1
|
Li X, Zou Y, Geng B, Liu P, Cao L, Zhang Z, Hu S, Wang C, Zhao Y, Wu Q, Tan J. Transcriptome analysis reveals that defects in cell cycle regulation contribute to preimplantation embryo arrest. Genomics 2024; 116:110946. [PMID: 39326642 DOI: 10.1016/j.ygeno.2024.110946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/09/2024] [Accepted: 09/23/2024] [Indexed: 09/28/2024]
Abstract
Patients with preimplantation embryo arrest (PREMBA) often experience assisted reproductive failure primarily due to the lack of transferable embryos, and the molecular mechanisms underlying PREMBA remain unclear. In our study, the embryos from five women with recurrent preimplantation embryo arrest and three women with tubal factor infertility were used for single-embryo transcriptome sequencing. Meanwhile, the transcriptomes of normal human preimplantation embryos obtained from GSE36552 were utilized to perform a comparative analysis with the transcriptomes of PREMBA embryos. Our results showed dysregulation of the cell cycle phase transition might be a potential pathogenic factor contributing to PREMBA. Through integrated analysis of the differentially expressed genes (DEGs) and weighted gene co-expression network analysis (WGCNA), we identified a number of hub genes using the protein-protein interaction network. The top 5 hub genes were as follows: CCNB2, BUB1B, CDC25A, CCNB3, and PLK3. The expression of hub genes was validated in PREMBA embryos and donated embryos using RT-qPCR. The knockdown of Ccnb2 in mouse zygotes led to an increase in embryo fragmentation, a rise in apoptosis, and a reduction in blastocyst formation. Furthermore, silencing the expression of CDC25A in HEK293T cells resulted in a decrease in cell proliferation and an increase in apoptosis, providing further support for our findings. Our findings could predict the development outcomes of preimplantation embryos and be used as potential therapeutic targets to prevent recurrent failures of IVF/ICSI attempts.
Collapse
Affiliation(s)
- Xin Li
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Yang Zou
- Central Laboratory, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Baobao Geng
- Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, SuZhou, Jiangsu Province, China
| | - Peipei Liu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Liyun Cao
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Zhiqin Zhang
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Shaofeng Hu
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Changhua Wang
- JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China
| | - Yan Zhao
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| | - Qiongfang Wu
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| | - Jun Tan
- Reproductive Medicine Center, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China; JXHC Key Laboratory of Fertility Preservation, Jiangxi Maternal and Child Health Hospital, Nanchang, Jiangxi Province, China.
| |
Collapse
|
2
|
Pavlovic D, Niciforovic D, Papic D, Milojevic K, Markovic M. CDK4/6 inhibitors: basics, pros, and major cons in breast cancer treatment with specific regard to cardiotoxicity - a narrative review. Ther Adv Med Oncol 2023; 15:17588359231205848. [PMID: 37841752 PMCID: PMC10571689 DOI: 10.1177/17588359231205848] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Breast cancer is characterized by the uncontrolled proliferation of breast cells, with a high incidence reported in 2020 to have affected over 2 million women. In recent years, the conventional methods of treating breast cancer have involved radiotherapy and chemotherapy. However, the emergence of CDK4/6 inhibitors has shown potential as a promising cancer therapy. Cyclin-dependent kinases (CDK) inhibitors are a class of molecules that impede the formation of an active kinase complex, thereby hindering its activity and consequently halting the progression of the cell cycle. It was discovered that they have a significant impact on impeding the progression of the cancer. This is evident with the Food and Drug Administration's approval of drugs such as palbociclib, ribociclib, and abemaciclib for hormone receptor-positive metastatic breast cancer in combination with specific endocrine therapies. In spite of enormous success in breast cancer treatment, certain obstacles have emerged, such as therapy resistance, side effects, and most of all, cardiotoxicity. Some of these drawbacks have been successfully overcome by dosage reduction, different combinations of the drugs, and the assessment of each patient's condition and suitability prior to treatment. Yet other drawbacks still require tenacious research, especially certain cases of cardiotoxicities. This article delves into the biological mechanisms of CDK4/6 in the cell cycle and cancer, as well as the clinical advantages and most common adverse events (AEs) associated with CDK4/6 inhibitors. The primary objective of this review is to provide a comprehensive analysis of cardiotoxic AEs and elucidate the underlying pathophysiological mechanisms responsible for the cardiotoxicity of CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Dragica Pavlovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, Kragujevac 34000, Serbia
| | - Danijela Niciforovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Dragana Papic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Katarina Milojevic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marina Markovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
3
|
Rowland RJ, Heath R, Maskell D, Thompson RF, Ranson NA, Blaza JN, Endicott JA, Noble MEM, Salamina M. Cryo-EM structure of SKP1-SKP2-CKS1 in complex with CDK2-cyclin A-p27KIP1. Sci Rep 2023; 13:10718. [PMID: 37400515 PMCID: PMC10318019 DOI: 10.1038/s41598-023-37609-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/24/2023] [Indexed: 07/05/2023] Open
Abstract
p27KIP1 (cyclin-dependent kinase inhibitor 1B, p27) is a member of the CIP/KIP family of CDK (cyclin dependent kinase) regulators that inhibit cell cycle CDKs. p27 phosphorylation by CDK1/2, signals its recruitment to the SCFSKP2 (S-phase kinase associated protein 1 (SKP1)-cullin-SKP2) E3 ubiquitin ligase complex for proteasomal degradation. The nature of p27 binding to SKP2 and CKS1 was revealed by the SKP1-SKP2-CKS1-p27 phosphopeptide crystal structure. Subsequently, a model for the hexameric CDK2-cyclin A-CKS1-p27-SKP1-SKP2 complex was proposed by overlaying an independently determined CDK2-cyclin A-p27 structure. Here we describe the experimentally determined structure of the isolated CDK2-cyclin A-CKS1-p27-SKP1-SKP2 complex at 3.4 Å global resolution using cryogenic electron microscopy. This structure supports previous analysis in which p27 was found to be structurally dynamic, transitioning from disordered to nascent secondary structure on target binding. We employed 3D variability analysis to further explore the conformational space of the hexameric complex and uncovered a previously unidentified hinge motion centred on CKS1. This flexibility gives rise to open and closed conformations of the hexameric complex that we propose may contribute to p27 regulation by facilitating recognition with SCFSKP2. This 3D variability analysis further informed particle subtraction and local refinement approaches to enhance the local resolution of the complex.
Collapse
Affiliation(s)
- Rhianna J Rowland
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Richard Heath
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Daniel Maskell
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - Rebecca F Thompson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
- Life Sciences Electron Microscopy, Thermo Fisher Scientific, Leeds, UK
| | - Neil A Ranson
- Astbury Centre for Structural Molecular Biology, School of Molecular and Cellular Biology, University of Leeds, Leeds, LS2 9JT, UK
| | - James N Blaza
- Department of Chemistry, York Structural Biology Laboratory and York Biomedical Research Institute, University of York, Heslington, YO10 5DD, York, UK
| | - Jane A Endicott
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
| | - Martin E M Noble
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Marco Salamina
- Translational and Clinical Research Institute, Newcastle University Centre for Cancer, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK.
- Evotec (UK) Ltd., Milton, Abingdon, OX14 4RZ, UK.
| |
Collapse
|
4
|
Mughal MJ, Bhadresha K, Kwok HF. CDK inhibitors from past to present: A new wave of cancer therapy. Semin Cancer Biol 2023; 88:106-122. [PMID: 36565895 DOI: 10.1016/j.semcancer.2022.12.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022]
Abstract
Deregulation of the cell cycle machinery, which has been linked to dysregulation of cyclin-dependent kinases (CDKs), is a defining characteristic of cancer, eventually promoting abnormal proliferation that feeds tumorigenesis and disease development. In this regard, several CDK inhibitors (CDKIs) have been developed during the last few decades (1st, 2nd, and 3rd generation CDKIs) to inhibit cancer cell proliferation. 1st and 2nd generation CDKIs have not received much clinical attention for the treatment of cancer patients because of their limited specificity and high toxicity. However, the recent development of combination strategies allowed us to reduce the toxicity and side effects of these CDKIs, paving the way for their potential application in clinical settings. The 3rd generation CDKIs have yielded the most promising results at the preclinical and clinical levels, propelling them into the advanced stages of clinical trials against multiple malignancies, especially breast cancer, and revolutionizing traditional treatment strategies. In this review, we discuss the most-investigated candidates from the 1st, 2nd, and 3rd generations of CDKIs, their basic mechanisms of action, the reasons for their failure in the past, and their current clinical development for the treatment of different malignancies. Additionally, we briefly highlighted the most recent clinical trial results and advances in the development of 3rd generation FDA-approved selective CDK4/6 inhibitors that combat the most prevalent cancer. Overall, this review will provide a thorough knowledge of CDKIs from the past to the present, allowing researchers to rethink and develop innovative cancer therapeutic regimens.
Collapse
Affiliation(s)
- Muhammad Jameel Mughal
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; MOE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biochemistry and Molecular Medicine, School of Medicine and Health Sciences, George Washington University, Washington, DC, United States
| | - Kinjal Bhadresha
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Hematology/Oncology Division, School of Medicine, Indiana University Indianapolis, IN, United States
| | - Hang Fai Kwok
- Cancer Centre, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR; MOE Frontiers Science Center for Precision Oncology, University of Macau, Avenida de Universidade, Taipa, Macau SAR; Department of Biomedical Sciences, Faculty of Health Sciences, University of Macau, Avenida de Universidade, Taipa, Macau SAR.
| |
Collapse
|
5
|
Syahirah R, Hsu AY, Deng Q. A curious case of cyclin‐dependent kinases in neutrophils. J Leukoc Biol 2022; 111:1057-1068. [PMID: 35188696 PMCID: PMC9035055 DOI: 10.1002/jlb.2ru1021-573r] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 01/21/2022] [Accepted: 01/31/2022] [Indexed: 12/11/2022] Open
Abstract
Neutrophils are terminally differentiated, short-lived white blood cells critical for innate immunity. Although cyclin-dependent kinases (CDKs) are typically related to cell cycle progression, increasing evidence has shown that they regulate essential functions of neutrophils. This review highlights the roles of CDKs and their partners, cyclins, in neutrophils, outside of cell cycle regulation. CDK1-10 and several cyclins are expressed in neutrophils, albeit at different levels. Observed phenotypes associated with specific inhibition or genetic loss of CDK2 indicate its role in modulating neutrophil migration. CDK4 and 6 regulate neutrophil extracellular traps (NETs) formation, while CDK5 regulates neutrophil degranulation. CDK7 and 9 are critical in neutrophil apoptosis, contributing to inflammation resolution. In addition to the CDKs that regulate mature neutrophil functions, cyclins are essential in hematopoiesis and granulopoiesis. The pivotal roles of CDKs in neutrophils present an untapped potential in targeting CDKs for treating neutrophil-dominant inflammatory diseases and understanding the regulation of the neutrophil life cycle.
Collapse
Affiliation(s)
- Ramizah Syahirah
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
| | - Alan Y. Hsu
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
- Department of Pathology Harvard Medical School Boston Massachusetts USA
- Department of Laboratory Medicine The Stem Cell Program, Boston Children's Hospital Boston Massachusetts USA
| | - Qing Deng
- Department of Biological Sciences Purdue University West Lafayette Indiana USA
- Purdue Institute of Inflammation Immunology and Infectious Disease, Purdue University West Lafayette Indiana USA
- Purdue University Center for Cancer Research, Purdue University West Lafayette Indiana USA
| |
Collapse
|
6
|
Ke XG, Xiong YY, Yu B, Yuan C, Chen PY, Yang YF, Wu HZ. Mollugin induced oxidative DNA damage via up-regulating ROS that caused cell cycle arrest in hepatoma cells. Chem Biol Interact 2022; 353:109805. [PMID: 35007525 DOI: 10.1016/j.cbi.2022.109805] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/30/2021] [Accepted: 01/05/2022] [Indexed: 01/10/2023]
Abstract
Mollugin has been proven to have anti-tumor activity. However, its potential anti-tumor mechanism remains to be fully elaborated. Herein, we investigated the growth inhibition of HepG2 cells, as well as the anti-tumor effect of mollugin and its molecular mechanism on H22-tumor bearing mice. In vitro, mollugin was shown to have a strong inhibitory effect on HepG2 cells in a concentration-dependent manner. Mollugin induced S-phase arrest of HepG2 cells, and increased intracellular reactive oxygen species (ROS) levels. Comet assay demonstrated that mollugin induced DNA damage in HepG2 cells, as well as an increase in the expression of p-H2AX. In addition, mollugin induced changes in cyclin A2 and CDK2. However, the addition of antioxidant glutathione (GSH) was able to reverse the effect of mollugin. In vivo, mollugin significantly inhibited tumor growth and reduced the tendency of tumor volume growth in mice. The tumor cell density was found to be decreased in the administration group, and the content of ROS in the tumor tissue significantly increased. The expression of p-H2AX, cyclin A2 and CDK2 were consistent with in vitro results. Mollugin demonstrated anti-hepatocellular carcinoma activity in vitro and in vivo, and its anti-hepatocellular carcinoma activity was found to be related to DNA damage and cell cycle arrest induced by excessive ROS production in cells.
Collapse
Affiliation(s)
- Xin-Ge Ke
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yi-Yi Xiong
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Bing Yu
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Chong Yuan
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Peng-Yu Chen
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China
| | - Yan-Fang Yang
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Wuhan, 430065, China.
| | - He-Zhen Wu
- Faculty of Pharmacy, Hubei University of Chinese Medicine, Wuhan, 430065, China; Key Laboratory of Traditional Chinese Medicine Resources and Chemistry of Hubei Province, Wuhan, 430065, China.
| |
Collapse
|
7
|
Dai L, Prabhu N, Yu LY, Bacanu S, Ramos AD, Nordlund P. Horizontal Cell Biology: Monitoring Global Changes of Protein Interaction States with the Proteome-Wide Cellular Thermal Shift Assay (CETSA). Annu Rev Biochem 2019; 88:383-408. [DOI: 10.1146/annurev-biochem-062917-012837] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The cellular thermal shift assay (CETSA) is a biophysical technique allowing direct studies of ligand binding to proteins in cells and tissues. The proteome-wide implementation of CETSA with mass spectrometry detection (MS-CETSA) has now been successfully applied to discover targets for orphan clinical drugs and hits from phenotypic screens, to identify off-targets, and to explain poly-pharmacology and drug toxicity. Highly sensitive multidimensional MS-CETSA implementations can now also access binding of physiological ligands to proteins, such as metabolites, nucleic acids, and other proteins. MS-CETSA can thereby provide comprehensive information on modulations of protein interaction states in cellular processes, including downstream effects of drugs and transitions between different physiological cell states. Such horizontal information on ligandmodulation in cells is largely orthogonal to vertical information on the levels of different proteins and therefore opens novel opportunities to understand operational aspects of cellular proteomes.
Collapse
Affiliation(s)
- Lingyun Dai
- School of Biological Sciences, Nanyang Technological University, Singapore 138673;, ,
| | - Nayana Prabhu
- School of Biological Sciences, Nanyang Technological University, Singapore 138673;, ,
| | - Liang Ying Yu
- School of Biological Sciences, Nanyang Technological University, Singapore 138673;, ,
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden;, ,
| | - Smaranda Bacanu
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden;, ,
| | - Anderson Daniel Ramos
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden;, ,
| | - Pär Nordlund
- School of Biological Sciences, Nanyang Technological University, Singapore 138673;, ,
- Department of Oncology and Pathology, Karolinska Institutet, 171 77 Stockholm, Sweden;, ,
- Institute of Molecular and Cellular Biology, A*STAR, Singapore 138673
| |
Collapse
|
8
|
MiR-199b represses porcine muscle satellite cells proliferation by targeting JAG1. Gene 2019; 691:24-33. [DOI: 10.1016/j.gene.2018.12.052] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 12/17/2018] [Accepted: 12/21/2018] [Indexed: 02/02/2023]
|
9
|
Jindal A, Thadi A, Shailubhai K. Hepatocellular Carcinoma: Etiology and Current and Future Drugs. J Clin Exp Hepatol 2019; 9:221-232. [PMID: 31024205 PMCID: PMC6477125 DOI: 10.1016/j.jceh.2019.01.004] [Citation(s) in RCA: 171] [Impact Index Per Article: 28.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Accepted: 01/14/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is swiftly increasing in prevalence globally with a high mortality rate. The progression of HCC in patients is induced with advanced fibrosis, mainly cirrhosis, and hepatitis. The absence of proper preventive or curative treatment methods encouraged extensive research against HCC to develop new therapeutic strategies. The Food and Drug Administration-approved Nexavar (sorafenib) is used in the treatment of patients with unresectable HCC. In 2017, Stivarga (regorafenib) and Opdivo (nivolumab) got approved for patients with HCC after being treated with sorafenib, and in 2018, Lenvima (lenvatinib) got approved for patients with unresectable HCC. But, owing to the rapid drug resistance development and toxicities, these treatment options are not completely satisfactory. Therefore, there is an urgent need for new systemic combination therapies that target different signaling mechanisms, thereby decreasing the prospect of cancer cells developing resistance to treatment. In this review, HCC etiology and new therapeutic strategies that include currently approved drugs and other potential candidates of HCC such as Milciclib, palbociclib, galunisertib, ipafricept, and ramucirumab are evaluated.
Collapse
Key Words
- AMP, adenosine monophosphate
- AMPK, AMP-activated protein kinase
- ATP, adenosine 5′-triphosphate
- BMF, Bcl2 modifying factor
- BMI, body mass index
- CDK, cyclin-dependent kinase
- CTGF, connective tissue growth factor
- CTL, cytotoxic T lymphocyte
- CTLA, cytotoxic T-lymphocyte-associated protein
- ECM, extracellular matrix
- EFGR, endothelial growth factor receptor
- EGFR, epidermal growth factor receptor
- EMT, Epithelial–mesenchymal transition
- ERK, extracellular signal-regulated kinase
- FDA, Food and Drug Administration
- GFG, fibroblast growth factor
- HBV, hepatitis B virus
- HBcAg, hepatitis B core antibody
- HBsAg, HBV surface antigen
- HCC, Hepatocellular carcinoma
- HCV, hepatitis B virus
- HDV, hepatitis D virus
- HIF, hypoxia-inducible factor
- HIV, human immunodeficiency virus
- IGFR, insulin-like growth factor
- JAK, janus kinase
- MAPK, mitogen-activated protein kinase
- MDSC, myeloid-derived suppressor cell
- NASH, nonalcoholic steatohepatitis
- NK, natural killer
- NKT, natural killer T cell
- ORR, objective response rate
- OS, overall survival
- PAPSS1, 3′-phosphoadenosine 5′-phosphosulfate synthase 1
- PD-L1, programmed death ligand1
- PD1, programmed cell death protein 1
- PDGFR, platelet-derived growth factor receptor
- PEDF, pigment epithelium-derived factor
- PFS, progression-free survival
- PI3K, phosphoinositide 3-kinases
- PTEN, phosphatase and tensin homolog
- PUMA, p53 upregulated modulator of apoptosis
- RFA, radiofrequency ablation
- Rb, retinoblastoma protein
- SCF, stem cell factor
- SHP1, src homology 2 domain–containing phosphatase 1
- STAT3, signal transducer and activator of transcription 3
- TACE, transarterial chemoembolization
- TGF 1, transforming growth factor-1
- TK, tyrosine kinase
- TKI, Tyrosine kinase inhibitor
- TRKA, tropomyosin receptor kinase A
- Treg, regulatory T cells
- VEGF, vascular endothelial growth factor
- VEGFR, vascular endothelial growth factor receptor
- bFGF, basic fibroblast growth factor
- combination therapy
- cyclin-dependent kinase inhibitors
- hepatocellular carcinoma
- hepatology
- tyrosine kinase inhibitors
Collapse
Affiliation(s)
- Aastha Jindal
- Research and Development Center, Baruch S. Blumberg Institute, Doylestown, PA 18902, USA
- Address for correspondence: Aastha Jindal, Research and Development Center, Baruch S. Blumberg Institute, Doylestown, PA 18902, USA.
| | - Anusha Thadi
- Research and Development Center, Baruch S. Blumberg Institute, Doylestown, PA 18902, USA
| | - Kunwar Shailubhai
- Research and Development Center, Baruch S. Blumberg Institute, Doylestown, PA 18902, USA
- Research & Development, Tiziana Lifesciences, Doylestown, PA 18902, USA
| |
Collapse
|
10
|
García-Reyes B, Kretz AL, Ruff JP, von Karstedt S, Hillenbrand A, Knippschild U, Henne-Bruns D, Lemke J. The Emerging Role of Cyclin-Dependent Kinases (CDKs) in Pancreatic Ductal Adenocarcinoma. Int J Mol Sci 2018; 19:E3219. [PMID: 30340359 PMCID: PMC6214075 DOI: 10.3390/ijms19103219] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 09/27/2018] [Accepted: 10/11/2018] [Indexed: 02/07/2023] Open
Abstract
The family of cyclin-dependent kinases (CDKs) has critical functions in cell cycle regulation and controlling of transcriptional elongation. Moreover, dysregulated CDKs have been linked to cancer initiation and progression. Pharmacological CDK inhibition has recently emerged as a novel and promising approach in cancer therapy. This idea is of particular interest to combat pancreatic ductal adenocarcinoma (PDAC), a cancer entity with a dismal prognosis which is owed mainly to PDAC's resistance to conventional therapies. Here, we review the current knowledge of CDK biology, its role in cancer and the therapeutic potential to target CDKs as a novel treatment strategy for PDAC.
Collapse
Affiliation(s)
- Balbina García-Reyes
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Anna-Laura Kretz
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Jan-Philipp Ruff
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Silvia von Karstedt
- Department of Translational Genomics, University Hospital Cologne, Weyertal 115b, 50931 Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann-Straße 26, 50931 Cologne, Germany.
| | - Andreas Hillenbrand
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Doris Henne-Bruns
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Johannes Lemke
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| |
Collapse
|
11
|
Bayarkhangai B, Noureldin S, Yu L, Zhao N, Gu Y, Xu H, Guo C. A comprehensive and perspective view of oncoprotein SET in cancer. Cancer Med 2018; 7:3084-3094. [PMID: 29749127 PMCID: PMC6051184 DOI: 10.1002/cam4.1526] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 03/25/2018] [Accepted: 04/05/2018] [Indexed: 12/16/2022] Open
Abstract
SET is a multifunctional oncoprotein which is ubiquitously expressed in all kinds of cells. The SET protein participates in many cellular processes including cell cycle, cell migration, apoptosis, transcription, and DNA repair. Accumulating evidence demonstrates that the expression and activity of SET correlate with cancer occurrence, metastasis, and prognosis. Therefore, the SET protein is regarded as a potential target for cancer therapy and several inhibitors are being developed for clinical use. Herein, we comprehensively review the physiological and pathological functions of SET as well as its structure-function relationship. Additionally, the regulatory mechanisms of SET at both transcriptional and posttranslational levels are also discussed.
Collapse
Affiliation(s)
- Buuvee Bayarkhangai
- State Key of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Suzan Noureldin
- State Key of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Liting Yu
- State Key of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Na Zhao
- State Key of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Yaru Gu
- State Key of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Hanmei Xu
- State Key of Natural Medicine, China Pharmaceutical University, Nanjing, China
| | - Changying Guo
- State Key of Natural Medicine, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
12
|
Dai L, Zhao T, Bisteau X, Sun W, Prabhu N, Lim YT, Sobota RM, Kaldis P, Nordlund P. Modulation of Protein-Interaction States through the Cell Cycle. Cell 2018; 173:1481-1494.e13. [PMID: 29706543 DOI: 10.1016/j.cell.2018.03.065] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 01/25/2018] [Accepted: 03/26/2018] [Indexed: 11/25/2022]
Abstract
Global profiling of protein expression through the cell cycle has revealed subsets of periodically expressed proteins. However, expression levels alone only give a partial view of the biochemical processes determining cellular events. Using a proteome-wide implementation of the cellular thermal shift assay (CETSA) to study specific cell-cycle phases, we uncover changes of interaction states for more than 750 proteins during the cell cycle. Notably, many protein complexes are modulated in specific cell-cycle phases, reflecting their roles in processes such as DNA replication, chromatin remodeling, transcription, translation, and disintegration of the nuclear envelope. Surprisingly, only small differences in the interaction states were seen between the G1 and the G2 phase, suggesting similar hardwiring of biochemical processes in these two phases. The present work reveals novel molecular details of the cell cycle and establishes proteome-wide CETSA as a new strategy to study modulation of protein-interaction states in intact cells.
Collapse
Affiliation(s)
- Lingyun Dai
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Tianyun Zhao
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Xavier Bisteau
- Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore
| | - Wendi Sun
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Nayana Prabhu
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Yan Ting Lim
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore
| | - Radoslaw M Sobota
- Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore; Institute of Medical Biology, A(∗)STAR, Singapore 138648, Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore; Department of Biochemistry, National University of Singapore, Singapore 117597, Singapore
| | - Pär Nordlund
- School of Biological Sciences, Nanyang Technological University, Singapore 637551, Singapore; Institute of Molecular and Cell Biology, A(∗)STAR, Singapore 138673, Singapore; Department of Oncology and Pathology, Karolinska Institutet, 17177 Stockholm, Sweden.
| |
Collapse
|
13
|
Wang J, Tian Y, Chen H, Li H, Zheng S. Key signaling pathways, genes and transcription factors associated with hepatocellular carcinoma. Mol Med Rep 2018; 17:8153-8160. [PMID: 29658607 PMCID: PMC5983994 DOI: 10.3892/mmr.2018.8871] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Accepted: 04/03/2018] [Indexed: 01/30/2023] Open
Abstract
The purpose of the present study was to investigate the underlying molecular mechanism of hepatocellular carcinoma (HCC) using bioinformatics approaches. The microarray dataset GSE64041 was downloaded from the Gene Expression Omnibus database, which included 60 tumor liver samples and 60 matched control samples. Differentially expressed genes (DEGs) between HCC and control groups were identified. Then functional enrichment analyses, protein‑protein interaction (PPI) network, sub‑network and integrated transcription factor (TF)‑microRNA (miRNA)‑target network analyses were performed for these DEGs. A total of 378 DEGs were obtained, including 101 upregulated and 277 downregulated DEGs. In addition, functional enrichment analysis for DEGs in the sub‑network revealed 'cell division' and 'cell cycle' as key Gene Ontology (GO) terms and pathways. Topoisomerase (DNA) IIα (TOP2A) and integrin subunit α2 (ITGA2) were hub nodes in the PPI network. TOP2A, cyclin dependent kinase 1 (CDK1) and polo like kinase 1 (PLK1) were revealed to be hub nodes in the sub‑network. Finally, 4 TFs including forkhead box M1 (FOXM1), E2F transcription factor 4 (E2F4), SIN3 transcription regulator family member A (SIN3A) and transcription factor 7 like 1 (TCF7L1) were obtained through integrated network analysis. TOP2A, ITGA2, PLK1 and CDK1 may be key genes involved in HCC development. 'Cell division' and 'cell cycle' were indicated to act as key GO terms and Kyoto Encyclopedia of Genes and Genomes pathways in HCC. In addition, FOXM1, TCF7L1, E2F4 and SIN3A were revealed to be key TFs associated with HCC.
Collapse
Affiliation(s)
- Jingcheng Wang
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| | - Yang Tian
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| | - Hui Chen
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| | - Hui Li
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| | - Shusen Zheng
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University, Key Laboratory of Combined Multi‑Organ Transplantation, Ministry of Public Health Key Laboratory of Organ Transplantation, Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
14
|
Liu X, Tan N, Liao H, Pan G, Xu Q, Zhu R, Zou L, He S, Zhu H. High GSTP1 inhibits cell proliferation by reducing Akt phosphorylation and is associated with a better prognosis in hepatocellular carcinoma. Oncotarget 2017; 9:8957-8971. [PMID: 29507666 PMCID: PMC5823662 DOI: 10.18632/oncotarget.23420] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Accepted: 09/21/2017] [Indexed: 12/15/2022] Open
Abstract
Glutathione S-transferase (GST) family members promote carcinogenesis and cancer progression. We assessed GST pi 1 (GSTP1) mRNA and protein levels in hepatocellular carcinoma (HCC) using genome databases and tissue microarray (TMA) technology. We found that in cancerous tissues, GSTP1 mRNA was down-regulated in genome databases, and immunohistochemical staining of GSTP1 in 237 HCC cases varied from negative to strongly positive. GSTP1 levels correlated negatively with tumor size and serum alpha-fetoprotein (AFP) in HCC patients, and higher GSTP1 levels associated with longer overall survival (OS) and disease-free survival (DFS). We also found that GSTP1 overexpression restrained HepG2 and Huh7 liver cancer cell proliferation in vivo and in vitro. GSTP1 arrested the cell cycle at G1/S by up-regulating p21 and p27 and down-regulating p-Akt. Interrupting GSTP1 gene expression promoted liver cancer cell proliferation and increased the percentage of cells in S phase by decreasing levels of p21 and p27 and increasing p-Akt. These results suggest high GSTP1 levels provide a better prognosis through suppression of tumorigenesis in HCC.
Collapse
Affiliation(s)
- Xiaojia Liu
- Department of Pathology, Basic Medical School, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Division of Surgical Pathology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Ning Tan
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin 541001, China
| | - Hongtao Liao
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin 541001, China
| | - Guangdong Pan
- Department of Hepatobiliary Surgery, The People's Hospital of Liuzhou, Liuzhou 545001, China
| | - Qing Xu
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin 541001, China
| | - Rong Zhu
- Department of Pathology, Basic Medical School, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Liping Zou
- Department of Pathology, Basic Medical School, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Division of Surgical Pathology, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Songqing He
- Laboratory of Liver Injury and Repair Molecular Medicine, Guilin Medical University, Guilin 541001, China.,Department of Hepatobiliary Surgery, Affiliated Hospital of Guangxi Medical University, Nanning 530021, China
| | - Hongguang Zhu
- Department of Pathology, Basic Medical School, Shanghai Medical College, Fudan University, Shanghai 200032, China.,Division of Surgical Pathology, Huashan Hospital, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
MiR-34c represses muscle development by forming a regulatory loop with Notch1. Sci Rep 2017; 7:9346. [PMID: 28839212 PMCID: PMC5571228 DOI: 10.1038/s41598-017-09688-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/19/2017] [Indexed: 11/08/2022] Open
Abstract
Since pork accounts for about 40% of global meat consumption, the pig is an important economic animal for meat production. Pig is also a useful medical model for humans due to its similarity in size and physiology. Understanding the mechanism of muscle development has great implication for animal breeding and human health. Previous studies showed porcine muscle satellite cells (PSCs) are important for postnatal skeletal muscle growth, and Notch1 signaling pathway and miRNAs regulate the skeletal muscle development. Notch1 signal pathway regulates the transcription of certain types of miRNAs which further affects target gene expression. However, the specific relationship between Notch1 and miRNAs during muscle development has not been established. We found miR-34c is decreased in PSCs overexpressed N1ICD. Through the overexpression and inhibition of mi-34c, we demonstrated that miR-34c inhibits PSCs proliferation and promotes PSCs differentiation. Using dual-luciferase reporter assay and Chromatin immunoprecipitation, we demonstrate there is a reciprocal regulatory loop between Notch1 and miR-34c. Furthermore, injection of miR-34c lentivirus into mice caused repression of gastrocnemius muscle development. In summary, our data revealed that miR-34c can form a regulatory loop with Notch1 to repress muscle development, and this result expands our understanding of muscle development mechanism.
Collapse
|
16
|
Cdc25A Is a Critical Mediator of Ischemic Neuronal Death In Vitro and In Vivo. J Neurosci 2017; 37:6729-6740. [PMID: 28607169 DOI: 10.1523/jneurosci.3017-16.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 05/26/2017] [Accepted: 05/30/2017] [Indexed: 11/21/2022] Open
Abstract
Dysregulation of cell cycle machinery is implicated in a number of neuronal death contexts, including stroke. Increasing evidence suggests that cyclin-dependent kinases (Cdks) are inappropriately activated in mature neurons under ischemic stress conditions. We previously demonstrated a functional role for the cyclin D1/Cdk4/pRb (retinoblastoma tumor suppressor protein) pathway in delayed neuronal death induced by ischemia. However, the molecular signals leading to cyclin D/Cdk4/pRb activation following ischemic insult are presently not clear. Here, we investigate the cell division cycle 25 (Cdc25) dual-specificity phosphatases as potential upstream regulators of ischemic neuronal death and Cdk4 activation. We show that a pharmacologic inhibitor of Cdc25 family members (A, B, and C) protects mouse primary neurons from hypoxia-induced delayed death. The major contributor to the death process appears to be Cdc25A. shRNA-mediated knockdown of Cdc25A protects neurons in a delayed model of hypoxia-induced death in vitro Similar results were observed in vivo following global ischemia in the rat. In contrast, neurons singly or doubly deficient for Cdc25B/C were not significantly protective. We show that Cdc25A activity, but not level, is upregulated in vitro following hypoxia and global ischemic insult in vivo Finally, we show that shRNA targeting Cdc25A blocks Ser795 pRb phosphorylation. Overall, our results indicate a role for Cdc25A in delayed neuronal death mediated by ischemia.SIGNIFICANCE STATEMENT A major challenge in stroke is finding an effective neuroprotective strategy to treat cerebral ischemic injury. Cdc25 family member A (Cdc25A) is a phosphatase normally activated during cell division in proliferating cells. We found that Cdc25A is activated in neurons undergoing ischemic stress mediated by hypoxia in vitro and global cerebral ischemia in rats in vivo We show that pharmacologic or genetic inhibition of Cdc25A activity protects neurons from delayed death in vitro and in vivo Downregulation of Cdc25A led to reduction in retinoblastoma tumor suppressor protein (pRb) phosphorylation. An increase in pRb phosphorylation has been previously linked to ischemic neuronal death. Our results identify Cdc25A as a potential target for neuroprotectant strategy for the treatment of delayed ischemic neuronal death.
Collapse
|
17
|
Chang CT, Hseu YC, Thiyagarajan V, Huang HC, Hsu LS, Huang PJ, Liu JY, Liao JW, Yang HL. Antrodia salmonea induces G 2 cell-cycle arrest in human triple-negative breast cancer (MDA-MB-231) cells and suppresses tumor growth in athymic nude mice. JOURNAL OF ETHNOPHARMACOLOGY 2017; 196:9-19. [PMID: 27986611 DOI: 10.1016/j.jep.2016.12.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 10/19/2016] [Accepted: 12/10/2016] [Indexed: 06/06/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Antrodia salmonea (AS), is a well-known folk medicinal mushroom in Taiwan, has been reported to exhibit anti-oxidant, anti-angiogenic, and anti-inflammatory effects. MATERIALS AND METHODS In the present study, we examined the effects of AS on cell-cycle arrest in vitro in MDA-MB-231 cells and on tumor regression in vivo using an athymic nude mice model. RESULTS AS (0-200μg/mL) treatment significantly induced G2 cell-cycle arrest in MDA-MB-231 cells by reducing the levels of cyclin B1, cyclin A, cyclin E, and CDC2 proteins. In addition, N-acetylcysteine (NAC) pretreatment prevented AS induced G2 cell-cycle arrest, indicating that ROS accumulation and subsequent cell cycle arrest might be a major mechanism of AS-induced cytotoxicity. Further, AS treatment decreased COX-2 expression and induced PARP cleavage was significantly reversed by NAC pretreatment in MDA-MB-231 cells. The in vivo study results revealed that AS treatment was effective in terms of delaying the tumor incidence and reducing the tumor growth in MDA-MB-231-xenografted nude mice. TUNEL assay, immunohistochemical staining and Western blotting confirmed that AS significantly modulated the xenografted tumor progression as demonstrated by induction of apoptosis, autophagy, and cell-cycle arrest. CONCLUSION Our data strongly suggest that Antrodia salmonea could be an anti-cancer agent for human breast cancer.
Collapse
Affiliation(s)
- Chia-Ting Chang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung 40402, Taiwan
| | - You-Cheng Hseu
- Department of Cosmeceutics,Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung 40402, Taiwan; Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Varadharajan Thiyagarajan
- Department of Cosmeceutics,Cosmeceutics, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung 40402, Taiwan
| | - Hui-Chi Huang
- School of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung 40402, Taiwan
| | - Li-Sung Hsu
- Institute of Biochemistry and Biotechnology, Chung Shan Medical University, 40402 Taichung, Taiwan
| | - Pei-Jane Huang
- Department of Health and Nutrition Biotechnology, Asia University, Taichung 41354, Taiwan
| | - Jer-Yuh Liu
- Graduate Institute of Cancer Biology, China Medical University, Taichung 40402, Taiwan
| | - Jiunn-Wang Liao
- Graduate Institute of Veterinary Pathology, National Chung Hsing University, Taichung 402, Taiwan
| | - Hsin-Ling Yang
- Institute of Nutrition, College of Biopharmaceutical and Food Sciences, China Medical University, Taichung 40402, Taiwan.
| |
Collapse
|
18
|
Affiliation(s)
- Philipp Kaldis
- Cell Division and Cancer Research, Institute of Molecular and Cell Biology, Agency for Science, Technology and ResearchSingapore, Singapore; Department of Biochemistry, National University of SingaporeSingapore, Singapore
| |
Collapse
|
19
|
Czerwinska AM, Nowacka J, Aszer M, Gawrzak S, Archacka K, Fogtman A, Iwanicka-Nowicka R, Jańczyk-Ilach K, Koblowska M, Ciemerych MA, Grabowska I. Cell cycle regulation of embryonic stem cells and mouse embryonic fibroblasts lacking functional Pax7. Cell Cycle 2016; 15:2931-2942. [PMID: 27610933 PMCID: PMC5105925 DOI: 10.1080/15384101.2016.1231260] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The transcription factor Pax7 plays a key role during embryonic myogenesis and in adult organisms in that it sustains the proper function of satellite cells, which serve as adult skeletal muscle stem cells. Recently we have shown that lack of Pax7 does not prevent the myogenic differentiation of pluripotent stem cells. In the current work we show that the absence of functional Pax7 in differentiating embryonic stem cells modulates cell cycle facilitating their proliferation. Surprisingly, deregulation of Pax7 function also positively impacts at the proliferation of mouse embryonic fibroblasts. Such phenotypes seem to be executed by modulating the expression of positive cell cycle regulators, such as cyclin E.
Collapse
Affiliation(s)
- Areta M Czerwinska
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Joanna Nowacka
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Magdalena Aszer
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Sylwia Gawrzak
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Karolina Archacka
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Anna Fogtman
- b Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Warsaw , Poland
| | - Roksana Iwanicka-Nowicka
- b Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Warsaw , Poland.,c Department of Systems Biology , Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Katarzyna Jańczyk-Ilach
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Marta Koblowska
- b Laboratory of Microarray Analysis, Institute of Biochemistry and Biophysics, Polish Academy of Sciences , Warsaw , Poland.,c Department of Systems Biology , Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Maria A Ciemerych
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| | - Iwona Grabowska
- a Department of Cytology , Institute of Zoology, Faculty of Biology, University of Warsaw , Warsaw , Poland
| |
Collapse
|
20
|
Abstract
The preimplantation development stage of mammalian embryogenesis consists of a series of highly conserved, regulated, and predictable cell divisions. This process is essential to allow the rapid expansion and differentiation of a single-cell zygote into a multicellular blastocyst containing cells of multiple developmental lineages. This period of development, also known as the germinal stage, encompasses several important developmental transitions, which are accompanied by dramatic changes in cell cycle profiles and dynamics. These changes are driven primarily by differences in the establishment and enforcement of cell cycle checkpoints, which must be bypassed to facilitate the completion of essential cell cycle events. Much of the current knowledge in this area has been amassed through the study of knockout models in mice. These mouse models are powerful experimental tools, which have allowed us to dissect the relative dependence of the early embryonic cell cycles on various aspects of the cell cycle machinery and highlight the extent of functional redundancy between members of the same gene family. This chapter will explore the ways in which the cell cycle machinery, their accessory proteins, and their stimuli operate during mammalian preimplantation using mouse models as a reference and how this allows for the usually well-defined stages of the cell cycle to be shaped and transformed during this unique and critical stage of development.
Collapse
|
21
|
Pinhero R, Yankulov K. Expression and Purification of Recombinant CDKs: CDK7, CDK8, and CDK9. Methods Mol Biol 2016; 1336:13-28. [PMID: 26231705 DOI: 10.1007/978-1-4939-2926-9_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Cyclin-dependent kinases have established roles in the regulation of cell cycle, in gene expression and in cell differentiation. Many of these kinases have been considered as drug targets and numerous efforts have been made to develop specific and potent inhibitors against them. The first step in all of these attempts and in many other biochemical analyses is the production of highly purified and reliable kinase, most frequently in a recombinant form. In this chapter we describe our experience in the cloning, expression, and purification of CDKs using CDK7/CycH, CDK8/CycC, and CDK9/CycT1 as an example.
Collapse
Affiliation(s)
- Reena Pinhero
- Department of Molecular Biology and Genetics, University of Guelph, 50 Stone Road East, Guelph, ON, Canada, N1G 2W1
| | | |
Collapse
|
22
|
Abstract
Neurons are usually regarded as postmitotic cells that undergo apoptosis in response to cell cycle reactivation. Nevertheless, recent evidence indicates the existence of a defined developmental program that induces DNA replication in specific populations of neurons, which remain in a tetraploid state for the rest of their adult life. Similarly, de novo neuronal tetraploidization has also been described in the adult brain as an early hallmark of neurodegeneration. The aim of this review is to integrate these recent developments in the context of cell cycle regulation and apoptotic cell death in neurons. We conclude that a variety of mechanisms exists in neuronal cells for G1/S and G2/M checkpoint regulation. These mechanisms, which are connected with the apoptotic machinery, can be modulated by environmental signals and the neuronal phenotype itself, thus resulting in a variety of outcomes ranging from cell death at the G1/S checkpoint to full proliferation of differentiated neurons.
Collapse
Key Words
- AD, Alzheimer disease
- BDNF, brain-derived neurotrophic factor
- BrdU, 5-bromo-2′-deoxyuridine
- CKI, Cdk-inhibitor
- CNS, central nervous system
- Cdk, cyclin-dependent kinase
- Cip/Kip, cyclin inhibitor protein/kinase inhibitor protein
- G0, quiescent state
- G1, growth phase 1
- G2, growth phase 2
- Ink, inhibitor of kinase
- Mcm2, minichromosome maintenance 2
- PCNA, proliferating cell nuclear antigen
- PD, Parkinson disease
- RGCs, retinal ganglion cells
- Rb, Retinoblastoma
- S-phase
- S-phase, synthesis phase.
- apoptosis
- cell cycle re-entry
- mitosis
- neuron
- p38MAPK, p38 mitogen-activated protein kinase
- p75NTR, neurotrophin receptor p75
- tetraploid
Collapse
Affiliation(s)
- José M Frade
- a Department of Molecular, Cellular and Developmental Neurobiology; Instituto Cajal; Consejo Superior de Investigaciones Científicas (IC-CSIC) ; Madrid , Spain
| | | |
Collapse
|
23
|
Herrero-Ruiz J, Mora-Santos M, Giráldez S, Sáez C, Japón MA, Tortolero M, Romero F. βTrCP controls the lysosome-mediated degradation of CDK1, whose accumulation correlates with tumor malignancy. Oncotarget 2015; 5:7563-74. [PMID: 25149538 PMCID: PMC4202144 DOI: 10.18632/oncotarget.2274] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
In mammals, cell cycle progression is controlled by cyclin-dependent kinases, among which CDK1 plays important roles in the regulation of the G2/M transition, G1 progression and G1/S transition. CDK1 is highly regulated by its association to cyclins, phosphorylation and dephosphorylation, changes in subcellular localization, and by direct binding of CDK inhibitor proteins. CDK1 steady-state protein levels are held constant throughout the cell cycle by a coordinated regulation of protein synthesis and degradation. We show that CDK1 is ubiquitinated by the E3 ubiquitin ligase SCFβTrCP and degraded by the lysosome. Furthermore, we found that DNA damage not only triggers the stabilization of inhibitory phosphorylation sites on CDK1 and repression of CDK1 gene expression, but also regulates βTrCP-induced CDK1 degradation in a cell type-dependent manner. Specifically, treatment with the chemotherapeutic agent doxorubicin in certain cell lines provokes CDK1 degradation and induces apoptosis, whereas in others it inhibits destruction of the protein. These observations raise the possibility that different tumor types, depending on their pathogenic spectrum mutations, may display different sensitivity to βTrCP-induced CDK1 degradation after DNA damage. Finally, we found that CDK1 accumulation in patients’ tumors shows a negative correlation with βTrCP and a positive correlation with the degree of tumor malignancy.
Collapse
Affiliation(s)
- Joaquín Herrero-Ruiz
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Mar Mora-Santos
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Servando Giráldez
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Carmen Sáez
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Anatomía Patológica, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Miguel A Japón
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla and Departamento de Anatomía Patológica, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Maria Tortolero
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Francisco Romero
- Departamento de Microbiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
24
|
Dupont CA, Dardalhon-Cuménal D, Kyba M, Brock HW, Randsholt NB, Peronnet F. Drosophila Cyclin G and epigenetic maintenance of gene expression during development. Epigenetics Chromatin 2015; 8:18. [PMID: 25995770 PMCID: PMC4438588 DOI: 10.1186/s13072-015-0008-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 04/01/2015] [Indexed: 12/31/2022] Open
Abstract
Background Cyclins and cyclin-dependent kinases (CDKs) are essential for cell cycle regulation and are functionally associated with proteins involved in epigenetic maintenance of transcriptional patterns in various developmental or cellular contexts. Epigenetic maintenance of transcription patterns, notably of Hox genes, requires the conserved Polycomb-group (PcG), Trithorax-group (TrxG), and Enhancer of Trithorax and Polycomb (ETP) proteins, particularly well studied in Drosophila. These proteins form large multimeric complexes that bind chromatin and appose or recognize histone post-translational modifications. PcG genes act as repressors, counteracted by trxG genes that maintain gene activation, while ETPs interact with both, behaving alternatively as repressors or activators. Drosophila Cyclin G negatively regulates cell growth and cell cycle progression, binds and co-localizes with the ETP Corto on chromatin, and participates with Corto in Abdominal-B Hox gene regulation. Here, we address further implications of Cyclin G in epigenetic maintenance of gene expression. Results We show that Cyclin G physically interacts and extensively co-localizes on chromatin with the conserved ETP Additional sex combs (ASX), belonging to the repressive PR-DUB complex that participates in H2A deubiquitination and Hox gene silencing. Furthermore, Cyclin G mainly co-localizes with RNA polymerase II phosphorylated on serine 2 that is specific to productive transcription. CycG interacts with Asx, PcG, and trxG genes in Hox gene maintenance, and behaves as a PcG gene. These interactions correlate with modified ectopic Hox protein domains in imaginal discs, consistent with a role for Cyclin G in PcG-mediated Hox gene repression. Conclusions We show here that Drosophila CycG is a Polycomb-group gene enhancer, acting in epigenetic maintenance of the Hox genes Sex combs reduced (Scr) and Ultrabithorax (Ubx). However, our data suggest that Cyclin G acts alternatively as a transcriptional activator or repressor depending on the developmental stage, the tissue or the target gene. Interestingly, since Cyclin G interacts with several CDKs, Cyclin G binding to the ETPs ASX or Corto suggests that their activity could depend on Cyclin G-mediated phosphorylation. We discuss whether Cyclin G fine-tunes transcription by controlling H2A ubiquitination and transcriptional elongation via interaction with the ASX subunit of PR-DUB. Electronic supplementary material The online version of this article (doi:10.1186/s13072-015-0008-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Camille A Dupont
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR 7622, Developmental Biology, 9, quai Saint-Bernard, F-75005 Paris, France ; CNRS, IBPS, UMR 7622, Developmental Biology, 9, quai Saint-Bernard, F-75005 Paris, France
| | - Delphine Dardalhon-Cuménal
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR 7622, Developmental Biology, 9, quai Saint-Bernard, F-75005 Paris, France ; CNRS, IBPS, UMR 7622, Developmental Biology, 9, quai Saint-Bernard, F-75005 Paris, France
| | - Michael Kyba
- Lillehei Heart Institute and Department of Pediatrics, University of Minnesota, 2231 6th Street SE, Minneapolis, MN 55455 USA
| | - Hugh W Brock
- Department of Zoology, University of British Columbia, 6270 University Boulevard, V6T 1Z4 Vancouver, BC Canada
| | - Neel B Randsholt
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR 7622, Developmental Biology, 9, quai Saint-Bernard, F-75005 Paris, France ; CNRS, IBPS, UMR 7622, Developmental Biology, 9, quai Saint-Bernard, F-75005 Paris, France
| | - Frédérique Peronnet
- Sorbonne Universités, UPMC Univ Paris 06, Institut de Biologie Paris-Seine (IBPS), UMR 7622, Developmental Biology, 9, quai Saint-Bernard, F-75005 Paris, France ; CNRS, IBPS, UMR 7622, Developmental Biology, 9, quai Saint-Bernard, F-75005 Paris, France
| |
Collapse
|
25
|
Takeda S, Sasagawa S, Oyama T, Searleman AC, Westergard TD, Cheng EH, Hsieh JJ. Taspase1-dependent TFIIA cleavage coordinates head morphogenesis by limiting Cdkn2a locus transcription. J Clin Invest 2015; 125:1203-14. [PMID: 25664857 DOI: 10.1172/jci77075] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2014] [Accepted: 01/05/2015] [Indexed: 01/06/2023] Open
Abstract
Head morphogenesis requires complex signal relays to enable precisely coordinated proliferation, migration, and patterning. Here, we demonstrate that, during mouse head formation, taspase1-mediated (TASP1-mediated) cleavage of the general transcription factor TFIIA ensures proper coordination of rapid cell proliferation and morphogenesis by maintaining limited transcription of the negative cell cycle regulators p16Ink4a and p19Arf from the Cdkn2a locus. In mice, loss of TASP1 function led to catastrophic craniofacial malformations that were associated with inadequate cell proliferation. Compound deficiency of Cdkn2a, especially p16Ink4a deficiency, markedly reduced the craniofacial anomalies of TASP1-deficent mice. Furthermore, evaluation of mice expressing noncleavable TASP1 targets revealed that TFIIA is the principal TASP1 substrate that orchestrates craniofacial morphogenesis. ChIP analyses determined that noncleaved TFIIA accumulates at the p16Ink4a and p19Arf promoters to drive transcription of these negative regulators. In summary, our study elucidates a regulatory circuit comprising proteolysis, transcription, and proliferation that is pivotal for construction of the mammalian head.
Collapse
|
26
|
Pino MTL, Verstraeten SV. Tl(I) and Tl(III) alter the expression of EGF-dependent signals and cyclins required for pheochromocytoma (PC12) cell-cycle resumption and progression. J Appl Toxicol 2014; 35:952-69. [PMID: 25534134 DOI: 10.1002/jat.3096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 10/16/2014] [Accepted: 10/31/2014] [Indexed: 01/06/2023]
Abstract
The effects of thallium [Tl(I) and Tl(III)] on the PC12 cell cycle were evaluated without (EGF(-)) or with (EGF(+)) media supplementation with epidermal growth factor (EGF). The following markers of cell-cycle phases were analyzed: cyclin D1 (G1 ); E2F-1, cyclin E and cytosolic p21 (G1 →S transition); nuclear PCNA and cyclin A (S); and cyclin B1 (G2). The amount of cells in each phase and the activation of the signaling cascade triggered by EGF were also analyzed. Tl(I) and Tl(III) (5-100 μM) caused dissimilar effects on PC12 cell proliferation. In EGF(-) cells, Tl(I) increased the expression of G1 →S transition markers and nuclear PCNA, without affecting cyclin A or cyclin B1. In addition to those, cyclin B1 was also increased in EGF(+) cells. In EGF(-) cells, Tl(III) increased the expression of cyclin D1, all the G1→S and S phase markers and cyclin B1. In EGF(+) cells, Tl(III) increased cyclin D1 expression and decreased all the markers of G1 →S transition and the S phase. Even when these cations did not induce the activation of EGF receptor (EGFR) in EGF(-) cells, they promoted the phosphorylation of ERK1/2 and Akt. In the presence of EGF, the cations anticipated EGFR phosphorylation without affecting the kinetics of EGF-dependent ERK1/2 and Akt phosphorylation. Altogether, results indicate that Tl(I) promoted cell proliferation in both EGF(-) and EGF(+) cells. In contrast, Tl(III) promoted the proliferation of EGF(-) cells but delayed it in EGF(+) cells, which may be related to the toxic effects of this cation in PC12 cells.
Collapse
Affiliation(s)
- María T L Pino
- Department of Biological Chemistry, IQUIFIB (UBA-CONICET), School of Pharmacy and Biochemistry, University of Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
| | - Sandra V Verstraeten
- Department of Biological Chemistry, IQUIFIB (UBA-CONICET), School of Pharmacy and Biochemistry, University of Buenos Aires, Junín 956, C1113AAD, Buenos Aires, Argentina
| |
Collapse
|
27
|
Ren J, Yang J, Xu Y, Huang Q, Yang M, Hu K. Lupiwighteone induces cell cycle arrest and apoptosis and activates the Nrf2/ARE pathway in human neuroblastoma cells. Biomed Pharmacother 2014; 69:153-61. [PMID: 25661352 DOI: 10.1016/j.biopha.2014.11.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 11/09/2014] [Indexed: 12/22/2022] Open
Abstract
Lupiwighteone (Lup) is a kind of natural isoflavone, but its pharmacological effect and active mechanism are rarely reported. This study aimed to investigate the anticancer and cancer preventive effects of Lup on human neuroblastoma (SH-SY5Y) cells. We found that Lup could inhibit SH-SY5Y cells growth in a concentration- and time-dependent manner. Further studies suggested that Lup could induce G2/M phase arrest associated with an evident decrease in cyclin B1/D1 and cyclin dependent kinase (CDK) 1/2/4/6 protein expressions. Moreover, Lup could regulate the changes of mitochondrial membrane potential and increase intracellular reactive oxygen species (ROS) production. After the cells were treated with Lup, topical morphological characteristics were observed; apoptosis-related protein expressions, such as Bax, cytochrome c, cleaved caspase-9, cleaved caspase-3 and cleaved PARP-1 were increased; and protein expressions, such as Bcl-2, procaspase-9, PARP-1 and P-Akt were decreased. These changes were observed simultaneously. In addition, Nrf2 transcription factor activation was detected by an ARE-GFP reporter assay. Nrf2 nuclear localization was then investigated using a fluorescence microscope. Furthermore, Nrf2 and Keap1 protein levels were determined by western blot. Our results may provide a scientific basis for the application of the anticancer and cancer preventive effects of Lup on SH-SY5Y cells.
Collapse
Affiliation(s)
- Jie Ren
- School of Pharmaceutical Engineering & Life Science, Changzhou University, 1, Gehu Road, Changzhou, Jiangsu 213164, PR China.
| | - Jie Yang
- School of Pharmaceutical Engineering & Life Science, Changzhou University, 1, Gehu Road, Changzhou, Jiangsu 213164, PR China
| | - Yuanyuan Xu
- School of Pharmaceutical Engineering & Life Science, Changzhou University, 1, Gehu Road, Changzhou, Jiangsu 213164, PR China
| | - Qianhui Huang
- School of Pharmaceutical Engineering & Life Science, Changzhou University, 1, Gehu Road, Changzhou, Jiangsu 213164, PR China
| | - Meng Yang
- School of Pharmaceutical Engineering & Life Science, Changzhou University, 1, Gehu Road, Changzhou, Jiangsu 213164, PR China
| | - Kun Hu
- School of Pharmaceutical Engineering & Life Science, Changzhou University, 1, Gehu Road, Changzhou, Jiangsu 213164, PR China.
| |
Collapse
|
28
|
Cachat E, Liu W, Hohenstein P, Davies JA. A library of mammalian effector modules for synthetic morphology. J Biol Eng 2014; 8:26. [PMID: 25478005 PMCID: PMC4255936 DOI: 10.1186/1754-1611-8-26] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/02/2014] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND In mammalian development, the formation of most tissues is achieved by a relatively small repertoire of basic morphogenetic events (e.g. cell adhesion, locomotion, apoptosis, etc.), permutated in various sequences to form different tissues. Together with cell differentiation, these mechanisms allow populations of cells to organize themselves into defined geometries and structures, as simple embryos develop into complex organisms. The control of tissue morphogenesis by populations of engineered cells is a potentially very powerful but neglected aspect of synthetic biology. RESULTS We have assembled a modular library of synthetic morphogenetic driver genes to control (separately) mammalian cell adhesion, locomotion, fusion, proliferation and elective cell death. Here we describe this library and demonstrate its use in the T-REx-293 human cell line to induce each of these desired morphological behaviours on command. CONCLUSIONS Building on from the simple test systems described here, we want to extend engineered control of morphogenetic cell behaviour to more complex 3D structures that can inform embryologists and may, in the future, be used in surgery and regenerative medicine, making synthetic morphology a powerful tool for developmental biology and tissue engineering.
Collapse
Affiliation(s)
- Elise Cachat
- University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
| | - Weijia Liu
- University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
| | - Peter Hohenstein
- The Roslin Institute, University of Edinburgh, Easter Bush Campus, Midlothian, EH25 9RG UK
| | - Jamie A Davies
- University of Edinburgh, Centre for Integrative Physiology, Hugh Robson Building, George Square, Edinburgh, EH8 9XD UK
| |
Collapse
|
29
|
Tao L, Fu R, Wang X, Yao J, Zhou Y, Dai Q, Li Z, Lu N, Wang W. LL-202, a newly synthesized flavonoid, inhibits tumor growth via inducing G2/M phase arrest and cell apoptosis in MCF-7 human breast cancer cells in vitro and in vivo. Toxicol Lett 2014; 228:1-12. [DOI: 10.1016/j.toxlet.2014.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/31/2014] [Accepted: 04/03/2014] [Indexed: 10/25/2022]
|
30
|
Li S, Song W, Jiang M, Zeng L, Zhu X, Chen J. Phosphorylation of cyclin Y by CDK14 induces its ubiquitination and degradation. FEBS Lett 2014; 588:1989-96. [PMID: 24794231 DOI: 10.1016/j.febslet.2014.04.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Revised: 04/05/2014] [Accepted: 04/13/2014] [Indexed: 02/06/2023]
Abstract
Cyclin Y, a membrane associated cyclin, is capable of binding and activating CDK14. Here we report that human cyclin Y (CCNY) is a phosphoprotein in vivo and that phosphorylation of CCNY by CDK14 triggers its ubiquitination and degradation. Inactivation of either CDK14 or Cul1 results in accumulation of CCNY. An in vivo and in vitro mapping of CCNY phosphorylation sites by mass spectrometry revealed that the flanking regions of the conserved cyclin box are heavily phosphorylated. Phosphorylation of CCNY at Serines 71 and 73 creates a putative phospho-degron that controls its association with an SCF complex. Mutation of serine to alanine at these two sites stabilized CCNY and enhanced the activity of CCNY/CDK14 on phosphorylation of LRP6. Our results provide insight into autoregulation of the cyclin Y/CDK14 pair in CDK14 activation and cyclin Y turnover which is a process that is involved in membrane proximal signaling.
Collapse
Affiliation(s)
- Shan Li
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Wei Song
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Mei Jiang
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Liyong Zeng
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Xueliang Zhu
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China
| | - Jiangye Chen
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China.
| |
Collapse
|
31
|
The Complex Relationship between Liver Cancer and the Cell Cycle: A Story of Multiple Regulations. Cancers (Basel) 2014; 6:79-111. [PMID: 24419005 PMCID: PMC3980619 DOI: 10.3390/cancers6010079] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 12/24/2013] [Accepted: 01/03/2014] [Indexed: 12/14/2022] Open
Abstract
The liver acts as a hub for metabolic reactions to keep a homeostatic balance during development and growth. The process of liver cancer development, although poorly understood, is related to different etiologic factors like toxins, alcohol, or viral infection. At the molecular level, liver cancer is characterized by a disruption of cell cycle regulation through many molecular mechanisms. In this review, we focus on the mechanisms underlying the lack of regulation of the cell cycle during liver cancer, focusing mainly on hepatocellular carcinoma (HCC). We also provide a brief summary of novel therapies connected to cell cycle regulation.
Collapse
|
32
|
Stephens DN, Klein RH, Salmans ML, Gordon W, Ho H, Andersen B. The Ets transcription factor EHF as a regulator of cornea epithelial cell identity. J Biol Chem 2013; 288:34304-24. [PMID: 24142692 DOI: 10.1074/jbc.m113.504399] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The cornea is the clear, outermost portion of the eye composed of three layers: an epithelium that provides a protective barrier while allowing transmission of light into the eye, a collagen-rich stroma, and an endothelium monolayer. How cornea development and aging is controlled is poorly understood. Here we characterize the mouse cornea transcriptome from early embryogenesis through aging and compare it with transcriptomes of other epithelial tissues, identifying cornea-enriched genes, pathways, and transcriptional regulators. Additionally, we profiled cornea epithelium and stroma, defining genes enriched in these layers. Over 10,000 genes are differentially regulated in the mouse cornea across the time course, showing dynamic expression during development and modest expression changes in fewer genes during aging. A striking transition time point for gene expression between postnatal days 14 and 28 corresponds with completion of cornea development at the transcriptional level. Clustering classifies co-expressed, and potentially co-regulated, genes into biologically informative categories, including groups that exhibit epithelial or stromal enriched expression. Based on these findings, and through loss of function studies and ChIP-seq, we show that the Ets transcription factor EHF promotes cornea epithelial fate through complementary gene activating and repressing activities. Furthermore, we identify potential interactions between EHF, KLF4, and KLF5 in promoting cornea epithelial differentiation. These data provide insights into the mechanisms underlying epithelial development and aging, identifying EHF as a regulator of cornea epithelial identity and pointing to interactions between Ets and KLF factors in promoting epithelial fate. Furthermore, this comprehensive gene expression data set for the cornea is a powerful tool for discovery of novel cornea regulators and pathways.
Collapse
|
33
|
Wierstra I. The transcription factor FOXM1 (Forkhead box M1): proliferation-specific expression, transcription factor function, target genes, mouse models, and normal biological roles. Adv Cancer Res 2013; 118:97-398. [PMID: 23768511 DOI: 10.1016/b978-0-12-407173-5.00004-2] [Citation(s) in RCA: 127] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
FOXM1 (Forkhead box M1) is a typical proliferation-associated transcription factor, which stimulates cell proliferation and exhibits a proliferation-specific expression pattern. Accordingly, both the expression and the transcriptional activity of FOXM1 are increased by proliferation signals, but decreased by antiproliferation signals, including the positive and negative regulation by protooncoproteins or tumor suppressors, respectively. FOXM1 stimulates cell cycle progression by promoting the entry into S-phase and M-phase. Moreover, FOXM1 is required for proper execution of mitosis. Accordingly, FOXM1 regulates the expression of genes, whose products control G1/S-transition, S-phase progression, G2/M-transition, and M-phase progression. Additionally, FOXM1 target genes encode proteins with functions in the execution of DNA replication and mitosis. FOXM1 is a transcriptional activator with a forkhead domain as DNA binding domain and with a very strong acidic transactivation domain. However, wild-type FOXM1 is (almost) inactive because the transactivation domain is repressed by three inhibitory domains. Inactive FOXM1 can be converted into a very potent transactivator by activating signals, which release the transactivation domain from its inhibition by the inhibitory domains. FOXM1 is essential for embryonic development and the foxm1 knockout is embryonically lethal. In adults, FOXM1 is important for tissue repair after injury. FOXM1 prevents premature senescence and interferes with contact inhibition. FOXM1 plays a role for maintenance of stem cell pluripotency and for self-renewal capacity of stem cells. The functions of FOXM1 in prevention of polyploidy and aneuploidy and in homologous recombination repair of DNA-double-strand breaks suggest an importance of FOXM1 for the maintenance of genomic stability and chromosomal integrity.
Collapse
|
34
|
Abstract
Cyclin-dependent kinases (Cdks) are serine/threonine kinases and their catalytic activities are modulated by interactions with cyclins and Cdk inhibitors (CKIs). Close cooperation between this trio is necessary for ensuring orderly progression through the cell cycle. In addition to their well-established function in cell cycle control, it is becoming increasingly apparent that mammalian Cdks, cyclins and CKIs play indispensable roles in processes such as transcription, epigenetic regulation, metabolism, stem cell self-renewal, neuronal functions and spermatogenesis. Even more remarkably, they can accomplish some of these tasks individually, without the need for Cdk/cyclin complex formation or kinase activity. In this Review, we discuss the latest revelations about Cdks, cyclins and CKIs with the goal of showcasing their functional diversity beyond cell cycle regulation and their impact on development and disease in mammals.
Collapse
Affiliation(s)
- Shuhui Lim
- Institute of Molecular and Cell Biology IMCB, A*STAR Agency for Science, Technology and Research, Singapore 138673, Republic of Singapore
| | | |
Collapse
|
35
|
Anti-tumor effects of Mfn2 in gastric cancer. Int J Mol Sci 2013; 14:13005-21. [PMID: 23797661 PMCID: PMC3742171 DOI: 10.3390/ijms140713005] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2013] [Revised: 05/19/2013] [Accepted: 06/07/2013] [Indexed: 11/24/2022] Open
Abstract
Mitofusin-2 (Mfn2) is a mitochondrial outer membrane protein involved in mitochondrial fusion. Its mutation can cause Charcot-Marie-Tooth disease. Recent studies of Mfn2 in cancer research have not included gastric cancer. We confirmed that Mfn2 expression was lower in tumor tissue than in normal gastric mucosal tissue and that it was negatively correlated with tumor size, indicating an anti-tumor role for Mfn2. In vitro experiments showed that Mfn2 overexpression suppressed gastric cancer cell proliferation and colony formation, weakened the invasion and migratory ability of cancer cells by downregulating MMP-2 and MMP-9, halted the cell cycle and induced apoptosis. Western blotting indicated the likely involvement of P21 and PI3K/Akt signaling. Therefore, Mfn2 is a potential anti-tumor gene and a potential therapeutic target for treating gastric cancer. The progress of gastric cancer may be delayed by controlling Mfn2 expression.
Collapse
|
36
|
Monnerat S, Almeida Costa CI, Forkert AC, Benz C, Hamilton A, Tetley L, Burchmore R, Novo C, Mottram JC, Hammarton TC. Identification and Functional Characterisation of CRK12:CYC9, a Novel Cyclin-Dependent Kinase (CDK)-Cyclin Complex in Trypanosoma brucei. PLoS One 2013; 8:e67327. [PMID: 23805309 PMCID: PMC3689728 DOI: 10.1371/journal.pone.0067327] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 05/20/2013] [Indexed: 11/19/2022] Open
Abstract
The protozoan parasite, Trypanosoma brucei, is spread by the tsetse fly and causes trypanosomiasis in humans and animals. Both the life cycle and cell cycle of the parasite are complex. Trypanosomes have eleven cdc2-related kinases (CRKs) and ten cyclins, an unusually large number for a single celled organism. To date, relatively little is known about the function of many of the CRKs and cyclins, and only CRK3 has previously been shown to be cyclin-dependent in vivo. Here we report the identification of a previously uncharacterised CRK:cyclin complex between CRK12 and the putative transcriptional cyclin, CYC9. CRK12:CYC9 interact to form an active protein kinase complex in procyclic and bloodstream T. brucei. Both CRK12 and CYC9 are essential for the proliferation of bloodstream trypanosomes in vitro, and we show that CRK12 is also essential for survival of T. brucei in a mouse model, providing genetic validation of CRK12:CYC9 as a novel drug target for trypanosomiasis. Further, functional characterisation of CRK12 and CYC9 using RNA interference reveals roles for these proteins in endocytosis and cytokinesis, respectively.
Collapse
Affiliation(s)
- Séverine Monnerat
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Cristina I. Almeida Costa
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Andrea C. Forkert
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Corinna Benz
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Alana Hamilton
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Laurence Tetley
- School of Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Richard Burchmore
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Carlos Novo
- Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Jeremy C. Mottram
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| | - Tansy C. Hammarton
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
37
|
Harrison Pitner MK, Saavedra HI. Cdk4 and nek2 signal binucleation and centrosome amplification in a her2+ breast cancer model. PLoS One 2013; 8:e65971. [PMID: 23776583 PMCID: PMC3679029 DOI: 10.1371/journal.pone.0065971] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Accepted: 05/02/2013] [Indexed: 12/02/2022] Open
Abstract
Centrosome amplification (CA) is a contributor to carcinogenesis, generating aneuploidy, and chromosome instability. Previous work shows that breast adenocarcinomas have a higher frequency of centrosome defects compared to normal breast tissues. Abnormal centrosome phenotypes are found in pre-malignant lesions, suggesting an early role in breast carcinogenesis. However, the role of CA in breast cancers remains elusive. Identification of pathways and regulatory molecules involved in the generation of CA is essential to understanding its role in breast tumorigenesis. We established a breast cancer model of CA using Her2-positive cells. Our goal was to identify centrosome cycle molecules that are deregulated by aberrant Her2 signaling and the mechanisms driving CA. Our results show some Her2+ breast cancer cell lines harbor both CA and binucleation. Abolishing the expression of Cdk4 abrogated both CA and binucleation in these cells. We also found the source of binucleation in these cells to be defective cytokinesis that is normalized by downregulation of Cdk4. Protein levels of Nek2 diminish upon Cdk4 knockdown and vice versa, suggesting a molecular connection between Cdk4 and Nek2. Knockdown of Nek2 reduces CA and binucleation in this model while its overexpression further enhances centrosome amplification. We conclude that CA is modulated through Cdk4 and Nek2 signaling and that binucleation is a likely source of CA in Her2+ breast cancer cells.
Collapse
Affiliation(s)
- Mary Kathryn Harrison Pitner
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, United States of America
| | - Harold I. Saavedra
- Department of Radiation Oncology, Emory University School of Medicine, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
38
|
Decitabine, a DNA methyltransferases inhibitor, induces cell cycle arrest at G2/M phase through p53-independent pathway in human cancer cells. Biomed Pharmacother 2013; 67:305-11. [PMID: 23582784 DOI: 10.1016/j.biopha.2013.01.004] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2013] [Accepted: 01/24/2013] [Indexed: 12/24/2022] Open
Abstract
Decitabine (5-aza-2'-deoxycytidine), an inhibitor of DNA methyltransferases, has a wide range of anti-metabolic and anti-cancer activities. Decitabine also induces cell cycle arrest at G2/M phase and apoptosis in human cancer cells. However, the cellular and molecular mechanisms of this cell cycle arrest are poorly understood. In the present study, we investigated the roles of the tumor suppressor p53 and the cyclin-dependent kinase (Cdk) inhibitor p21 following decitabine-induced G2/M arrest in human cancer cells. DNA flow cytometric analyses indicated that decitabine induced a G2/M arrest in AGS gastric and A549 lung carcinoma cell lines, which have wild type p53. Western blot analyses using whole cell lysates from AGS cells demonstrated that decitabine treatment did not change the steady-state level of Cdks and Cdk inhibitor p27, but it partially inhibited expression of cyclin A, cyclin B1, and Cdc25C proteins. However, similar results were found using the A549 cell line, where decitabine induced a dramatic up-regulation of both p53 and p21 expression, and the increased levels of p21 were associated with increased binding of p21 with Cdks, cyclin A, and cyclin B1. Knockdown of p53 by small interfering RNA (siRNA) markedly abolished p53 induction by decitabine in AGS cells, yet p53 siRNA had no attenuating effect on p21 induction. In addition, depletion of p21 expression with siRNA, but not p53, significantly attenuated decitabine-induced G2/M arrest. We also observed that decitabine strongly induced G2/M arrest associated with p21 induction in both p53 allele-null (-/-) HCT116 and wild type p53 (+/+) HCT116 cell lines. Therefore, our data indicated that p21 plays a crucial role in decitabine-induced G2/M arrest and operates in a p53-independent manner.
Collapse
|
39
|
Torres-Guzmán R, Chu S, Velasco JA, Lallena MJ. Multiparametric Cell-Based Assay for the Evaluation of Transcription Inhibition by High-Content Imaging. ACTA ACUST UNITED AC 2013; 18:556-66. [DOI: 10.1177/1087057112472539] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Loss of normal cell cycle regulation is a hallmark of human cancer. Cyclin-dependent kinases (CDKs) are key regulators of the cell cycle and have been actively pursued as promising therapeutic targets. Likewise, members of the CDK family are functionally related to transcriptional modulation, a molecular pathway suitable for therapeutic intervention. We used a set of 2500 compounds in the U2OS cell line to evaluate its effect in the cell division process. Interestingly, out of this analysis, we identified a subpopulation of compounds that are able to inhibit RNA polymerase activity, thus interfering with gene transcription processes. After this finding, we developed, validated, and fully automated a multiparameter high-content imaging (HCI) assay to measure phosphorylation of the RNA polymerase II carboxyl terminal domain (pCTD). Simultaneously, we measured both the DNA content and cell proliferation index in the treated cells. The linear regression analysis comparing the IC50 for pCTD and the 4N EC50 for DNA content or IC50 for cell proliferation showed an excellent agreement ( r2 = 0.84 and r2 = 0.94, respectively). Our results confirm that this method allows discriminating between cell cycle and transcription inhibition and confirms HCI as a powerful technology for the identification of compounds with an effective and selective pathway phenotype.
Collapse
Affiliation(s)
- Raquel Torres-Guzmán
- Translational Sciences & Technologies, Lilly Research Laboratories, Eli Lilly & Company, Lilly Research Laboratories, Alcobendas, Madrid, Spain
| | - Shaoyou Chu
- Translational Sciences & Technologies, Lilly Research Laboratories, Eli Lilly & Company, Lilly Research Laboratories, Indianapolis, IN, USA
| | - Juan A. Velasco
- Translational Sciences & Technologies, Lilly Research Laboratories, Eli Lilly & Company, Lilly Research Laboratories, Alcobendas, Madrid, Spain
| | - María José Lallena
- Translational Sciences & Technologies, Lilly Research Laboratories, Eli Lilly & Company, Lilly Research Laboratories, Alcobendas, Madrid, Spain
| |
Collapse
|
40
|
Hernández-Ortega S, Bru S, Ricco N, Ramírez S, Casals N, Jiménez J, Isasa M, Crosas B, Clotet J. Defective in mitotic arrest 1 (Dma1) ubiquitin ligase controls G1 cyclin degradation. J Biol Chem 2012; 288:4704-14. [PMID: 23264631 DOI: 10.1074/jbc.m112.426593] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Progression through the G(1) phase of the cell cycle is controlled by diverse cyclin-dependent kinases (CDKs) that might be associated to numerous cyclin isoforms. Given such complexity, regulation of cyclin degradation should be crucial for coordinating progression through the cell cycle. In Saccharomyces cerevisiae, SCF is the only E3 ligase known to date to be involved in G(1) cyclin degradation. Here, we report the design of a genetic screening that uncovered Dma1 as another E3 ligase that targets G(1) cyclins in yeast. We show that the cyclin Pcl1 is ubiquitinated in vitro and in vivo by Dma1, and accordingly, is stabilized in dma1 mutants. We demonstrate that Pcl1 must be phosphorylated by its own CDK to efficiently interact with Dma1 and undergo degradation. A nonphosphorylatable version of Pcl1 accumulates throughout the cell cycle, demonstrating the physiological relevance of the proposed mechanism. Finally, we present evidence that the levels of Pcl1 and Cln2 are independently controlled in response to nutrient availability. This new previously unknown mechanism for G(1) cyclin degradation that we report here could help elucidate the specific roles of the redundant CDK-cyclin complexes in G(1).
Collapse
Affiliation(s)
- Sara Hernández-Ortega
- Departament de Ciències Bàsiques, Universitat Internacional de Catalunya, 08017 Barcelona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Abstract
Apigenin (4',5,7-trihydroxyflavone, 5,7-dihydroxy-2-(4-hydroxyphenyl)-4H-1-benzopyran-4-one) is a flavonoid found in many fruits, vegetables, and herbs, the most abundant sources being the leafy herb parsley and dried flowers of chamomile. Present in dietary sources as a glycoside, it is cleaved in the gastrointestinal lumen to be absorbed and distributed as apigenin itself. For this reason, the epithelium of the gastrointestinal tract is exposed to higher concentrations of apigenin than tissues at other locations. This would also be true for epithelial cancers of the gastrointestinal tract. We consider the evidence for actions of apigenin that might hinder the ability of gastrointestinal cancers to progress and spread. Apigenin has been shown to inhibit cell growth, sensitize cancer cells to elimination by apoptosis, and hinder the development of blood vessels to serve the growing tumor. It also has actions that alter the relationship of the cancer cells with their microenvironment. Apigenin is able to reduce cancer cell glucose uptake, inhibit remodeling of the extracellular matrix, inhibit cell adhesion molecules that participate in cancer progression, and oppose chemokine signaling pathways that direct the course of metastasis into other locations. As such, apigenin may provide some additional benefit beyond existing drugs in slowing the emergence of metastatic disease.
Collapse
Affiliation(s)
- Émilie C Lefort
- Department of Pathology, Dalhousie University, Halifax, Nova Scotia, Canada
| | | |
Collapse
|
42
|
Segar JL, Volk KA, Lipman MHB, Scholz TD. Thyroid hormone is required for growth adaptation to pressure load in the ovine fetal heart. Exp Physiol 2012; 98:722-33. [PMID: 23104936 DOI: 10.1113/expphysiol.2012.069435] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Thyroid hormone exerts broad effects on the adult heart, but little is known regarding the role of thyroid hormone in the regulation of cardiac growth early in development and in response to pathophysiological conditions. To address this issue, we determined the effects of fetal thyroidectomy on cardiac growth and growth-related gene expression in control and pulmonary-artery-banded fetal sheep. Fetal thyroidectomy (THX) and/or placement of a restrictive pulmonary artery band (PAB) were performed at 126 ± 1 days of gestation (term, 145 days). Four groups of animals [n = 5-6 in each group; (i) control; (ii) fetal THX; (iii) fetal PAB; and (iv) fetal PAB + THX] were monitored for 1 week prior to being killed. Fetal heart rate was significantly lower in the two THX groups compared with the non-THX groups, while mean arterial blood pressure was similar among groups. Combined left and right ventricle free wall + septum weight, expressed per kilogram of fetal weight, was significantly increased in PAB (6.27 ± 0.85 g kg(-1)) compared with control animals (4.72 ± 0.12 g kg(-1)). Thyroidectomy significantly attenuated the increase in cardiac mass associated with PAB (4.94 ± 0.13 g kg(-1)), while THX alone had no detectable effect on heart mass (4.95 ± 0.27 g kg(-1)). The percentage of binucleated cardiomyocytes was significantly decreased in THX and PAB +THX groups (∼16%) compared with the non-THX groups (∼27%). No differences in levels of activated Akt, extracellular signal-regulated kinase or c-Jun N-terminal kinase were detected among the groups. Markers of cellular proliferation but not apoptosis or expression of growth-related genes were lower in the THX and THX+ PAB groups relative to thyroid-intact animals. These findings suggest that in the late-gestation fetal heart, thyroid hormone has important cellular growth functions in both physiological and pathophysiological states. Specifically, thyroid hormone is required for adaptive fetal cardiac growth in response to pressure overload.
Collapse
Affiliation(s)
- Jeffrey L Segar
- Department of Pediatrics, University of Iowa Carver College of Medicine, University of Iowa Children's Hospital, 200 Hawkins Drive, Iowa City, IA 52242, USA.
| | | | | | | |
Collapse
|
43
|
Lim S, Kaldis P. Loss of Cdk2 and Cdk4 induces a switch from proliferation to differentiation in neural stem cells. Stem Cells 2012; 30:1509-20. [PMID: 22532528 DOI: 10.1002/stem.1114] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
During neurogenesis, cell cycle regulators play a pivotal role in ensuring proper proliferation, cell cycle exit, and differentiation of neural precursors. However, the precise role of cyclin-dependent kinases (Cdks) in these processes is not well understood. We generated Cdk2 and Cdk4 double knockout (DKO) mice and found a striking ablation of the intermediate zone and cortical plate in mouse embryonic brain. When neural stem cells (NSCs) were isolated and analyzed, DKO NSCs proliferated comparable to wild type as Cdk1 now binds to cyclin D1 and E1 and assumes the role vacated by the loss of Cdk2 and Cdk4 in phosphorylating Rb. Although compensation was sufficient for the maintenance of self-renewal and multilineage potential, DKO NSCs displayed an altered cell cycle profile and were more prone to neuronal differentiation. This was manifested in vivo as a marked reduction in S-phase length and an increased tendency for neurogenic divisions that prevented proper expansion of the basal progenitor pool. Our data thus demonstrate the induction of neurogenic divisions in the absence of critical mediators of G1/S transition-Cdk2 and Cdk4, and highlight their evolutionary importance in the determination of cortical thickness.
Collapse
Affiliation(s)
- Shuhui Lim
- Institute of Molecular and Cell Biology (IMCB), A*STAR (Agency for Science, Technology and Research), Singapore, Singapore
| | | |
Collapse
|
44
|
Inhibition of human cytomegalovirus immediate-early gene expression by cyclin A2-dependent kinase activity. J Virol 2012; 86:9369-83. [PMID: 22718829 DOI: 10.1128/jvi.07181-11] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Human cytomegalovirus (HCMV) starts its lytic replication cycle only in the G(0)/G(1) phase of the cell division cycle. S/G(2) cells can be infected but block the onset of immediate-early (IE) gene expression. This block can be overcome by inhibition of cyclin-dependent kinases (CDKs), suggesting that cyclin A2, the only cyclin with an S/G(2)-specific activity profile, may act as a negative regulator of viral gene expression. To directly test this hypothesis, we generated derivatives of an HCMV-permissive glioblastoma cell line that express cyclin A2 in a constitutive, cell cycle-independent manner. We demonstrate that even moderate cyclin A2 overexpression in G(1) was sufficient to severely compromise the HCMV replicative cycle after high-multiplicity infection. This negative effect was composed of a strong but transient inhibition of IE gene transcription and a more sustained alteration of IE mRNA processing, resulting in reduced levels of UL37 and IE2, an essential transactivator of viral early gene expression. Consistently, cyclin A2-overexpressing cells showed a strong delay of viral early and late gene expression, as well as virus reproduction. All effects were dependent on CDK activity, as a cyclin A2 mutant deficient in CDK binding was unable to interfere with the HCMV infectious cycle. Interestingly, murine CMV, whose IE gene expression is known to be cell cycle independent, is not affected by cyclin A2. Instead, it upregulates cyclin A2-associated kinase activity upon infection. Understanding the mechanisms behind the HCMV-specific action of cyclin A2-CDK might reveal new targets for antiviral strategies.
Collapse
|
45
|
Recent progress in studies of arterivirus- and coronavirus-host interactions. Viruses 2012; 4:980-1010. [PMID: 22816036 PMCID: PMC3397358 DOI: 10.3390/v4060980] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 05/30/2012] [Accepted: 06/14/2012] [Indexed: 12/15/2022] Open
Abstract
Animal coronaviruses, such as infectious bronchitis virus (IBV), and arteriviruses, such as porcine reproductive and respiratory syndrome virus (PRRSV), are able to manifest highly contagious infections in their specific native hosts, thereby arising in critical economic damage to animal industries. This review discusses recent progress in studies of virus-host interactions during animal and human coronavirus and arterivirus infections, with emphasis on IBV-host cell interactions. These interactions may be directly involved in viral replication or lead to the alteration of certain signaling pathways, such as cell stress response and innate immunity, to facilitate viral replication and pathogenesis.
Collapse
|
46
|
Abstract
The recent Company of Biologists workshop 'Growth, Division and Differentiation: Understanding Developmental Control', which was held in September 2011 at Wiston House, West Sussex, UK, brought together researchers aiming to understand cell proliferation and differentiation in various metazoans, ranging from flies to mice. Here, we review the common themes that emerged from the meeting, highlighting novel insights into the interplay between regulators of cell proliferation and differentiation during development.
Collapse
Affiliation(s)
- Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore.
| | | |
Collapse
|
47
|
Kubiak JZ, El Dika M. Canonical and Alternative Pathways in Cyclin-Dependent Kinase 1/Cyclin B Inactivation upon M-Phase Exit in Xenopus laevis Cell-Free Extracts. Enzyme Res 2011; 2011:523420. [PMID: 21755042 PMCID: PMC3132491 DOI: 10.4061/2011/523420] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 04/01/2011] [Accepted: 04/18/2011] [Indexed: 11/20/2022] Open
Abstract
Cyclin-Dependent Kinase 1 (CDK1) is the major M-phase kinase known also as the M-phase Promoting Factor or MPF. Studies performed during the last decade have shown many details of how CDK1 is regulated and also how it regulates the cell cycle progression. Xenopus laevis cell-free extracts were widely used to elucidate the details and to obtain a global view of the role of CDK1 in M-phase control. CDK1 inactivation upon M-phase exit is a primordial process leading to the M-phase/interphase transition during the cell cycle. Here we discuss two closely related aspects of CDK1 regulation in Xenopus laevis cell-free extracts: firstly, how CDK1 becomes inactivated and secondly, how other actors, like kinases and phosphatases network and/or specific inhibitors, cooperate with CDK1 inactivation to assure timely exit from the M-phase.
Collapse
Affiliation(s)
- Jacek Z Kubiak
- Cell Cycle Group, Institute of Genetics & Development, University of Rennes 1, CNRS-UMR 6061, Faculty of Medicine, 2 Avenue Prof. Léon Bernard, CS 34317, 35043 Rennes Cedex, France
| | | |
Collapse
|
48
|
Momčilović O, Navara C, Schatten G. Cell cycle adaptations and maintenance of genomic integrity in embryonic stem cells and induced pluripotent stem cells. Results Probl Cell Differ 2011; 53:415-458. [PMID: 21630155 DOI: 10.1007/978-3-642-19065-0_18] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Pluripotent stem cells have the capability to undergo unlimited self-renewal and differentiation into all somatic cell types. They have acquired specific adjustments in the cell cycle structure that allow them to rapidly proliferate, including cell cycle independent expression of cell cycle regulators and lax G(1) to S phase transition. However, due to the developmental role of embryonic stem cells (ES) it is essential to maintain genomic integrity and prevent acquisition of mutations that would be transmitted to multiple cell lineages. Several modifications in DNA damage response of ES cells accommodate dynamic cycling and preservation of genetic information. The absence of a G(1)/S cell cycle arrest promotes apoptotic response of damaged cells before DNA changes can be fixed in the form of mutation during the S phase, while G(2)/M cell cycle arrest allows repair of damaged DNA following replication. Furthermore, ES cells express higher level of DNA repair proteins, and exhibit enhanced repair of multiple types of DNA damage. Similarly to ES cells, induced pluripotent stem (iPS) cells are poised to proliferate and exhibit lack of G(1)/S cell cycle arrest, extreme sensitivity to DNA damage, and high level of expression of DNA repair genes. The fundamental mechanisms by which the cell cycle regulates genomic integrity in ES cells and iPS cells are similar, though not identical.
Collapse
Affiliation(s)
- Olga Momčilović
- Department of Human Genetics, University of Pittsburgh, Pittsburgh, PA, USA.
| | | | | |
Collapse
|
49
|
Haccard O, Jessus C. Greatwall kinase, ARPP-19 and protein phosphatase 2A: shifting the mitosis paradigm. Results Probl Cell Differ 2011; 53:219-234. [PMID: 21630148 DOI: 10.1007/978-3-642-19065-0_11] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Control of entry into mitosis has long been seen in terms of an explosive activation of cyclin-dependent kinase 1, the mitotic driver ensuring the phosphorylation of hundreds of proteins required for cell division. However, if these phosphorylations are maintained during M-phase, they must be removed when cells exit mitosis. It has been surmised that an "antimitotic" phosphatase must be inhibited to allow mitosis entry and activated for returning to interphase. This chapter discusses a series of recent works conducted on Xenopus egg extracts that provide the answers regarding the identity and the regulation of such a phosphatase. PP2A-B55δ is the major phosphatase controlling exit from mitosis; it is negatively regulated by the kinase Greatwall that phosphorylates the small protein ARPP-19 and converts it into a potent PP2A inhibitor. These findings provide a new element of paramount importance in the control of mitosis.
Collapse
Affiliation(s)
- Olivier Haccard
- UMR-CNRS 7622 Biologie du Développement, Université Paris 6, 9 quai Saint-Bernard, 75005 Paris, France
| | | |
Collapse
|