer development and progression. Most of the ER-negative breast cancers display more aggressive clinical behaviors and are resistant to antiestrogen therapies. In addition, many ER-negative tumors show insensitivity to many chemotherapeutic drugs and radiotherapy, although mechanisms underlying this phenotype are less clear. EXPERIMENTAL DESIGN We conducted immunohistochemistry on 296 cases of breast cancer tissues using a variety of antibodies. On the basis of the clinical data, we conducted siRNA knockdown to study the role of ERα on ATM expression in breast cancer cell lines. Furthermore, we used antisense oligonucleotides against micro RNAs (miRNA) or miRNA overexpression plasmids to study the role of miR-18a and -106a on ATM expression. Finally we used in situ hybridization to assess miR-18a and -106a expression in breast cancer tissues. RESULTS We found that in ER-negative breast cancer tissues, expression of the ATM kinase, a critical DNA damage-response protein, is aberrantly upregulated. We also found that the locoregional recurrence rate after radiotherapy positively correlates with ATM expression. On the cellular level, we showed that ERα, but not ERβ, negatively regulates ATM expression. Furthermore, we identified that ERα activates miR-18a and -106a to downregulate ATM expression. We also showed that miR-18a and -106a were significantly underexpressed in ER-negative breast cancer tissues. CONCLUSIONS We reveal a novel mechanism involving ERα and miR-18a and -106a regulation of ATM in breast cancer.
Collapse
Affiliation(s)
- Xiaojing Guo
- Authors' Affiliations: Department of Breast Pathology and Lab, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China; Departments of Radiation Oncology, and Nanomedicine, The Methodist Hospital Research Institute, Houston, Texas; and Department of Biochemistry and Molecular Biology, Southern Research Institute, Birmingham, Alabama
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Abstract
The cell cycle ensures genome maintenance by coordinating the processes of DNA replication and chromosome segregation. Of particular importance is the irreversible transition from the G1 phase of the cell cycle to S phase. This transition marks the switch from preparing chromosomes for replication ("origin licensing") to active DNA synthesis ("origin firing"). Ubiquitin-mediated proteolysis is essential for restricting DNA replication to only once per cell cycle and is the major mechanism regulating the G1 to S phase transition. Although some changes in protein levels are attributable to regulated mRNA abundance, protein degradation elicits very rapid changes in protein abundance and is critical for the sharp and irreversible transition from one cell cycle stage to the next. Not surprisingly, regulation of the G1-to-S phase transition is perturbed in most cancer cells, and deregulation of key molecular events in G1 and S phase drives not only cell proliferation but also genome instability. In this review we focus on the mechanisms by which E3 ubiquitin ligases control the irreversible transition from G1 to S phase in mammalian cells.
Collapse
Affiliation(s)
- Lindsay F Rizzardi
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | |
Collapse
|
32
|
Galons H, Oumata N, Gloulou O, Meijer L. Cyclin-dependent kinase inhibitors closer to market launch? Expert Opin Ther Pat 2013; 23:945-63. [PMID: 23600454 DOI: 10.1517/13543776.2013.789861] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Interest in cyclin-dependent kinase (CDK) inhibitors was stimulated by the demonstration that their pharmacological activities could lead to therapies for numerous diseases. Until now, despite the clinical introduction of a dozen compounds belonging to other classes of kinase inhibitors, no CDK inhibitor has reached the marketplace. AREAS COVERED This review covers CDK inhibitor patents published between 2009 and September 2012. It presents compounds currently undergoing clinical development, along with our earlier (2010) review of the same topic, as well as descriptions of recently published compounds not disclosed in the patent literature. It provides the reader with an update of all chemical structures of current interest in the CDK inhibitor field. EXPERT OPINION Though cancer remains the most obvious application for CDK inhibition, other indications, such as HIV infection, could potentially be treated with CDK inhibitors.
Collapse
Affiliation(s)
- Hervé Galons
- Laboratoire de Chimie Organique 2, INSERM U 1022, Université Paris - Descartes, 4 avenue de l'Observatoire, 75270 Paris cedex 06, France.
| | | | | | | |
Collapse
|
33
|
Khattar V, Thottassery JV. Cks1: Structure, Emerging Roles and Implications in Multiple Cancers. ACTA ACUST UNITED AC 2013; 4:1341-1354. [PMID: 24563807 PMCID: PMC3930463 DOI: 10.4236/jct.2013.48159] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Deregulation of the cell cycle results in loss of normal control mechanisms that prevent aberrant cell proliferation and cancer progression. Regulation of the cell cycle is a highly complex process with many layers of control. One of these mechanisms involves timely degradation of CDK inhibitors (CKIs) like p27Kip1 by the ubiquitin proteasomal system (UPS). Cks1 is a 9 kDa protein which is frequently overexpressed in different tumor subtypes, and has pleiotropic roles in cell cycle progression, many of which remain to be fully characterized. One well characterized molecular role of Cks1 is that of an essential adaptor that regulates p27Kip1 abundance by facilitating its interaction with the SCF-Skp2 E3 ligase which appends ubiquitin to p27Kip1 and targets it for degradation through the UPS. In addition, emerging research has uncovered p27Kip1-independent roles of Cks1 which have provided crucial insights into how it may be involved in cancer progression. We review here the structural features of Cks1 and their functional implications, and also some recently identified Cks1 roles and their involvement in breast and other cancers.
Collapse
Affiliation(s)
| | - Jaideep V Thottassery
- Southern Research Institute, Birmingham, USA ; University of Alabama Comprehensive Cancer Center, Birmingham, USA
| |
Collapse
|