1
|
Kwiatkowski N, Liang T, Sha Z, Collier PN, Yang A, Sathappa M, Paul A, Su L, Zheng X, Aversa R, Li K, Mehovic R, Kolodzy C, Breitkopf SB, Chen D, Howarth CL, Yuan K, Jo H, Growney JD, Weiss M, Williams J. CDK2 heterobifunctional degraders co-degrade CDK2 and cyclin E resulting in efficacy in CCNE1-amplified and overexpressed cancers. Cell Chem Biol 2025; 32:556-569.e24. [PMID: 40250405 DOI: 10.1016/j.chembiol.2025.03.006] [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: 09/11/2024] [Revised: 01/27/2025] [Accepted: 03/18/2025] [Indexed: 04/20/2025]
Abstract
CCNE1 amplification drives aberrant CDK2-cyclin E1 activity in cancer. Despite activity of CDK2 inhibitors, their therapeutic margins are limited by poor CDK selectivity. We developed a degrader with high selectivity for CDK2 over CDK1 that also unexpectedly led to cyclin E1 degradation and potent and complete suppression of RB phosphorylation at concentrations with low CDK2 occupancy and negligible CDK1 degradation. Co-depletion of CDK2 and cyclin E1 also resensitized palbociclib-adapted breast cancer cells to cell cycle blockade. Overall, the improved potency and selectivity of the degrader for CDK2 over small-molecule inhibitors drives antiproliferative activity with greater specificity for CCNE1amp cancer cells and RB dependency. Using an orally administered degrader, we demonstrate deep and sustained RB pathway suppression, which is needed to induce stasis in CCNE1amp tumors. These results highlight the potential of this modality to target CDK2 potently and selectivity in this biomarker-defined patient population with high unmet need.
Collapse
Affiliation(s)
- Nicholas Kwiatkowski
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Tong Liang
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Zhe Sha
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Philip N Collier
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Annan Yang
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Murugappan Sathappa
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Atanu Paul
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Lijing Su
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Xiaozhang Zheng
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Robert Aversa
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Kunhua Li
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Revonda Mehovic
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Christina Kolodzy
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Susanne B Breitkopf
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Dapeng Chen
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Charles L Howarth
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Karen Yuan
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Hakryul Jo
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Joseph D Growney
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Matthew Weiss
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA
| | - Juliet Williams
- Kymera Therapeutics, Inc., Biological Sciences, 500 North Beacon St. 4th Floor, Watertown, MA 02472, USA.
| |
Collapse
|
2
|
Xu R, Hao Y, Liu Y, Ji B, Tian W, Zhang W. Functional mechanisms and potential therapeutic strategies for lactylation in liver diseases. Life Sci 2025; 363:123395. [PMID: 39809380 DOI: 10.1016/j.lfs.2025.123395] [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: 11/07/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Lactylation, a novel form of lactate-mediated protein post-translational modification (PTM), has been identified as a crucial regulator of gene expression and protein function through the modification of both histone and non-histone proteins. Liver disease is frequently characterized by a reprogramming of glucose metabolism and subsequent lactate accumulation. Recent research has implicated lactylation in a diverse array of hepatic pathologies, including liver injury, non-alcoholic fatty liver disease, liver fibrosis, and hepatocellular carcinoma. Consequently, lactylation has emerged as a pivotal regulatory mechanism in liver disease pathogenesis. This review aims to elucidate the intricate regulatory and functional mechanisms underlying lactylation, synthesize recent advancements in its role in various liver diseases, and highlight its potential as a therapeutic target for future interventions in hepatic disorders.
Collapse
Affiliation(s)
- Rong Xu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Yitong Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Yahui Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Bai Ji
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Weibo Tian
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China
| | - Wei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun 130021, Jilin Province, China.
| |
Collapse
|
3
|
Shen X, Yan S, Zeng T, Xia F, Jiang D, Wan G, Cao D, Wu R. TarIKGC: A Target Identification Tool Using Semantics-Enhanced Knowledge Graph Completion with Application to CDK2 Inhibitor Discovery. J Med Chem 2025; 68:1793-1809. [PMID: 39745279 DOI: 10.1021/acs.jmedchem.4c02543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
Target identification is a critical stage in the drug discovery pipeline. Various computational methodologies have been dedicated to enhancing the classification performance of compound-target interactions, yet significant room remains for improving the recommendation performance. To address this challenge, we developed TarIKGC, a tool for target prioritization that leverages semantics enhanced knowledge graph (KG) completion. This method harnesses knowledge representation learning within a heterogeneous compound-target-disease network. Specifically, TarIKGC combines an attention-based aggregation graph neural network with a multimodal feature extractor network to simultaneously learn internal semantic features from biomedical entities and topological features from the KG. Furthermore, a KG embedding model is employed to identify missing relationships among compounds and targets. In silico evaluations highlighted the superior performance of TarIKGC in drug repositioning tasks. In addition, TarIKGC successfully identified two potential cyclin-dependent kinase 2 (CDK2) inhibitors with novel scaffolds through reverse target fishing. Both compounds exhibited antiproliferative activities across multiple therapeutic indications targeting CDK2.
Collapse
Affiliation(s)
- Xiaojuan Shen
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Shijia Yan
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Tao Zeng
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Fei Xia
- School of Chemistry and Molecular Engineering, East China Normal University, Shanghai 200062, China
| | - Dejun Jiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Guohui Wan
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| | - Dongsheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan 410013, China
| | - Ruibo Wu
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China
| |
Collapse
|
4
|
Kim D, Chung H, Liu W, Jeong K, Ozmen TY, Ozmen F, Rames MJ, Kim S, Guo X, Jameson N, de Jong PR, Yea S, Harford L, Li J, Mathews CA, Doroshow DB, Charles VJ, Kim D, Fischer K, Samatar AA, Jubb A, Bunker KD, Blackwell K, Simpkins F, Meric-Bernstam F, Mills GB, Harismendy O, Ma J, Lackner MR. Cyclin E1/CDK2 activation defines a key vulnerability to WEE1 kinase inhibition in gynecological cancers. NPJ Precis Oncol 2025; 9:3. [PMID: 39755818 DOI: 10.1038/s41698-024-00787-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 12/13/2024] [Indexed: 01/06/2025] Open
Abstract
Upregulation of Cyclin E1 and subsequent activation of CDK2 accelerates cell cycle progression from G1 to S phase and is a common oncogenic driver in gynecological malignancies. WEE1 kinase counteracts the effects of Cyclin E1/CDK2 activation by regulating multiple cell cycle checkpoints. Here we characterized the relationship between Cyclin E1/CDK2 activation and sensitivity to the selective WEE1 inhibitor azenosertib. We found that ovarian cancer cell lines with high levels of endogenous Cyclin E1 expression or forced overexpression were exquisitely sensitive to azenosertib and these results extended to in vivo models of ovarian and uterine serous carcinoma. Models with high Cyclin E1 expression showed higher baseline levels of replication stress and enhanced cellular responses to azenosertib treatment. We found azenosertib synergized with different classes of chemotherapy and described distinct underlying mechanisms. Finally, we provided early evidence from an ongoing phase I study demonstrating the clinical activity of monotherapy azenosertib in patients with Cyclin E1/CDK2-activated ovarian and uterine serous carcinomas.
Collapse
Affiliation(s)
- Daehwan Kim
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA
| | | | - Wen Liu
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA
| | - Kangjin Jeong
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Tugba Y Ozmen
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Furkan Ozmen
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Matthew J Rames
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
- Cancer Early Detection Advanced Research Center (CEDAR), Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | - Sangyub Kim
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA
| | - Xiao Guo
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA
| | | | | | - Steven Yea
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA
| | | | - Jiali Li
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA
| | - Cara A Mathews
- Program in Women's Oncology, Women & Infants Hospital, Legorreta Cancer Center of Alpert Medical School at Brown University, Providence, RI, USA
| | - Deborah B Doroshow
- Early Phase Trials Unit, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Vincent J Charles
- The University of Chicago Medicine & Biological Sciences, Chicago, IL, USA
| | - Doris Kim
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA
| | | | | | - Adrian Jubb
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA
| | | | | | - Fiona Simpkins
- Ovarian Cancer Research Center, Division of Gynecologic Oncology, Department of Obstetrics & Gynecology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Funda Meric-Bernstam
- Department of Investigational Cancer Therapeutics, Division of Cancer Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon B Mills
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, USA
| | | | - Jianhui Ma
- Zentalis Pharmaceuticals, Inc., San Diego, CA, USA.
| | | |
Collapse
|
5
|
Zhang N, Zhang H, Li S, Wu W, Luo P, Liu Z, Chen Y, Xia Z, Huang C, Cheng Q. Uncovering the predictive and immunomodulatory potential of transient receptor potential melastatin family-related CCNE1 in pan-cancer. Mol Cancer 2024; 23:258. [PMID: 39551726 PMCID: PMC11572178 DOI: 10.1186/s12943-024-02169-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Accepted: 11/05/2024] [Indexed: 11/19/2024] Open
Abstract
Millions of new cases of cancer are diagnosed worldwide each year, making it a serious public health concern. Developments in customized therapy and early detection have significantly enhanced treatment for and results from cancer. Therefore, it is important to investigate new molecular biomarkers. In this study, we created an efficient transient receptor potential melastatin (TRPM) family members-related TRPM-Score for 17 solid tumors. CCNE1, produced from TRPM-Score, was found to be an exceptional biomarker through several sophisticated machine learning and deep learning computational techniques. TRPM-Score and CCNE1 immunotherapeutic prediction, immunological characteristics, and predictive value were thoroughly assessed. In most cancer types, CCNE1 was a substantially dangerous marker. Additional in vitro tests validated CCNE1's immunomodulatory properties, demonstrating that silencing impeded macrophage movement and decreased PD-L1 expression. Additionally, CCNE1 may accurately predict responses to cancer immunotherapy. These findings indicate that the TRPM family-particularly CCNE1, which is associated with TRPM-is a significant player in the pan-cancer domain and can be utilized as a therapeutic target and prognostic biomarkers, especially in immuno-oncology. The thorough characterization of the TRPM family and the discovery of CCNE1 as a crucial downstream effector mark important developments in our comprehension of pan-cancer biology.
Collapse
Affiliation(s)
- Nan Zhang
- College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Zhang
- Department of Neurosurgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Shuyu Li
- Department of Thyroid and Breast Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wantao Wu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zhiwei Xia
- Department of Neurology, Hunan Aerospace Hospital, Hunan Normal University, Changsha, China.
| | - Chenshen Huang
- Department of Gastrointestinal Surgery, Fujian Provincial Hospital, Fuzhou University Affiliated Provincial Hospital, Fuzhou, China.
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.
- Xiangya Hospital, National Clinical Research Center for Geriatric Disorders, Central South University, Changsha, China.
| |
Collapse
|
6
|
Chen Y, Jia M, Ge L, Li Z, He H, Zhou X, Li F. A Negative Feedback Loop Compromises NMD-Mediated Virus Restriction by the Autophagy Pathway in Plants. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400978. [PMID: 39189522 PMCID: PMC11348178 DOI: 10.1002/advs.202400978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Revised: 05/21/2024] [Indexed: 08/28/2024]
Abstract
Nonsense-mediated decay (NMD) and autophagy play pivotal roles in restricting virus infection in plants. However, the interconnection between these two pathways in viral infections has not been explored. Here, it is shown that overexpression of NbSMG7 and NbUPF3 attenuates cucumber green mottle mosaic virus (CGMMV) infection by recognizing the viral internal termination codon and vice versa. NbSMG7 is subjected to autophagic degradation, which is executed by its interaction with one of the autophagy-related proteins, NbATG8i. Mutation of the ATG8 interacting motif (AIM) in NbSMG7 (SMG7mAIM1) abolishes the interaction and comprises its autophagic degradation. Silencing of NbSMG7 and NbATG8i, or NbUPF3 and NbATG8i, compared to silencing each gene individually, leads to more virus accumulations, but overexpression of NbSMG7 and NbATG8i fails to achieve more potent virus inhibition. When CGMMV is co-inoculated with NbSMG7mAIM1 or with NbUPF3, compared to co-inoculating with NbSMG7 in NbATG8i transgene plants, the inoculated plants exhibit milder viral phenotypes. These findings reveal that NMD-mediated virus inhibition is impaired by the autophagic degradation of SMG7 in a negative feedback loop, and a novel regulatory interplay between NMD and autophagy is uncovered, providing insights that are valuable in optimizing strategies to harness NMD and autophagy for combating viral infections.
Collapse
Affiliation(s)
- Yalin Chen
- State Key Laboratory for Biology of Plant Diseases and Insect PestsInstitute of Plant ProtectionChinese Academy of Agricultural SciencesBeijing100193China
| | - Mingxuan Jia
- State Key Laboratory for Biology of Plant Diseases and Insect PestsInstitute of Plant ProtectionChinese Academy of Agricultural SciencesBeijing100193China
| | - Linhao Ge
- State Key Laboratory for Biology of Plant Diseases and Insect PestsInstitute of Plant ProtectionChinese Academy of Agricultural SciencesBeijing100193China
| | - Zhaolei Li
- State Key Laboratory for Biology of Plant Diseases and Insect PestsInstitute of Plant ProtectionChinese Academy of Agricultural SciencesBeijing100193China
| | - Hao He
- State Key Laboratory for Biology of Plant Diseases and Insect PestsInstitute of Plant ProtectionChinese Academy of Agricultural SciencesBeijing100193China
| | - Xueping Zhou
- State Key Laboratory for Biology of Plant Diseases and Insect PestsInstitute of Plant ProtectionChinese Academy of Agricultural SciencesBeijing100193China
- State Key Laboratory of Rice BiologyInstitute of BiotechnologyZhejiang UniversityHangzhouZhejiang310058China
| | - Fangfang Li
- State Key Laboratory for Biology of Plant Diseases and Insect PestsInstitute of Plant ProtectionChinese Academy of Agricultural SciencesBeijing100193China
| |
Collapse
|
7
|
Lao J, Sun H, Wang A, Wu M, Liu D, Zhang Y, Chen C, Xia Q, Ma S. Effect of eIF6 on the development of silk glands and silk protein synthesis of the silkworm, Bombyx mori. Int J Biol Macromol 2024; 256:128316. [PMID: 38000606 DOI: 10.1016/j.ijbiomac.2023.128316] [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: 10/19/2023] [Revised: 11/18/2023] [Accepted: 11/19/2023] [Indexed: 11/26/2023]
Abstract
The silkworm is a lepidopteran domesticated from the wild silkworm, mostly valued for its efficient synthesis of silk protein. This species' ability to spin silk has supported the 5500-year-old silk industry and the globally known "Silk Road", making the transformation of mulberry leaves into silk of great concern. Therefore, research on the silk-related genes of silkworms and their regulatory mechanisms has attracted increasing attention. Previous studies have revealed that domestic silk gland cells are endoreduplication cells, and their high-copy genome and special chromatin conformation provide conditions for the high expression of silk proteins. In this study, we systematically investigate the expression pattern of eukaryotic initiation factors (eIFs) and identified the eIF6 as a eukaryotic translation initiation factor involved in the synthesis of silk proteins. We generated an eIF6 gene deletion mutant strain of silkworm using the CRISPR/Cas9 system and investigated the function of eIF6 in silk gland development and silk protein synthesis. The results showed that deletion of eIF6 inhibited the individual development of silkworm larvae, inhibited the development of silk glands, and significantly reduced the cocoon layer ratio. Therefore, we elucidated the function of eIF6 in the development of silk glands and the synthesis of silk proteins, which is important for further elucidation of the developmental process of silk glands and the mechanism underlying the ultra-high expression of silk proteins.
Collapse
Affiliation(s)
- Junjie Lao
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Hao Sun
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Aoming Wang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Mingke Wu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Dan Liu
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Yan Zhang
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Chaojie Chen
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China
| | - Qingyou Xia
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China; State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400716, China.
| | - Sanyuan Ma
- Integrative Science Center of Germplasm Creation in Western China (CHONGQING) Science City, Biological Science Research Center, Southwest University, Chongqing 400716, China; State Key Laboratory of Silkworm Genome Biology, Biological Science Research Center, Southwest University, Chongqing 400716, China.
| |
Collapse
|
8
|
Hild V, Mellert K, Möller P, Barth TFE. Giant Cells of Various Lesions Are Characterised by Different Expression Patterns of HLA-Molecules and Molecules Involved in the Cell Cycle, Bone Metabolism, and Lineage Affiliation: An Immunohistochemical Study with a Review of the Literature. Cancers (Basel) 2023; 15:3702. [PMID: 37509363 PMCID: PMC10377796 DOI: 10.3390/cancers15143702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/30/2023] [Accepted: 07/06/2023] [Indexed: 07/30/2023] Open
Abstract
Giant cells (GCs) are thought to originate from the fusion of monocytic lineage cells and arise amid multiple backgrounds. To compare GCs of different origins, we immunohistochemically characterised the GCs of reactive and neoplastic lesions (n = 47). We studied the expression of 15 molecules including HLA class II molecules those relevant to the cell cycle, bone metabolism and lineage affiliation. HLA-DR was detectable in the GCs of sarcoidosis, sarcoid-like lesions, tuberculosis, and foreign body granuloma. Cyclin D1 was expressed by the GCs of neoplastic lesions as well as the GCs of bony callus, fibroid epulis, and brown tumours. While cyclin E was detected in the GCs of all lesions, p16 and p21 showed a heterogeneous expression pattern. RANK was expressed by the GCs of all lesions except sarcoid-like lesions and xanthogranuloma. All GCs were RANK-L-negative, and the GCs of all lesions were osteoprotegerin-positive. Osteonectin was limited to the GCs of chondroblastoma. Osteopontin and TRAP were detected in the GCs of all lesions except xanthogranuloma. RUNX2 was heterogeneously expressed in the reactive and neoplastic cohort. The GCs of all lesions except foreign body granuloma expressed CD68, and all GCs were CD163- and langerin-negative. This profiling points to a functional diversity of GCs despite their similar morphology.
Collapse
Affiliation(s)
- Vivien Hild
- Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| | - Kevin Mellert
- Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| | - Peter Möller
- Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| | - Thomas F E Barth
- Institute of Pathology, University Hospital Ulm, 89081 Ulm, Germany
| |
Collapse
|
9
|
Jin J, Bai L, Wang D, Ding W, Cao Z, Yan P, Li Y, Xi L, Wang Y, Zheng X, Wei H, Ding C, Wang Y. SIRT3-dependent delactylation of cyclin E2 prevents hepatocellular carcinoma growth. EMBO Rep 2023; 24:e56052. [PMID: 36896611 PMCID: PMC10157311 DOI: 10.15252/embr.202256052] [Citation(s) in RCA: 89] [Impact Index Per Article: 44.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 01/30/2023] [Accepted: 02/20/2023] [Indexed: 03/11/2023] Open
Abstract
Lysine lactylation (Kla) is a recently discovered histone mark derived from metabolic lactate. The NAD+ -dependent deacetylase SIRT3, which can also catalyze removal of the lactyl moiety from lysine, is expressed at low levels in hepatocellular carcinoma (HCC) and has been suggested to be an HCC tumor suppressor. Here we report that SIRT3 can delactylate non-histone proteins and suppress HCC development. Using SILAC-based quantitative proteomics, we identify cyclin E2 (CCNE2) as one of the lactylated substrates of SIRT3 in HCC cells. Furthermore, our crystallographic study elucidates the mechanism of CCNE2 K348la delactylation by SIRT3. Our results further suggest that lactylated CCNE2 promotes HCC cell growth, while SIRT3 activation by Honokiol induces HCC cell apoptosis and prevents HCC outgrowth in vivo by regulating Kla levels of CCNE2. Together, our results establish a physiological function of SIRT3 as a delactylase that is important for suppressing HCC, and our structural data could be useful for the future design of activators.
Collapse
Affiliation(s)
- Jing Jin
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Lin Bai
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Human Phenome Institute, School of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Dongyao Wang
- Division of Life Sciences and Medicine, Department of Hematology, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
| | - Wei Ding
- Beijing National Laboratory for Condensed Matter Physics, Institute of PhysicsChinese Academy of SciencesBeijingChina
| | - Zhuoxian Cao
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Peidong Yan
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Yunjia Li
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Lulu Xi
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Yuxin Wang
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
| | - Xiaohu Zheng
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Haiming Wei
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| | - Chen Ding
- State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Human Phenome Institute, School of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
| | - Yi Wang
- Division of Life Sciences and Medicine, Department of Hepatobiliary Surgery, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHeifeiChina
- Institute of Immunology and the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medicine and Medical CenterUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
10
|
Kang E, Weir A, Meagher NS, Farrington K, Nelson GS, Ghatage P, Lee C, Riggan MJ, Bolithon A, Popovic G, Leung B, Tang K, Lambie N, Millstein J, Alsop J, Anglesio MS, Ataseven B, Barlow E, Beckmann MW, Berger J, Bisinotto C, Bösmüller H, Boros J, Brand AH, Brooks‐Wilson A, Brucker SY, Carney ME, Casablanca Y, Cazorla‐Jiménez A, Cohen PA, Conrads TP, Cook LS, Coulson P, Courtney‐Brooks M, Cramer DW, Crowe P, Cunningham JM, Cybulski C, Darcy KM, El‐Bahrawy MA, Elishaev E, Erber R, Farrell R, Fereday S, Fischer A, García MJ, Gayther SA, Gentry‐Maharaj A, Gilks CB, Grube M, Harnett PR, Harrington SP, Harter P, Hartmann A, Hecht JL, Heikaus S, Hein A, Heitz F, Hendley J, Hernandez BY, Polo SH, Heublein S, Hirasawa A, Høgdall E, Høgdall CK, Horlings HM, Huntsman DG, Huzarski T, Jewell A, Jimenez‐Linan M, Jones ME, Kaufmann SH, Kennedy CJ, Khabele D, Kommoss FKF, Kruitwagen RFPM, Lambrechts D, Le ND, Lener M, Lester J, Leung Y, Linder A, Loverix L, Lubiński J, Madan R, Maxwell GL, Modugno F, Neuhausen SL, Olawaiye A, Olbrecht S, Orsulic S, Palacios J, Pearce CL, Pike MC, Quinn CM, Mohan GR, Rodríguez‐Antona C, Ruebner M, Ryan A, Salfinger SG, et alKang E, Weir A, Meagher NS, Farrington K, Nelson GS, Ghatage P, Lee C, Riggan MJ, Bolithon A, Popovic G, Leung B, Tang K, Lambie N, Millstein J, Alsop J, Anglesio MS, Ataseven B, Barlow E, Beckmann MW, Berger J, Bisinotto C, Bösmüller H, Boros J, Brand AH, Brooks‐Wilson A, Brucker SY, Carney ME, Casablanca Y, Cazorla‐Jiménez A, Cohen PA, Conrads TP, Cook LS, Coulson P, Courtney‐Brooks M, Cramer DW, Crowe P, Cunningham JM, Cybulski C, Darcy KM, El‐Bahrawy MA, Elishaev E, Erber R, Farrell R, Fereday S, Fischer A, García MJ, Gayther SA, Gentry‐Maharaj A, Gilks CB, Grube M, Harnett PR, Harrington SP, Harter P, Hartmann A, Hecht JL, Heikaus S, Hein A, Heitz F, Hendley J, Hernandez BY, Polo SH, Heublein S, Hirasawa A, Høgdall E, Høgdall CK, Horlings HM, Huntsman DG, Huzarski T, Jewell A, Jimenez‐Linan M, Jones ME, Kaufmann SH, Kennedy CJ, Khabele D, Kommoss FKF, Kruitwagen RFPM, Lambrechts D, Le ND, Lener M, Lester J, Leung Y, Linder A, Loverix L, Lubiński J, Madan R, Maxwell GL, Modugno F, Neuhausen SL, Olawaiye A, Olbrecht S, Orsulic S, Palacios J, Pearce CL, Pike MC, Quinn CM, Mohan GR, Rodríguez‐Antona C, Ruebner M, Ryan A, Salfinger SG, Sasamoto N, Schildkraut JM, Schoemaker MJ, Shah M, Sharma R, Shvetsov YB, Singh N, Sonke GS, Steele L, Stewart CJR, Sundfeldt K, Swerdlow AJ, Talhouk A, Tan A, Taylor SE, Terry KL, Tołoczko A, Traficante N, Van de Vijver KK, van der Aa MA, Van Gorp T, Van Nieuwenhuysen E, van‐Wagensveld L, Vergote I, Vierkant RA, Wang C, Wilkens LR, Winham SJ, Wu AH, Benitez J, Berchuck A, Candido dos Reis FJ, DeFazio A, Fasching PA, Goode EL, Goodman MT, Gronwald J, Karlan BY, Kommoss S, Menon U, Sinn H, Staebler A, Brenton JD, Bowtell DD, Pharoah PDP, Ramus SJ, Köbel M. CCNE1 and survival of patients with tubo-ovarian high-grade serous carcinoma: An Ovarian Tumor Tissue Analysis consortium study. Cancer 2023; 129:697-713. [PMID: 36572991 PMCID: PMC10107112 DOI: 10.1002/cncr.34582] [Show More Authors] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 09/14/2022] [Accepted: 09/30/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Cyclin E1 (CCNE1) is a potential predictive marker and therapeutic target in tubo-ovarian high-grade serous carcinoma (HGSC). Smaller studies have revealed unfavorable associations for CCNE1 amplification and CCNE1 overexpression with survival, but to date no large-scale, histotype-specific validation has been performed. The hypothesis was that high-level amplification of CCNE1 and CCNE1 overexpression, as well as a combination of the two, are linked to shorter overall survival in HGSC. METHODS Within the Ovarian Tumor Tissue Analysis consortium, amplification status and protein level in 3029 HGSC cases and mRNA expression in 2419 samples were investigated. RESULTS High-level amplification (>8 copies by chromogenic in situ hybridization) was found in 8.6% of HGSC and overexpression (>60% with at least 5% demonstrating strong intensity by immunohistochemistry) was found in 22.4%. CCNE1 high-level amplification and overexpression both were linked to shorter overall survival in multivariate survival analysis adjusted for age and stage, with hazard stratification by study (hazard ratio [HR], 1.26; 95% CI, 1.08-1.47, p = .034, and HR, 1.18; 95% CI, 1.05-1.32, p = .015, respectively). This was also true for cases with combined high-level amplification/overexpression (HR, 1.26; 95% CI, 1.09-1.47, p = .033). CCNE1 mRNA expression was not associated with overall survival (HR, 1.00 per 1-SD increase; 95% CI, 0.94-1.06; p = .58). CCNE1 high-level amplification is mutually exclusive with the presence of germline BRCA1/2 pathogenic variants and shows an inverse association to RB1 loss. CONCLUSION This study provides large-scale validation that CCNE1 high-level amplification is associated with shorter survival, supporting its utility as a prognostic biomarker in HGSC.
Collapse
Affiliation(s)
- Eun‐Young Kang
- Department of Pathology and Laboratory MedicineUniversity of CalgaryFoothills Medical CenterCalgaryAlbertaCanada
| | - Ashley Weir
- School of Clinical MedicineUNSW Medicine and HealthUniversity of NSW SydneySydneyNew South WalesAustralia
- Adult Cancer ProgramLowy Cancer Research CentreUniversity of NSW SydneySydneyNew South WalesAustralia
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVictoriaAustralia
| | - Nicola S. Meagher
- School of Clinical MedicineUNSW Medicine and HealthUniversity of NSW SydneySydneyNew South WalesAustralia
- The Daffodil CentreThe University of SydneyA Joint Venture With Cancer Council NSWSydneyNew South WalesAustralia
| | - Kyo Farrington
- Department of Pathology and Laboratory MedicineUniversity of CalgaryFoothills Medical CenterCalgaryAlbertaCanada
| | - Gregg S. Nelson
- Department of OncologyDivision of Gynecologic OncologyCumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Prafull Ghatage
- Department of OncologyDivision of Gynecologic OncologyCumming School of MedicineUniversity of CalgaryCalgaryAlbertaCanada
| | - Cheng‐Han Lee
- Department of Pathology and Laboratory MedicineUniversity of AlbertaEdmontonAlbertaCanada
| | - Marjorie J. Riggan
- Department of Obstetrics and GynecologyDivision of Gynecologic OncologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Adelyn Bolithon
- Adult Cancer ProgramLowy Cancer Research CentreUniversity of NSW SydneySydneyNew South WalesAustralia
- School of Women's and Children's HealthFaculty of Medicine and HealthUniversity of NSW SydneySydneyNew South WalesAustralia
| | - Gordana Popovic
- Stats CentralMark Wainwright Analytical CentreUniversity of NSW SydneySydneyNew South WalesAustralia
| | - Betty Leung
- Prince of Wales Clinical SchoolUNSW Medicine and HealthUniversity of NSW SydneySydneyNew South WalesAustralia
| | - Katrina Tang
- Department of Anatomical PathologyPrince of Wales HospitalSydneyNew South WalesAustralia
| | - Neil Lambie
- Canterbury Health LaboratoriesChristchurchNew Zealand
| | - Joshua Millstein
- Division of BiostatisticsDepartment of Population and Public Health SciencesKeck School of MedicineUniversity of Southern CaliforniaLos AngelesCaliforniaUSA
| | - Jennifer Alsop
- Department of OncologyCentre for Cancer Genetic EpidemiologyUniversity of CambridgeCambridgeUK
| | - Michael S. Anglesio
- Department of Obstetrics and GynecologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- British Columbia's Gynecological Cancer Research Team (OVCARE)University of British ColumbiaBC Cancerand Vancouver General HospitalVancouverBritish ColumbiaCanada
| | - Beyhan Ataseven
- Department of Gynecology and Gynecologic OncologyEvangelische Kliniken Essen‐Mitte (KEM)EssenGermany
- Department of Obstetrics and GynecologyLudwig Maximilian University MunichMunichGermany
| | - Ellen Barlow
- Gynaecological Cancer CentreRoyal Hospital for WomenSydneyNew South WalesAustralia
| | - Matthias W. Beckmann
- Department of Gynecology and ObstetricsComprehensive Cancer Center Erlangen‐EMNFriedrich‐Alexander University Erlangen‐NurembergUniversity Hospital ErlangenErlangenGermany
| | - Jessica Berger
- Division of Gynecologic OncologyDepartment of Obstetrics, Gynecology and Reproductive SciencesUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Christiani Bisinotto
- Department of Gynecology and ObstetricsRibeirão Preto Medical SchoolUniversity of São PauloRibeirão PretoBrazil
| | - Hans Bösmüller
- Institute of Pathology and NeuropathologyTuebingen University HospitalTuebingenGermany
| | - Jessica Boros
- Centre for Cancer ResearchThe Westmead Institute for Medical ResearchUniversity of SydneySydneyNew South WalesAustralia
- Department of Gynaecological OncologyWestmead HospitalSydneyNew South WalesAustralia
- Discipline of Obstetrics and GynaecologyThe University of SydneySydneyNew South WalesAustralia
| | - Alison H. Brand
- Department of Gynaecological OncologyWestmead HospitalSydneyNew South WalesAustralia
- Discipline of Obstetrics and GynaecologyThe University of SydneySydneyNew South WalesAustralia
| | - Angela Brooks‐Wilson
- Canada's Michael Smith Genome Sciences CentreBC CancerVancouverBritish ColumbiaCanada
| | - Sara Y. Brucker
- Department of Women's HealthTuebingen University HospitalTuebingenGermany
| | - Michael E. Carney
- Department of Obstetrics and GynecologyJohn A. Burns School of MedicineUniversity of HawaiiHonoluluHawaiiUSA
| | - Yovanni Casablanca
- Uniformed Services of the Health Sciences Gynecologic Cancer Center of ExcellenceBethesdaMarylandUSA
| | | | - Paul A. Cohen
- Department of Gynaecological OncologySt John of God Subiaco HospitalSubiacoWestern AustraliaAustralia
- Division of Obstetrics and GynaecologyMedical SchoolUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
| | - Thomas P. Conrads
- Women's Health Integrated Research CenterInova Health SystemFalls ChurchVirginiaUSA
| | - Linda S. Cook
- EpidemiologySchool of Public HealthUniversity of ColoradoAuroraColoradoUSA
- Community Health SciencesUniversity of CalgaryCalgaryAlbertaCanada
| | - Penny Coulson
- Division of Genetics and EpidemiologyThe Institute of Cancer ResearchLondonUK
| | - Madeleine Courtney‐Brooks
- Division of Gynecologic OncologyDepartment of Obstetrics, Gynecology and Reproductive SciencesUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Daniel W. Cramer
- Obstetrics and Gynecology Epidemiology CenterDepartment of Obstetrics and GynecologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Philip Crowe
- Prince of Wales Clinical SchoolUNSW Medicine and HealthUniversity of NSW SydneySydneyNew South WalesAustralia
- Department of SurgeryPrince of Wales Private HospitalRandwickNew South WalesAustralia
| | - Julie M. Cunningham
- Department of Laboratory Medicine and PathologyMayo ClinicRochesterMinnesotaUSA
| | - Cezary Cybulski
- Department of Genetics and PathologyInternational Hereditary Cancer CenterPomeranian Medical UniversitySzczecinPoland
| | - Kathleen M. Darcy
- Gynecologic Cancer Center of ExcellenceDepartment of Gynecologic Surgery and ObstetricsUniformed Services University of the Health SciencesWalter Reed National Military Medical CenterBethesdaMarylandUSA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, IncBethesdaMarylandUSA
| | - Mona A. El‐Bahrawy
- Department of Metabolism, Digestion and ReproductionImperial College LondonHammersmith HospitalLondonUK
| | - Esther Elishaev
- Department of PathologyUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Ramona Erber
- Institute of PathologyComprehensive Cancer Center Erlangen‐EMNFriedrich‐Alexander University Erlangen‐NurembergUniversity Hospital ErlangenErlangenGermany
| | - Rhonda Farrell
- Prince of Wales Private HospitalRandwickNew South WalesAustralia
| | - Sian Fereday
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Anna Fischer
- Institute of Pathology and NeuropathologyTuebingen University HospitalTuebingenGermany
| | - María J. García
- Computational Oncology GroupStructural Biology ProgrammeSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Simon A. Gayther
- Center for Bioinformatics and Functional Genomics and the Cedars Sinai Genomics CoreCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | | | - C. Blake Gilks
- Department of Pathology and Laboratory MedicineUniversity of British ColumbiaVancouverBritish ColumbiaCanada
| | - AOCS Group
- Centre for Cancer ResearchThe Westmead Institute for Medical ResearchUniversity of SydneySydneyNew South WalesAustralia
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- QIMR Berghofer Medical Research InstituteBrisbaneQueenslandAustralia
| | - Marcel Grube
- Department of Women's HealthTuebingen University HospitalTuebingenGermany
| | - Paul R. Harnett
- Discipline of Obstetrics and GynaecologyThe University of SydneySydneyNew South WalesAustralia
- Crown Princess Mary Cancer CentreWestmead HospitalSydneyNew South WalesAustralia
| | - Shariska Petersen Harrington
- Division of Gynecologic OncologyDepartment of Obstetrics and GynecologyThe University of Kansas Medical CenterKansas CityKansasUSA
| | - Philipp Harter
- Department of Gynecology and Gynecologic OncologyEvangelische Kliniken Essen‐Mitte (KEM)EssenGermany
- Department of Gynecology and Gynecological OncologyHSK, Dr. Horst‐Schmidt KlinikWiesbadenWiesbadenGermany
| | - Arndt Hartmann
- Institute of PathologyComprehensive Cancer Center Erlangen‐EMNFriedrich‐Alexander University Erlangen‐NurembergUniversity Hospital ErlangenErlangenGermany
| | - Jonathan L. Hecht
- Department of PathologyBeth Israel Deaconess Medical Center and Harvard Medical SchoolBostonMassachusettsUSA
| | | | - Alexander Hein
- Department of Gynecology and ObstetricsComprehensive Cancer Center Erlangen‐EMNFriedrich‐Alexander University Erlangen‐NurembergUniversity Hospital ErlangenErlangenGermany
| | - Florian Heitz
- Department of Gynecology and Gynecologic OncologyEvangelische Kliniken Essen‐Mitte (KEM)EssenGermany
- Department of Gynecology and Gynecological OncologyHSK, Dr. Horst‐Schmidt KlinikWiesbadenWiesbadenGermany
- Center for PathologyEvangelische Kliniken Essen‐MitteEssenGermany
| | - Joy Hendley
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | | | | | - Sabine Heublein
- Department of Obstetrics and GynecologyUniversity Hospital HeidelbergHeidelbergGermany
| | - Akira Hirasawa
- Department of Clinical Genomic MedicineGraduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayama UniversityOkayamaJapan
| | - Estrid Høgdall
- Department of PathologyHerlev HospitalUniversity of CopenhagenCopenhagenDenmark
| | - Claus K. Høgdall
- Department of GynaecologyRigshospitaletUniversity of CopenhagenCopenhagenDenmark
| | - Hugo M. Horlings
- Division of Molecular PathologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - David G. Huntsman
- Department of Obstetrics and GynecologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- Department of Molecular OncologyBC Cancer Research CentreVancouverBritish ColumbiaCanada
| | - Tomasz Huzarski
- Department of Genetics and PathologyInternational Hereditary Cancer CenterPomeranian Medical UniversitySzczecinPoland
- Department of Genetics and PathologyUniversity of Zielona GoraZielona GoraPoland
| | - Andrea Jewell
- Division of Gynecologic OncologyDepartment of Obstetrics and GynecologyThe University of Kansas Medical CenterKansas CityKansasUSA
| | | | - Michael E. Jones
- Division of Genetics and EpidemiologyThe Institute of Cancer ResearchLondonUK
| | - Scott H. Kaufmann
- Division of Oncology Research and Department of Molecular Pharmacology & Experimental TherapeuticsMayo ClinicRochesterMinnesotaUSA
| | - Catherine J. Kennedy
- Centre for Cancer ResearchThe Westmead Institute for Medical ResearchUniversity of SydneySydneyNew South WalesAustralia
- Department of Gynaecological OncologyWestmead HospitalSydneyNew South WalesAustralia
- Discipline of Obstetrics and GynaecologyThe University of SydneySydneyNew South WalesAustralia
| | - Dineo Khabele
- Division of Gynecologic OncologyDepartment of Obstetrics and GynecologyWashington University in St. LouisSt. LouisMissouriUSA
| | | | - Roy F. P. M. Kruitwagen
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtThe Netherlands
- GROW – School for Oncology and ReproductionMaastricht University Medical CenterMaastrichtThe Netherlands
| | - Diether Lambrechts
- Department of Human GeneticsLaboratory for Translational GeneticsKU LeuvenLeuvenBelgium
- VIB Center for Cancer BiologyVIBLeuvenBelgium
| | - Nhu D. Le
- Cancer Control ResearchBC Cancer AgencyVancouverBritish ColumbiaCanada
| | - Marcin Lener
- International Hereditary Cancer CenterDepartment of Genetics and PathologyPomeranian Medical University in SzczecinSzczecinPoland
| | - Jenny Lester
- David Geffen School of MedicineDepartment of Obstetrics and GynecologyUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Yee Leung
- Division of Obstetrics and GynaecologyFaculty of Health and Medical SciencesUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
- Department of Gynaecological OncologyKing Edward Memorial HospitalSubiacoWestern AustraliaAustralia
- Australia New Zealand Gynaecological Oncology GroupCamperdownAustralia
| | - Anna Linder
- Department of Obstetrics and GynecologyInst of Clinical Science, Sahlgrenska Center for Cancer ResearchUniversity of GothenburgGothenburgSweden
| | - Liselore Loverix
- Division of Gynecologic OncologyDepartment of Gynecology and ObstetricsLeuven Cancer InstituteLeuvenBelgium
| | - Jan Lubiński
- Department of Genetics and PathologyInternational Hereditary Cancer CenterPomeranian Medical UniversitySzczecinPoland
| | - Rashna Madan
- Department of Pathology and Laboratory MedicineThe University of Kansas Medical CenterKansas CityKansasUSA
| | | | - Francesmary Modugno
- Division of Gynecologic OncologyDepartment of Obstetrics, Gynecology and Reproductive SciencesUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
- Department of EpidemiologyUniversity of Pittsburgh School of Public HealthPittsburghPennsylvaniaUSA
- Women's Cancer Research CenterMagee‐Womens Research Institute and Hillman Cancer CenterPittsburghPennsylvaniaUSA
| | - Susan L. Neuhausen
- Department of Population SciencesBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Alexander Olawaiye
- Division of Gynecologic OncologyDepartment of Obstetrics, Gynecology and Reproductive SciencesUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Siel Olbrecht
- Division of Gynecologic OncologyDepartment of Gynecology and ObstetricsLeuven Cancer InstituteLeuvenBelgium
| | - Sandra Orsulic
- David Geffen School of MedicineDepartment of Obstetrics and GynecologyUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - José Palacios
- Department of PathologyHospital Ramón y CajalInstituto Ramon y Cajal de Investigación Sanitaria (IRyCIS)CIBERONCUniversidad de AlcaláMadridSpain
| | - Celeste Leigh Pearce
- Department of EpidemiologyUniversity of Michigan School of Public HealthAnn ArborMichiganUSA
| | - Malcolm C. Pike
- Department of Epidemiology and BiostatisticsMemorial Sloan‐Kettering Cancer CenterNew YorkNew YorkUSA
- Department of Population Health and Public Health SciencesKeck School of MedicineUniversity of Southern California Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Carmel M. Quinn
- The Health Precincts BiobankUNSW Biospecimen ServicesMark Wainwright Analytical CentreUniversity of NSW SydneySydneyNew South WalesAustralia
| | - Ganendra Raj Mohan
- Department of Gynaecological OncologySt John of God Subiaco HospitalSubiacoWestern AustraliaAustralia
- Department of Gynaecological OncologyKing Edward Memorial HospitalSubiacoWestern AustraliaAustralia
| | - Cristina Rodríguez‐Antona
- Hereditary Endocrine Cancer GroupSpanish National Cancer Research Center (CNIO)MadridSpain
- Centre for Biomedical Network Research on Rare Diseases (CIBERER)Instituto de Salud Carlos IIIMadridSpain
| | - Matthias Ruebner
- Department of Gynecology and ObstetricsComprehensive Cancer Center Erlangen‐EMNFriedrich‐Alexander University Erlangen‐NurembergUniversity Hospital ErlangenErlangenGermany
| | - Andy Ryan
- MRC Clinical Trials UnitInstitute of Clinical Trials & MethodologyUniversity College LondonLondonUK
- Women's CancerInstitute for Women's HealthUniversity College LondonLondonUK
| | - Stuart G. Salfinger
- Department of Gynaecological OncologySt John of God Subiaco HospitalSubiacoWestern AustraliaAustralia
| | - Naoko Sasamoto
- Obstetrics and Gynecology Epidemiology CenterDepartment of Obstetrics and GynecologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Joellen M. Schildkraut
- Department of EpidemiologyRollins School of Public HealthEmory UniversityAtlantaGeorgiaUSA
| | | | - Mitul Shah
- Department of OncologyCentre for Cancer Genetic EpidemiologyUniversity of CambridgeCambridgeUK
| | - Raghwa Sharma
- Tissue Pathology and Diagnostic OncologyWestmead HospitalSydneyNew South WalesAustralia
| | | | - Naveena Singh
- Department of PathologyBarts Health National Health Service TrustLondonUK
| | - Gabe S. Sonke
- Department of Medical OncologyThe Netherlands Cancer Institute ‐ Antoni van Leeuwenhoek HospitalAmsterdamThe Netherlands
| | - Linda Steele
- Department of Population SciencesBeckman Research Institute of City of HopeDuarteCaliforniaUSA
| | - Colin J. R. Stewart
- School for Women's and Infants' HealthUniversity of Western AustraliaPerthAustralia
| | - Karin Sundfeldt
- Department of Obstetrics and GynecologyInst of Clinical Science, Sahlgrenska Center for Cancer ResearchUniversity of GothenburgGothenburgSweden
| | - Anthony J. Swerdlow
- Division of Genetics and EpidemiologyThe Institute of Cancer ResearchLondonUK
- Division of Breast Cancer ResearchThe Institute of Cancer ResearchLondonUK
| | - Aline Talhouk
- Department of Obstetrics and GynecologyUniversity of British ColumbiaVancouverBritish ColumbiaCanada
- British Columbia's Gynecological Cancer Research Team (OVCARE)University of British ColumbiaBC Cancerand Vancouver General HospitalVancouverBritish ColumbiaCanada
| | - Adeline Tan
- Division of Obstetrics and GynaecologyFaculty of Health and Medical SciencesUniversity of Western AustraliaCrawleyWestern AustraliaAustralia
- Gynaepath WAClinipath (Sonic Healthcare)Osbourne ParkAustralia
| | - Sarah E. Taylor
- Division of Gynecologic OncologyDepartment of Obstetrics, Gynecology and Reproductive SciencesUniversity of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| | - Kathryn L. Terry
- Obstetrics and Gynecology Epidemiology CenterDepartment of Obstetrics and GynecologyBrigham and Women's Hospital and Harvard Medical SchoolBostonMassachusettsUSA
- Department of EpidemiologyHarvard T.H. Chan School of Public HealthBostonMassachusettsUSA
| | - Aleksandra Tołoczko
- Department of Genetics and PathologyPomeranian Medical UniversitySzczecinPoland
| | - Nadia Traficante
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Koen K. Van de Vijver
- Department of PathologyGhent University HospitalCancer Research Institute Ghent (CRIG)GhentBelgium
- Department of PathologyAntwerp University HospitalAntwerpBelgium
| | - Maaike A. van der Aa
- Department of ResearchNetherlands Comprehensive Cancer Organization (IKNL)UtrechtThe Netherlands
| | - Toon Van Gorp
- Division of Gynecologic OncologyDepartment of Gynecology and ObstetricsLeuven Cancer InstituteLeuvenBelgium
| | - Els Van Nieuwenhuysen
- Division of Gynecologic OncologyDepartment of Gynecology and ObstetricsLeuven Cancer InstituteLeuvenBelgium
| | - Lilian van‐Wagensveld
- Department of Obstetrics and GynecologyMaastricht University Medical CentreMaastrichtThe Netherlands
- GROW – School for Oncology and ReproductionMaastricht University Medical CenterMaastrichtThe Netherlands
- Department of ResearchNetherlands Comprehensive Cancer Organization (IKNL)UtrechtThe Netherlands
| | - Ignace Vergote
- Division of Gynecologic OncologyDepartment of Gynecology and ObstetricsLeuven Cancer InstituteLeuvenBelgium
| | - Robert A. Vierkant
- Department of Quantitative Health SciencesDivision of Clinical Trials and BiostatisticsMayo ClinicRochesterMinnesotaUSA
| | - Chen Wang
- Department of Quantitative Health SciencesDivision of Computational BiologyMayo ClinicRochesterMinnesotaUSA
| | | | - Stacey J. Winham
- Department of Quantitative Health SciencesDivision of Computational BiologyMayo ClinicRochesterMinnesotaUSA
| | - Anna H. Wu
- Department of Population Health and Public Health SciencesKeck School of MedicineUniversity of Southern California Norris Comprehensive Cancer CenterLos AngelesCaliforniaUSA
| | - Javier Benitez
- Centre for Biomedical Network Research on Rare Diseases (CIBERER)Instituto de Salud Carlos IIIMadridSpain
- Human Genetics GroupSpanish National Cancer Research Centre (CNIO)MadridSpain
| | - Andrew Berchuck
- Department of Obstetrics and GynecologyDivision of Gynecologic OncologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | | | - Anna DeFazio
- The Daffodil CentreThe University of SydneyA Joint Venture With Cancer Council NSWSydneyNew South WalesAustralia
- Centre for Cancer ResearchThe Westmead Institute for Medical ResearchUniversity of SydneySydneyNew South WalesAustralia
- Department of Gynaecological OncologyWestmead HospitalSydneyNew South WalesAustralia
- Discipline of Obstetrics and GynaecologyThe University of SydneySydneyNew South WalesAustralia
| | - Peter A. Fasching
- Department of Gynecology and ObstetricsComprehensive Cancer Center Erlangen‐EMNFriedrich‐Alexander University Erlangen‐NurembergUniversity Hospital ErlangenErlangenGermany
| | - Ellen L. Goode
- Department of Quantitative Health SciencesDivision of EpidemiologyMayo ClinicRochesterMinnesotaUSA
| | - Marc T. Goodman
- Cancer Prevention and Control ProgramCedars‐Sinai CancerCedars‐Sinai Medical CenterLos AngelesCaliforniaUSA
| | - Jacek Gronwald
- Department of Genetics and PathologyInternational Hereditary Cancer CenterPomeranian Medical UniversitySzczecinPoland
| | - Beth Y. Karlan
- David Geffen School of MedicineDepartment of Obstetrics and GynecologyUniversity of California at Los AngelesLos AngelesCaliforniaUSA
| | - Stefan Kommoss
- Department of Women's HealthTuebingen University HospitalTuebingenGermany
| | - Usha Menon
- MRC Clinical Trials UnitInstitute of Clinical Trials & MethodologyUniversity College LondonLondonUK
| | - Hans‐Peter Sinn
- Institute of PathologyHeidelberg University HospitalHeidelbergGermany
| | - Annette Staebler
- Institute of Pathology and NeuropathologyTuebingen University HospitalTuebingenGermany
| | - James D. Brenton
- Cancer Research UK Cambridge InstituteUniversity of CambridgeCambridgeUK
| | - David D. Bowtell
- Peter MacCallum Cancer CentreMelbourneVictoriaAustralia
- Sir Peter MacCallum Department of OncologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Paul D. P. Pharoah
- Department of OncologyCentre for Cancer Genetic EpidemiologyUniversity of CambridgeCambridgeUK
- Department of Public Health and Primary CareCentre for Cancer Genetic EpidemiologyUniversity of CambridgeCambridgeUK
| | - Susan J. Ramus
- School of Clinical MedicineUNSW Medicine and HealthUniversity of NSW SydneySydneyNew South WalesAustralia
- Adult Cancer ProgramLowy Cancer Research CentreUniversity of NSW SydneySydneyNew South WalesAustralia
| | - Martin Köbel
- Department of Pathology and Laboratory MedicineUniversity of CalgaryFoothills Medical CenterCalgaryAlbertaCanada
| |
Collapse
|
11
|
Khlebodarova TM, Demenkov PS, Ivanisenko TV, Antropova EA, Lavrik IN, Ivanisenko VA. Primary and Secondary micro-RNA Modulation the Extrinsic Pathway of Apoptosis in Hepatocellular Carcinoma. Mol Biol 2023; 57:165-175. [PMID: 37128213 PMCID: PMC10131518 DOI: 10.1134/s0026893323020103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/09/2022] [Accepted: 09/09/2022] [Indexed: 05/03/2023]
Abstract
Abstract-One of the most common malignant liver diseases is hepatocellular carcinoma, which has a high recurrence rate and a low five-year survival rate. It is very heterogeneous both in structure and between patients, which complicates the diagnosis, prognosis and response to treatment. In this regard, an individualized, patient-centered approach becomes important, in which the use of mimetics and hsa-miRNA inhibitors involved in the pathogenesis of the disease may be determinative. From this point of view hsa-miRNAs are of interest, their aberrant expression is associated with poor prognosis for patients and is associated with tumor progression due to dysregulation of programmed cell death (apoptosis). However, the effect of hsa-miRNA on tumor development depends not only on its direct effect on expression of genes, the primary targets, but also on secondary targets mediated by regulatory pathways. While the former are actively studied, the role of secondary targets of these hsa-miRNAs in modulating apoptosis is still unclear. The present work summarizes data on hsa-miRNAs whose primary targets are key genes of the extrinsic pathway of apoptosis. Their aberrant expression is associated with early disease relapse and poor patient outcome. For these hsa-miRNAs, using the software package ANDSystem, we reconstructed the regulation of the expression of secondary targets and analyzed their impact on the activity of the extrinsic pathway of apoptosis. The potential effect of hsa-miRNAs mediated by action on secondary targets is shown to negatively correlate with the number of primary targets. It is also shown that hsa-miR-373, hsa-miR-106b and hsa-miR-96 have the highest priority as markers of hepatocellular carcinoma, whose action on secondary targets enhances their anti-apoptotic effect.
Collapse
Affiliation(s)
- T. M. Khlebodarova
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Kurchatov Genomic Center, Institute of Cytology and Genetics Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - P. S. Demenkov
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Kurchatov Genomic Center, Institute of Cytology and Genetics Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - T. V. Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Kurchatov Genomic Center, Institute of Cytology and Genetics Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - E. A. Antropova
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| | - I. N. Lavrik
- Translational Inflammation Research, Medical Faculty, Otto von Guericke University Magdeburg, 39106 Magdeburg, Germany
| | - V. A. Ivanisenko
- Institute of Cytology and Genetics, Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
- Kurchatov Genomic Center, Institute of Cytology and Genetics Siberian Branch, Russian Academy of Sciences, 630090 Novosibirsk, Russia
| |
Collapse
|
12
|
Sartorius K, Antwi SO, Chuturgoon A, Roberts LR, Kramvis A. RNA Therapeutic Options to Manage Aberrant Signaling Pathways in Hepatocellular Carcinoma: Dream or Reality? Front Oncol 2022; 12:891812. [PMID: 35600358 PMCID: PMC9115561 DOI: 10.3389/fonc.2022.891812] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 04/04/2022] [Indexed: 11/24/2022] Open
Abstract
Despite the early promise of RNA therapeutics as a magic bullet to modulate aberrant signaling in cancer, this field remains a work-in-progress. Nevertheless, RNA therapeutics is now a reality for the treatment of viral diseases (COVID-19) and offers great promise for cancer. This review paper specifically investigates RNAi as a therapeutic option for HCC and discusses a range of RNAi technology including anti-sense oligonucleotides (ASOs), Aptamers, small interfering RNA (siRNA), ribozymes, riboswitches and CRISPR/Cas9 technology. The use of these RNAi based interventions is specifically outlined in three primary strategies, namely, repressing angiogenesis, the suppression of cell proliferation and the promotion of apoptosis. We also discuss some of the inherent chemical and delivery problems, as well as targeting issues and immunogenic reaction to RNAi interventions.
Collapse
Affiliation(s)
- Kurt Sartorius
- Hepatitis Virus Diversity Research Unit, School of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
- The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL, United States
- Department of Surgery, KZN Kwazulu-Natal (UKZN) Gastrointestinal Cancer Research Centre, Durban, South Africa
| | - Samuel O. Antwi
- The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL, United States
- Division of Epidemiology, Department of Quantitative Health Sciences, Mayo Clinic, Jacksonville, FL, United States
| | - Anil Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, College of Health Science, University of KwaZulu-Natal, Durban, South Africa
| | - Lewis R. Roberts
- The Africa Hepatopancreatobiliary Cancer Consortium (AHPBCC), Mayo Clinic, Jacksonville, FL, United States
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, United States
| | - Anna Kramvis
- Hepatitis Virus Diversity Research Unit, School of Internal Medicine, University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
13
|
Limas JC, Littlejohn AN, House AM, Kedziora KM, Mouery BL, Ma B, Fleifel D, Walens A, Aleman MM, Dominguez D, Cook JG. Quantitative profiling of adaptation to cyclin E overproduction. Life Sci Alliance 2022; 5:e202201378. [PMID: 35173014 PMCID: PMC8860095 DOI: 10.26508/lsa.202201378] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 01/28/2022] [Accepted: 01/31/2022] [Indexed: 01/03/2023] Open
Abstract
Cyclin E/CDK2 drives cell cycle progression from G1 to S phase. Despite the toxicity of cyclin E overproduction in mammalian cells, the cyclin E gene is overexpressed in some cancers. To further understand how cells can tolerate high cyclin E, we characterized non-transformed epithelial cells subjected to chronic cyclin E overproduction. Cells overproducing cyclin E, but not cyclins D or A, briefly experienced truncated G1 phases followed by a transient period of DNA replication origin underlicensing, replication stress, and impaired proliferation. Individual cells displayed substantial intercellular heterogeneity in cell cycle dynamics and CDK activity. Each phenotype improved rapidly despite high cyclin E-associated activity. Transcriptome analysis revealed adapted cells down-regulated a cohort of G1-regulated genes. Withdrawing cyclin E from adapted cells only partially reversed underlicensing indicating that adaptation is at least partly non-genetic. This study provides evidence that mammalian cyclin E/CDK inhibits origin licensing indirectly through premature S phase onset and provides mechanistic insight into the relationship between CDKs and licensing. It serves as an example of oncogene adaptation that may recapitulate molecular changes during tumorigenesis.
Collapse
Affiliation(s)
- Juanita C Limas
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amiee N Littlejohn
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Amy M House
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Katarzyna M Kedziora
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Bioinformatics and Analytics Research Collaborative (BARC), University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Brandon L Mouery
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Boyang Ma
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Dalia Fleifel
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Andrea Walens
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Maria M Aleman
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Daniel Dominguez
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jeanette Gowen Cook
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Department of Biochemistry and Biophysics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Curriculum in Genetics and Molecular Biology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
- Lineberger Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
14
|
Abstract
Cyclin-dependent kinases 4 and 6 (CDK4 and CDK6) and their activating partners, D-type cyclins, link the extracellular environment with the core cell cycle machinery. Constitutive activation of cyclin D–CDK4/6 represents the driving force of tumorigenesis in several cancer types. Small-molecule inhibitors of CDK4/6 have been used with great success in the treatment of hormone receptor–positive breast cancers and are in clinical trials for many other tumor types. Unexpectedly, recent work indicates that inhibition of CDK4/6 affects a wide range of cellular functions such as tumor cell metabolism and antitumor immunity. We discuss how recent advances in understanding CDK4/6 biology are opening new avenues for the future use of cyclin D–CDK4/6 inhibitors in cancer treatment.
Collapse
Affiliation(s)
- Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Yan Geng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| |
Collapse
|
15
|
Liang Y, Fan Y, Liu Y, Fan H. HNRNPU promotes the progression of hepatocellular carcinoma by enhancing CDK2 transcription. Exp Cell Res 2021; 409:112898. [PMID: 34737140 DOI: 10.1016/j.yexcr.2021.112898] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/29/2021] [Accepted: 10/25/2021] [Indexed: 02/08/2023]
Abstract
The nuclear matrix-associated protein Heterogeneous Nuclear Ribonucleoprotein U (HNRNPU), also known as SAF-A, is known to maintain active chromatin structure in mouse hepatocytes. However, the functional roles and molecular mechanisms of HNRNPU in the development of hepatocellular carcinoma (HCC) remain largely unknown. Herein, we found that HNRNPU was upregulated in HCC, and the proliferation of HCC cells was inhibited in vitro and in vivo upon HNRNPU knockdown. Moreover, the upregulation of HNRNPU was correlated with poor prognosis in HCC. Mechanistically, HNRNPU bound to the CDK2 gene locus, a key factor in cell cycle regulation, where it was enriched with H3K27 acetylation (H3K27ac), H3K9 acetylation (H3K9ac), and H3K4 mono-methylation (H3K4me1). Furthermore, HNRNPU knockdown reduced the levels of H3K27ac and H3K9ac at the binding site, where the levels of H3K27 tri-methylation (H3K27me3) were increased, eventually leading to the downregulation of CDK2. Collectively, our results provide a new mechanism whereby HNRNPU promotes HCC development by enhancing the transcription of CDK2.
Collapse
Affiliation(s)
- Yi Liang
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Yao Fan
- Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, 646000, China
| | - Yu Liu
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Hui Fan
- The Key Laboratory of Molecular Biology of Infectious Diseases Designated By the Chinese Ministry of Education, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
16
|
Sonntag R, Penners C, Kohlhepp M, Haas U, Lambertz D, Kroh A, Cramer T, Ticconi F, Costa IG, Tacke F, Gassler N, Trautwein C, Liedtke C. Cyclin E1 in Murine and Human Liver Cancer: A Promising Target for Therapeutic Intervention during Tumour Progression. Cancers (Basel) 2021; 13:5680. [PMID: 34830835 PMCID: PMC8616292 DOI: 10.3390/cancers13225680] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 11/03/2021] [Accepted: 11/04/2021] [Indexed: 12/24/2022] Open
Abstract
Cyclin E1 (CCNE1) is a regulatory subunit of Cyclin-dependent kinase 2 (CDK2) and is thought to control the transition of quiescent cells into cell cycle progression. Recently, we identified CCNE1 and CDK2 as key factors for the initiation of hepatocellular carcinoma (HCC). In the present study, we dissected the contributions of CCNE1 and CDK2 for HCC progression in mice and patients. Therefore, we generated genetically modified mice allowing inducible deletion of Ccne1 or Cdk2. After initiation of HCC, using the hepatocarcinogen diethylnitrosamine (DEN), we deleted Ccne1 or Cdk2 and subsequently analysed HCC progression. The relevance of CCNE1 or CDK2 for human HCC progression was investigated by in silico database analysis. Interventional deletion of Ccne1, but not of Cdk2, substantially reduced the HCC burden in mice. Ccne1-deficient HCCs were characterised by attenuated proliferation, impaired DNA damage response and downregulation of markers for stemness and microinvasion. Additionally, the tumour microenvironment of Ccne1-deficient mice showed a reduction in immune mediators, myeloid cells and cancer-associated fibroblasts. In sharp contrast, Cdk2 was dispensable for HCC progression in mice. In agreement with our mouse data, CCNE1 was overexpressed in HCC patients independent of risk factors, and associated with reduced disease-free survival, a common signature for enhanced chromosomal instability, proliferation, dedifferentiation and invasion. However, CDK2 lacked diagnostic or prognostic value in HCC patients. In summary, CCNE1 drives HCC progression in a CDK2-independent manner in mice and man. Therefore, interventional inactivation of CCNE1 represents a promising strategy the treatment of liver cancer.
Collapse
Affiliation(s)
- Roland Sonntag
- Department of Internal Medicine III, University Hospital RWTH, 52074 Aachen, Germany; (C.P.); (U.H.); (D.L.); (C.T.)
| | - Christian Penners
- Department of Internal Medicine III, University Hospital RWTH, 52074 Aachen, Germany; (C.P.); (U.H.); (D.L.); (C.T.)
| | - Marlene Kohlhepp
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité University Medicine Berlin, 13353 Berlin, Germany; (M.K.); (F.T.)
| | - Ute Haas
- Department of Internal Medicine III, University Hospital RWTH, 52074 Aachen, Germany; (C.P.); (U.H.); (D.L.); (C.T.)
| | - Daniela Lambertz
- Department of Internal Medicine III, University Hospital RWTH, 52074 Aachen, Germany; (C.P.); (U.H.); (D.L.); (C.T.)
| | - Andreas Kroh
- Department of General, Visceral and Transplantation Surgery, University Hospital RWTH, 52074 Aachen, Germany; (A.K.); (T.C.)
| | - Thorsten Cramer
- Department of General, Visceral and Transplantation Surgery, University Hospital RWTH, 52074 Aachen, Germany; (A.K.); (T.C.)
- Department of Surgery, Maastricht University Medical Center, 6200 MD Maastricht, The Netherlands
- ESCAM—European Surgery Center Aachen Maastricht, 52074 Aachen, Germany
- ESCAM—European Surgery Center Aachen Maastricht, 6200 MD Maastricht, The Netherlands
| | - Fabio Ticconi
- IZKF Research Group Computational Biology and Bioinformatics, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany; (F.T.); (I.G.C.)
| | - Ivan G. Costa
- IZKF Research Group Computational Biology and Bioinformatics, Helmholtz Institute for Biomedical Engineering, RWTH Aachen University, 52074 Aachen, Germany; (F.T.); (I.G.C.)
| | - Frank Tacke
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum and Campus Charité Mitte, Charité University Medicine Berlin, 13353 Berlin, Germany; (M.K.); (F.T.)
| | - Nikolaus Gassler
- Section of Pathology, Institute of Forensic Medicine University Hospital Jena, 07747 Jena, Germany;
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital RWTH, 52074 Aachen, Germany; (C.P.); (U.H.); (D.L.); (C.T.)
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital RWTH, 52074 Aachen, Germany; (C.P.); (U.H.); (D.L.); (C.T.)
| |
Collapse
|
17
|
Jiang D, Xu J, Liu S, Nasser MI, Wei W, Mao T, Liu X, Zou X, Li J, Li X. Rosmanol induces breast cancer cells apoptosis by regulating PI3K/AKT and STAT3/JAK2 signaling pathways. Oncol Lett 2021; 22:631. [PMID: 34267823 PMCID: PMC8258625 DOI: 10.3892/ol.2021.12892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 06/07/2021] [Indexed: 12/31/2022] Open
Abstract
Breast cancer is one of the most frequently diagnosed cancers amongst women; however, there is currently no effective treatment. Natural compounds are considered to contribute to cancer prevention and have a pivotal role in modulating apoptosis. Rosmanol is a phenolic diterpene compound with antioxidant and anti-inflammatory properties. In the present study, the effects of Rosmanol on breast cancer cell proliferation/apoptosis were investigated, and it was demonstrated that it inhibited the proliferation of MCF-7 and MDA-MB 231 cells but did not have a significant effect on normal human breast MCF-10A cells. In addition, the apoptotic process was accelerated by Rosmanol, through mitochondrial pathways and reactive oxygen species (ROS) production caused by DNA damage, which function further demonstrated by the attenuation and addition of the ROS inhibitor, N-acetyl-cysteine. It was also demonstrated that Rosmanol accelerated cell apoptosis, and arrested breast cancer cells in the S phase. Moreover, Rosmanol inhibited proliferation and promoted apoptosis of cancer cells via the inhibition of ERK and STAT3 signals, attributable to the increase in p-p38, the overexpression of protein inhibitor of activated STAT3, and the decrease in PI3K/AKT, ERK and JAK2/STAT3.
Collapse
Affiliation(s)
- Dongjun Jiang
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Jiaqi Xu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Sitong Liu
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Moussa Ide Nasser
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Wei Wei
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Tianjiao Mao
- Department of Stomatogy, Affiliated Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Xintong Liu
- Bioprobe Application Research Unit, Chemical Biology Department, RIKEN-Max Planck Joint Research Division, RIkagaku KENkyusho/Institute of Physical and Chemical Research (RIKEN) Center for Sustainable Resource Science, Wako, Saitama 351-0198, Japan
| | - Xiaopan Zou
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| | - Jiang Li
- Department of Stomatogy, Affiliated Stomatological Hospital of Guangzhou Medical University, Guangzhou, Guangdong 510180, P.R. China
| | - Xiaomeng Li
- The Key Laboratory of Molecular Epigenetics of Ministry of Education (MOE), School of Life Sciences, Northeast Normal University, Changchun, Jilin 130024, P.R. China
| |
Collapse
|
18
|
Suski JM, Braun M, Strmiska V, Sicinski P. Targeting cell-cycle machinery in cancer. Cancer Cell 2021; 39:759-778. [PMID: 33891890 PMCID: PMC8206013 DOI: 10.1016/j.ccell.2021.03.010] [Citation(s) in RCA: 302] [Impact Index Per Article: 75.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 02/09/2021] [Accepted: 03/26/2021] [Indexed: 12/19/2022]
Abstract
Abnormal activity of the core cell-cycle machinery is seen in essentially all tumor types and represents a driving force of tumorigenesis. Recent studies revealed that cell-cycle proteins regulate a wide range of cellular functions, in addition to promoting cell division. With the clinical success of CDK4/6 inhibitors, it is becoming increasingly clear that targeting individual cell-cycle components may represent an effective anti-cancer strategy. Here, we discuss the potential of inhibiting different cell-cycle proteins for cancer therapy.
Collapse
Affiliation(s)
- Jan M Suski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Marcin Braun
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Chair of Oncology, Medical University of Lodz, 92-213 Lodz, Poland
| | - Vladislav Strmiska
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
19
|
Donaldson JS, Dale MP, Rosser SJ. Decoupling Growth and Protein Production in CHO Cells: A Targeted Approach. Front Bioeng Biotechnol 2021; 9:658325. [PMID: 34150726 PMCID: PMC8207133 DOI: 10.3389/fbioe.2021.658325] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 04/09/2021] [Indexed: 11/28/2022] Open
Abstract
Fed-batch cultures of Chinese Hamster Ovary cells have been used to produce high quantities of biotherapeutics, particularly monoclonal antibodies. However, a growing number of next-generation biotherapeutics, such as bi-specific antibodies and fusion proteins, are difficult to express using standard fed-batch processes. Decoupling cell growth and biotherapeutic production is becoming an increasingly desired strategy for the biomanufacturing industry, especially for difficult-to-express products. Cells are grown to a high cell density in the absence of recombinant protein production (the growth phase), then expression of the recombinant protein is induced and cell proliferation halted (the production phase), usually by combining an inducible gene expression system with a proliferation control strategy. Separating the growth and production phases allows cell resources to be more efficiently directed toward either growth or production, improving growth characteristics and enhancing the production of difficult to express proteins. However, current mammalian cell proliferation control methods rely on temperature shifts and chemical agents, which interact with many non-proliferation pathways, leading to variable impacts on product quality and culture viability. Synthetic biology offers an alternative approach by strategically targeting proliferation pathways to arrest cell growth but have largely remained unused in industrial bioproduction. Due to recent developments in microbial decoupling systems and advances in available mammalian cell engineering tools, we propose that the synthetic biology approach to decoupling growth and production needs revisiting.
Collapse
Affiliation(s)
- James S Donaldson
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Matthew P Dale
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| | - Susan J Rosser
- School of Biological Sciences, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
20
|
Chu C, Geng Y, Zhou Y, Sicinski P. Cyclin E in normal physiology and disease states. Trends Cell Biol 2021; 31:732-746. [PMID: 34052101 DOI: 10.1016/j.tcb.2021.05.001] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 04/29/2021] [Accepted: 05/03/2021] [Indexed: 01/17/2023]
Abstract
E-type cyclins, collectively called cyclin E, represent key components of the core cell cycle machinery. In mammalian cells, two E-type cyclins, E1 and E2, activate cyclin-dependent kinase 2 (CDK2) and drive cell cycle progression by phosphorylating several cellular proteins. Abnormally elevated activity of cyclin E-CDK2 has been documented in many human tumor types. Moreover, cyclin E overexpression mediates resistance of tumor cells to various therapeutic agents. Recent work has revealed that the role of cyclin E extends well beyond cell proliferation and tumorigenesis, and it may regulate a diverse array of physiological and pathological processes. In this review, we discuss these various cyclin E functions and the potential for therapeutic targeting of cyclin E and cyclin E-CDK2 kinase.
Collapse
Affiliation(s)
- Chen Chu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Yan Geng
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Yu Zhou
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA; Sichuan Provincial Key Laboratory for Human Disease Gene Study, Sichuan Academy of Medical Sciences, Sichuan Provincial People's Hospital, University of Electronic Science and Technology, Chengdu, China
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
21
|
Hasanin AH, Habib EK, El Gayar N, Matboli M. Promotive action of 2-acetylaminofluorene on hepatic precancerous lesions initiated by diethylnitrosamine in rats: Molecular study. World J Hepatol 2021; 13:328-342. [PMID: 33815676 PMCID: PMC8006078 DOI: 10.4254/wjh.v13.i3.328] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 12/14/2020] [Accepted: 03/02/2021] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Diethylnitrosamine (DEN) induces hepatic neoplastic lesions over a prolonged period. AIM To investigate the promotive action of 2-acetylaminofluorene (2-AAF) when combined with DEN in order to develop a rat model for induction of precancerous lesion and investigate the molecular mechanism underlying the activity of 2-AAF. METHODS The pre-precancerous lesions were initiated by intraperitoneal injection of DEN for three weeks consecutively, followed by one intraperitoneal injection of 2-AAF at three different doses (100, 200 and 300 mg/kg). Rats were separated into naïve, DEN, DEN + 100 mg 2-AAF, DEN + 200 mg 2-AAF, and DEN + 300 mg 2-AAF groups. Rats were sacrificed after 10 wk and 16 wk. Liver functions, level of alpha-fetoprotein, glutathione S-transferase-P and proliferating cell nuclear antigen staining of liver tissues were performed. The mRNA level of RAB11A, BAX, p53, and Cyclin E and epigenetic regulation by long-noncoding RNA (lncRNA) RP11-513I15.6, miR-1262 (microRNA), and miR-1298 were assessed in the sera and liver tissues of the rats. RESULTS 2-AAF administration significantly increased the percent area of the precancerous foci and cell proliferation along with a significant decrease in RAB11A, BAX, and p53 mRNA, and the increase in Cyclin E mRNA was associated with a marked decrease in lncRNA RP11-513I15.6 expression with a significant increase in both miR-1262 and miR-1298. CONCLUSION 2-AFF promoted hepatic precancerous lesions initiated through DEN by decreasing autophagy, apoptosis, and tumor suppression genes, along with increased cell proliferation, in a time- and dose-dependent manner. These actions were mediated under the epigenetic regulation of lncRNA RP11-513I15.6/miR-1262/miR-1298.
Collapse
Affiliation(s)
- Amany Helmy Hasanin
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo 11318, Egypt
| | - Eman K Habib
- Anatomy and Embryology Department, Faculty of Medicine, Ain Shams University, Cairo 11318, Egypt
| | - Nesreen El Gayar
- Clinical Pharmacology Department, Faculty of Medicine, Ain Shams University, Cairo 11318, Egypt
| | - Marwa Matboli
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo 11381, Egypt.
| |
Collapse
|
22
|
Tatum NJ, Endicott JA. Chatterboxes: the structural and functional diversity of cyclins. Semin Cell Dev Biol 2020; 107:4-20. [PMID: 32414682 DOI: 10.1016/j.semcdb.2020.04.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/28/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022]
Abstract
Proteins of the cyclin family have divergent sequences and execute diverse roles within the cell while sharing a common fold: the cyclin box domain. Structural studies of cyclins have played a key role in our characterization and understanding of cellular processes that they control, though to date only ten of the 29 CDK-activating cyclins have been structurally characterized by X-ray crystallography or cryo-electron microscopy with or without their cognate kinases. In this review, we survey the available structures of human cyclins, highlighting their molecular features in the context of their cellular roles. We pay particular attention to how cyclin activity is regulated through fine control of degradation motif recognition and ubiquitination. Finally, we discuss the emergent roles of cyclins independent of their roles as cyclin-dependent protein kinase activators, demonstrating the cyclin box domain to be a versatile and generalized scaffolding domain for protein-protein interactions across the cellular machinery.
Collapse
Affiliation(s)
- Natalie J Tatum
- Cancer Research UK Newcastle Drug Discovery Unit, Newcastle Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom
| | - Jane A Endicott
- Cancer Research UK Newcastle Drug Discovery Unit, Newcastle Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, United Kingdom.
| |
Collapse
|
23
|
Zhou Y, Geng Y, Zhang Y, Zhou Y, Chu C, Sharma S, Fassl A, Butter D, Sicinski P. The requirement for cyclin E in c-Myc overexpressing breast cancers. Cell Cycle 2020; 19:2589-2599. [PMID: 32975478 DOI: 10.1080/15384101.2020.1804720] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Basal-like triple-negative breast cancers frequently express high levels of c-Myc. This oncoprotein signals to the core cell cycle machinery by impinging on cyclin E. High levels of E-type cyclins (E1 and E2) are often seen in human triple-negative breast tumors. In the current study, we examined the requirement for E-type cyclins in the c-Myc-driven mouse model of breast cancer (MMTV-c-Myc mice). To do so, we crossed cyclin E1- (E1-/-) and E2- (E2-/-) deficient mice with MMTV-c-Myc animals, and observed the resulting cyclin E1-/-/MMTV-c-Myc and cyclin E2-/-/MMTV-c-Myc females for breast cancer incidence. We found that mice lacking cyclins E1 or E2 developed breast cancers like their cyclin Ewild-type counterparts. In contrast, further reduction of the dosage of E-cyclins in cyclin E1-/-E2+/-/MMTV-c-Myc and cyclin E1+/-E2-/-/MMTV-c-Myc animals significantly decreased the incidence of mammary carcinomas, revealing arole for E-cyclins in tumor initiation. We also observed that depletion of E-cyclins in human triple-negative breast cancer cell lines halted cell cycle progression, indicating that E-cyclins are essential for tumor cell proliferation. In contrast, we found that the catalytic partner of E-cyclins, the cyclin-dependent kinase 2 (CDK2), is dispensable for the proliferation of these cells. These results indicate that E-cyclins, but not CDK2, play essential and rate-limiting roles in driving the proliferation of c-Myc overexpressing breast cancer cells.
Collapse
Affiliation(s)
- Yu Zhou
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China , Chengdu, China
| | - Yan Geng
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA
| | - Yujiao Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA
| | - Yubin Zhou
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA.,Sichuan Provincial Key Laboratory for Human Disease Gene Study, Department of Laboratory Medicine, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China , Chengdu, China
| | - Chen Chu
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA
| | - Samanta Sharma
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA
| | - Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA
| | - Deborah Butter
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute and Department of Genetics, Blavatnik Institute, Harvard Medical School , Boston, MA, USA
| |
Collapse
|
24
|
Fassl A, Brain C, Abu-Remaileh M, Stukan I, Butter D, Stepien P, Feit AS, Bergholz J, Michowski W, Otto T, Sheng Q, Loo A, Michael W, Tiedt R, DeAngelis C, Schiff R, Jiang B, Jovanovic B, Nowak K, Ericsson M, Cameron M, Gray N, Dillon D, Zhao JJ, Sabatini DM, Jeselsohn R, Brown M, Polyak K, Sicinski P. Increased lysosomal biomass is responsible for the resistance of triple-negative breast cancers to CDK4/6 inhibition. SCIENCE ADVANCES 2020; 6:eabb2210. [PMID: 32704543 PMCID: PMC7360435 DOI: 10.1126/sciadv.abb2210] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/04/2020] [Indexed: 05/28/2023]
Abstract
Inhibitors of cyclin-dependent kinases CDK4 and CDK6 have been approved for treatment of hormone receptor-positive breast cancers. In contrast, triple-negative breast cancers (TNBCs) are resistant to CDK4/6 inhibition. Here, we demonstrate that a subset of TNBC critically requires CDK4/6 for proliferation, and yet, these TNBC are resistant to CDK4/6 inhibition due to sequestration of CDK4/6 inhibitors into tumor cell lysosomes. This sequestration is caused by enhanced lysosomal biogenesis and increased lysosomal numbers in TNBC cells. We developed new CDK4/6 inhibitor compounds that evade the lysosomal sequestration and are efficacious against resistant TNBC. We also show that coadministration of lysosomotropic or lysosome-destabilizing compounds (an antibiotic azithromycin, an antidepressant siramesine, an antimalaria compound chloroquine) renders resistant tumor cells sensitive to currently used CDK4/6 inhibitors. Lastly, coinhibition of CDK2 arrested proliferation of CDK4/6 inhibitor-resistant cells. These observations may extend the use of CDK4/6 inhibitors to TNBCs that are refractory to current anti-CDK4/6 therapies.
Collapse
Affiliation(s)
- Anne Fassl
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher Brain
- Novartis Institute for Biomedical Research, Cambridge, MA 02139, USA
| | - Monther Abu-Remaileh
- Whitehead Institutes for Biomedical Research and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Departments of Chemical Engineering and Genetics, Stanford University, Stanford, CA 94305, USA
- Institute for Chemistry, Engineering and Medicine for Human Health (ChEM-H), Stanford University, 290 Jane Stanford Way, Stanford, CA 94305, USA
| | - Iga Stukan
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Deborah Butter
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Piotr Stepien
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Avery S. Feit
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Johann Bergholz
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Wojciech Michowski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Tobias Otto
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Qing Sheng
- Novartis Institute for Biomedical Research, Cambridge, MA 02139, USA
| | - Alice Loo
- Novartis Institute for Biomedical Research, Cambridge, MA 02139, USA
| | - Walter Michael
- Novartis Institute for Biomedical Research, Cambridge, MA 02139, USA
| | - Ralph Tiedt
- Novartis Institutes for Biomedical Research, Oncology Disease Area, 4057 Basel, Switzerland
| | - Carmine DeAngelis
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Clinical Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Rachel Schiff
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine and Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Baishan Jiang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Bojana Jovanovic
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Karolina Nowak
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Maria Ericsson
- Electron Microscopy Facility, Harvard Medical School, Boston, MA 02115, USA
| | - Michael Cameron
- Department of Molecular Therapeutics, The Scripps Research Institute, Jupiter, FL 33458, USA
| | - Nathanael Gray
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
| | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jean J. Zhao
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA 02115, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
| | - David M. Sabatini
- Whitehead Institutes for Biomedical Research and Massachusetts Institute of Technology, Cambridge, MA 02142, USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA
- Howard Hughes Medical Institute, Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Rinath Jeselsohn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Myles Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA
- Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Piotr Sicinski
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
- Department of Genetics, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
25
|
Palmer N, Kaldis P. Less-well known functions of cyclin/CDK complexes. Semin Cell Dev Biol 2020; 107:54-62. [PMID: 32386818 DOI: 10.1016/j.semcdb.2020.04.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 04/07/2020] [Accepted: 04/08/2020] [Indexed: 12/31/2022]
Abstract
Cyclin-dependent kinases (CDKs) are activated by cyclins, which play important roles in dictating the actions of CDK/cyclin complexes. Cyclin binding influences the substrate specificity of these complexes in addition to their susceptibility to inhibition or degradation. CDK/cyclin complexes are best known to promote cell cycle progression in the mitotic cell cycle but are also crucial for important cellular processes not strictly associated with cellular division. This chapter primarily explores the understudied topic of CDK/cyclin complex functionality during the DNA damage response. We detail how CDK/cyclin complexes perform dual roles both as targets of DNA damage checkpoint signaling as well as effectors of DNA repair. Additionally, we discuss the potential CDK-independent roles of cyclins in these processes and the impact of such roles in human diseases such as cancer. Our goal is to place the spotlight on these important functions of cyclins either acting as independent entities or within CDK/cyclin complexes which have attracted less attention in the past. We consider that this will be important for a more complete understanding of the intricate functions of cell cycle proteins in the DNA damage response.
Collapse
Affiliation(s)
- Nathan Palmer
- Institute of Molecular and Cell Biology (IMCB), A⁎STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore; National University of Singapore (NUS), Department of Biochemistry, Singapore, 117597, Republic of Singapore
| | - Philipp Kaldis
- Institute of Molecular and Cell Biology (IMCB), A⁎STAR (Agency for Science, Technology and Research), 61 Biopolis Drive, Proteos, Singapore, 138673, Republic of Singapore; National University of Singapore (NUS), Department of Biochemistry, Singapore, 117597, Republic of Singapore; Department of Clinical Sciences, Lund University, Clinical Research Centre (CRC), Box 50332, SE-202 13, Malmö, Sweden.
| |
Collapse
|
26
|
Yang J, Dong Z, Ren A, Fu G, Zhang K, Li C, Wang X, Cui H. Antibiotic tigecycline inhibits cell proliferation, migration and invasion via down-regulating CCNE2 in pancreatic ductal adenocarcinoma. J Cell Mol Med 2020; 24:4245-4260. [PMID: 32141702 PMCID: PMC7171345 DOI: 10.1111/jcmm.15086] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Revised: 10/17/2019] [Accepted: 01/15/2020] [Indexed: 12/17/2022] Open
Abstract
Recently, many researches have reported that antibiotic tigecycline has significant effect on cancer treatment. However, biomedical functions and molecular mechanisms of tigecycline in human pancreatic ductal adenocarcinoma (PDAC) remain unclear. In the current study, we tried to assess the effect of tigecycline in PDAC cells. AsPC‐1 and HPAC cells were treated with indicated concentrations of tigecycline for indicated time, and then, MTT, BrdU and soft agar assay were used to test cell proliferation. The effect of tigecycline on cell cycle and cellular apoptosis was tested by cytometry. Migration and invasion were detected by wound healing assay and transwell migration/invasion assay. Expressions of cell cycle‐related and migration/invasion‐related protein were determined by using Western blot. The results revealed that tigecycline observably suppressed cell proliferation by inducing cell cycle arrest at G0/G1 phase and blocked cell migration/invasion via holding back the epithelial‐mesenchymal transition (EMT) process in PDAC. In addition, tigecycline also remarkably blocked tumorigenecity in vivo. Furthermore, the effects of tigecycline alone or combined with gemcitabine in vitro or on PDAC xenografts were also performed. The results showed that tigecycline enhanced the chemosensitivity of PDAC cells to gemcitabine. Interestingly, we found CCNE2 expression was declined distinctly after tigecycline treatment. Then, CCNE2 was overexpressed to rescue tigecycline‐induced effect. The results showed that CCNE2 overexpression significantly rescued tigecycline‐inhibited cell proliferation and migration/invasion. Collectively, we showed that tigecycline inhibits cell proliferation, migration and invasion via down‐regulating CCNE2, and tigecycline might be used as a potential drug for PDAC treatment alone or combined with gemcitabine.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Zhen Dong
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Aishu Ren
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Gang Fu
- College of Stomatology, Chongqing Medical University, Chongqing, China
| | - Kui Zhang
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Changhong Li
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| | - Xiangwei Wang
- Department of Urology, Carson International Cancer Center, Shenzhen University General Hospital & Shenzhen University Clinical Medical Academy Center, Shenzhen University, Shenzhen, China
| | - Hongjuan Cui
- State Key Laboratory of Silkworm Genome Biology, Institute of Sericulture and Systems Biology, Southwest University, Beibei, Chongqing, China.,Cancer Center, Medical Research Institute, Southwest University, Beibei, Chongqing, China.,Engineering Research Center for Cancer Biomedical and Translational Medicine, Southwest University, Beibei, Chongqing, China.,Chongqing Engineering and Technology Research Center for Silk Biomaterials and Regenerative Medicine, Southwest University, Beibei, Chongqing, China
| |
Collapse
|
27
|
Huang S, Zhang C, Sun C, Hou Y, Zhang Y, Tam NL, Wang Z, Yu J, Huang B, Zhuang H, Zhou Z, Ma Z, Sun Z, He X, Zhou Q, Hou B, Wu L. Obg-like ATPase 1 (OLA1) overexpression predicts poor prognosis and promotes tumor progression by regulating P21/CDK2 in hepatocellular carcinoma. Aging (Albany NY) 2020; 12:3025-3041. [PMID: 32045367 PMCID: PMC7041778 DOI: 10.18632/aging.102797] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 01/12/2020] [Indexed: 12/19/2022]
Abstract
Background: Obg-like ATPase 1 (OLA1) has been found to have a dual role in cancers. However, the relationship between OLA1 and hepatocellular carcinoma (HCC) remains unclear. Results: High expression of OLA1 in HCC was detected in public datasets and clinical samples, and correlated with poor prognosis. Downregulation of OLA1 significantly inhibited the proliferation, migration, invasion and tumorigenicity of HCC cells. Mechanistically, GSEA showed that OLA1 might promote tumor progression by regulating the cell cycle and apoptosis. In addition, OLA1 knockdown resulted in G0/G1 phase arrest and high levels of apoptosis. OLA1 could bind with P21 and upregulate CDK2 expression to promote HCC progression. Conclusions: Overall, these findings uncover a role for OLA1 in regulating the proliferation and apoptosis of HCC cells. Materials and methods: The Cancer Genome Atlas and Gene Expression Omnibus datasets were analyzed to identify gene expression. Immunohistochemistry staining, western blot and real-time polymerase chain reaction were performed to evaluate OLA1 expression in samples. Cell count Kit-8, wound-healing, transwell and flow cytometry assays were used to analyze HCC cell progression. Subcutaneous xenotransplantation models were used to investigate the role of OLA1 in vivo. Coimmunoprecipitation was used to analyze protein interactions.
Collapse
Affiliation(s)
- Shanzhou Huang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Chuanzhao Zhang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Chengjun Sun
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yuchen Hou
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Yixi Zhang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Nga Lei Tam
- The Fifth Affiliated Hospital of Sun Yat-Sen University, Division of Hepatobiliary Surgery, Zhuhai 519000, China
| | - Zekang Wang
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Jia Yu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Bowen Huang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Hongkai Zhuang
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Zixuan Zhou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Zuyi Ma
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Zhonghai Sun
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Xiaoshun He
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| | - Qi Zhou
- Department of Liver Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510080, China.,China Department of General Surgery, Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-Sen University, Huizhou, Guangdong 516081, China
| | - Baohua Hou
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, School of Medicine, South China University of Technology, Guangzhou 510080, China
| | - Linwei Wu
- Organ Transplant Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510080, China
| |
Collapse
|
28
|
Thomas R, Weihua Z. Rethink of EGFR in Cancer With Its Kinase Independent Function on Board. Front Oncol 2019; 9:800. [PMID: 31508364 PMCID: PMC6716122 DOI: 10.3389/fonc.2019.00800] [Citation(s) in RCA: 120] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 08/06/2019] [Indexed: 12/23/2022] Open
Abstract
The epidermal growth factor receptor (EGFR) is one of most potent oncogenes that are commonly altered in cancers. As a receptor tyrosine kinase, EGFR's kinase activity has been serving as the primary target for developing cancer therapeutics, namely the EGFR inhibitors including small molecules targeting its ATP binding pocket and monoclonal antibodies targeting its ligand binding domains. EGFR inhibitors have produced impressive therapeutic benefits to responsive types of cancers. However, acquired and innate resistances have precluded current anti-EGFR agents from offering sustainable benefits to initially responsive cancers and benefits to EGFR-positive cancers that are innately resistant. Recent years have witnessed a realization that EGFR possesses kinase-independent (KID) pro-survival functions in cancer cells. This new knowledge has offered a different angle of understanding of EGFR in cancer and opened a new avenue of targeting EGFR for cancer therapy. There are already many excellent reviews on the role of EGFR with a focus on its kinase-dependent functions and mechanisms of resistance to EGFR targeted therapies. The present opinion aims to initiate a fresh discussion about the function of EGFR in cancer cells by laying out some unanswered questions pertaining to EGFR in cancer cells, by rethinking the unmet therapeutic challenges from a view of EGFR's KID function, and by proposing novel approaches to target the KID functions of EGFR for cancer treatment.
Collapse
Affiliation(s)
- Rintu Thomas
- Department of Biology and Biochemistry, College of Natural Science and Mathematics, University of Houston, Houston, TX, United States
| | - Zhang Weihua
- Department of Biology and Biochemistry, College of Natural Science and Mathematics, University of Houston, Houston, TX, United States
| |
Collapse
|
29
|
Aziz K, Limzerwala JF, Sturmlechner I, Hurley E, Zhang C, Jeganathan KB, Nelson G, Bronk S, Velasco RF, van Deursen EJ, O’Brien DR, Kocher JPA, Youssef SA, van Ree JH, de Bruin A, van den Bos H, Spierings DC, Foijer F, van de Sluis B, Roberts LR, Gores G, Li H, van Deursen JM. Ccne1 Overexpression Causes Chromosome Instability in Liver Cells and Liver Tumor Development in Mice. Gastroenterology 2019; 157:210-226.e12. [PMID: 30878468 PMCID: PMC6800187 DOI: 10.1053/j.gastro.2019.03.016] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 02/15/2019] [Accepted: 03/07/2019] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS The CCNE1 locus, which encodes cyclin E1, is amplified in many types of cancer cells and is activated in hepatocellular carcinomas (HCCs) from patients infected with hepatitis B virus or adeno-associated virus type 2, due to integration of the virus nearby. We investigated cell-cycle and oncogenic effects of cyclin E1 overexpression in tissues of mice. METHODS We generated mice with doxycycline-inducible expression of Ccne1 (Ccne1T mice) and activated overexpression of cyclin E1 from age 3 weeks onward. At 14 months of age, livers were collected from mice that overexpress cyclin E1 and nontransgenic mice (controls) and analyzed for tumor burden and by histology. Mouse embryonic fibroblasts (MEFs) and hepatocytes from Ccne1T and control mice were analyzed to determine the extent to which cyclin E1 overexpression perturbs S-phase entry, DNA replication, and numbers and structures of chromosomes. Tissues from 4-month-old Ccne1T and control mice (at that age were free of tumors) were analyzed for chromosome alterations, to investigate the mechanisms by which cyclin E1 predisposes hepatocytes to transformation. RESULTS Ccne1T mice developed more hepatocellular adenomas and HCCs than control mice. Tumors developed only in livers of Ccne1T mice, despite high levels of cyclin E1 in other tissues. Ccne1T MEFs had defects that promoted chromosome missegregation and aneuploidy, including incomplete replication of DNA, centrosome amplification, and formation of nonperpendicular mitotic spindles. Whereas Ccne1T mice accumulated near-diploid aneuploid cells in multiple tissues and organs, polyploidization was observed only in hepatocytes, with losses and gains of whole chromosomes, DNA damage, and oxidative stress. CONCLUSIONS Livers, but not other tissues of mice with inducible overexpression of cyclin E1, develop tumors. More hepatocytes from the cyclin E1-overexpressing mice were polyploid than from control mice, and had losses or gains of whole chromosomes, DNA damage, and oxidative stress; all of these have been observed in human HCC cells. The increased risk of HCC in patients with hepatitis B virus or adeno-associated virus type 2 infection might involve activation of cyclin E1 and its effects on chromosomes and genomes of liver cells.
Collapse
Affiliation(s)
- Khaled Aziz
- Departments of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Jazeel F. Limzerwala
- Departments of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA
| | - Ines Sturmlechner
- Departments of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA,Department of Pediatrics, and, University Medical Center Groningen, Groningen, The Netherlands
| | - Erin Hurley
- Departments of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Cheng Zhang
- Departments of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Karthik B. Jeganathan
- Departments of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Grace Nelson
- Departments of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Steve Bronk
- Departments of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Raul Fierro Velasco
- Departments of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Erik-Jan van Deursen
- Departments of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Daniel R. O’Brien
- Departments of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA,Departments of Health Sciences Research, and, Mayo Clinic, Rochester, MN 55905, USA
| | - Jean-Pierre A. Kocher
- Departments of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, MN 55905, USA,Departments of Health Sciences Research, and, Mayo Clinic, Rochester, MN 55905, USA
| | - Sameh A. Youssef
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Janine H. van Ree
- Departments of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Alain de Bruin
- Department of Pathobiology, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands,Department of Pediatrics, and, University Medical Center Groningen, Groningen, The Netherlands
| | - Hilda van den Bos
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Diana C.J. Spierings
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Bart van de Sluis
- Department of Pediatrics, and, University Medical Center Groningen, Groningen, The Netherlands
| | - Lewis R. Roberts
- Departments of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Gregory Gores
- Departments of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Hu Li
- Departments of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Jan M. van Deursen
- Departments of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN 55905, USA,Departments of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN 55905, USA,Department of Pediatrics, and, University Medical Center Groningen, Groningen, The Netherlands,Correspondence: Please address all correspondence to Dr. Jan M. van Deursen, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, USA. Phone: 507.284.2524;
| |
Collapse
|
30
|
Li YL, Rao MJ, Zhang NY, Wu LW, Lin NM, Zhang C. BAY 87-2243 sensitizes hepatocellular carcinoma Hep3B cells to histone deacetylase inhibitors treatment via GSK-3β activation. Exp Ther Med 2019; 17:4547-4553. [PMID: 31186678 DOI: 10.3892/etm.2019.7500] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Accepted: 03/12/2019] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is associated with some of the highest cancer-associated mortality rates. Histone deacetylase (HDAC) inhibitors anti-HCC activities have been shown to promote Snail-induced metastasis. In the present study, it was shown that BAY 87-2243, a hypoxia-inducible transcription factor-1α inhibitor, could enhance the anti-HCC effects of HDAC inhibitors, including trichostatin A and vorinostat. In addition, BAY 87-2243 plus HDAC inhibitors exhibited synergistic cytotoxicity and induced significant cell death in Hep3B cells. Additionally, BAY 87-2243 combined with HDAC inhibitors-treated Hep3B cells formed fewer and smaller colonies as compared with either the control or single agent-treated cells. Furthermore, glycogen synthase kinase-3β might be involved in the enhanced cell death induced by BAY 87-2243 plus HDAC inhibitors. The present data also indicated that BAY 87-2243 combined with HDAC inhibitors could suppress the migration of Hep3B cells, and BAY 87-2243 could reverse the HDAC inhibitor-induced Snail activation in Hep3B cells. In conclusion, BAY 87-2243 combined with HDAC inhibitors might be an attractive chemotherapy strategy for HCC therapy.
Collapse
Affiliation(s)
- Yang-Ling Li
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China.,Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China
| | - Ming-Jun Rao
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China.,Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China.,Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, P.R. China
| | - Ning-Yu Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, P.R. China
| | - Lin-Wen Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Neng-Ming Lin
- Department of Clinical Pharmacology, Hangzhou First People's Hospital, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 310006, P.R. China.,Department of Clinical Pharmacology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, P.R. China.,Institute of Pharmacology, College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang 311402, P.R. China
| | - Chong Zhang
- School of Medicine, Zhejiang University City College, Hangzhou, Zhejiang 310015, P.R. China
| |
Collapse
|
31
|
Wood DJ, Endicott JA. Structural insights into the functional diversity of the CDK-cyclin family. Open Biol 2019; 8:rsob.180112. [PMID: 30185601 PMCID: PMC6170502 DOI: 10.1098/rsob.180112] [Citation(s) in RCA: 168] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 08/10/2018] [Indexed: 12/17/2022] Open
Abstract
Since their characterization as conserved modules that regulate progression through the eukaryotic cell cycle, cyclin-dependent protein kinases (CDKs) in higher eukaryotic cells are now also emerging as significant regulators of transcription, metabolism and cell differentiation. The cyclins, though originally characterized as CDK partners, also have CDK-independent roles that include the regulation of DNA damage repair and transcriptional programmes that direct cell differentiation, apoptosis and metabolic flux. This review compares the structures of the members of the CDK and cyclin families determined by X-ray crystallography, and considers what mechanistic insights they provide to guide functional studies and distinguish CDK- and cyclin-specific activities. Aberrant CDK activity is a hallmark of a number of diseases, and structural studies can provide important insights to identify novel routes to therapy.
Collapse
Affiliation(s)
- Daniel J Wood
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Medical School, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| | - Jane A Endicott
- Newcastle Cancer Centre, Northern Institute for Cancer Research, Medical School, Newcastle University, Paul O'Gorman Building, Framlington Place, Newcastle upon Tyne NE2 4HH, UK
| |
Collapse
|
32
|
Isolation and in silico prediction of potential drug-like compounds from Anethum sowa L. root extracts targeted towards cancer therapy. Comput Biol Chem 2019; 78:242-259. [DOI: 10.1016/j.compbiolchem.2018.11.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 10/22/2018] [Accepted: 11/28/2018] [Indexed: 12/16/2022]
|
33
|
Chen Y, Wang Q, Wang Q, Liu J, Jiang X, Zhang Y, Liu Y, Zhou F, Liu H. DEAD-Box Helicase 5 Interacts With Transcription Factor 12 and Promotes the Progression of Osteosarcoma by Stimulating Cell Cycle Progression. Front Pharmacol 2019; 9:1558. [PMID: 30733679 PMCID: PMC6353832 DOI: 10.3389/fphar.2018.01558] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 12/21/2018] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma (OS) is a common malignant primary bone tumor. Its mechanism of development and progression is poorly understood. Currently, there is no effective therapeutic regimens available for the treatment of OS. DEAD-box helicase 5 (DDX5) is involved in oncogenic processes. This study aimed to explore the role of DDX5 in the development and progression of OS and its relationship with transcription factor 12 (TCF12), which is as an important molecule of Wnt signaling pathway. We found that the expressions of DDX5 and TCF12 protein were significantly higher in OS patients tissues and in the MG63 cells than in the corresponding normal tissues and human osteoblast cell hFOB 1.19. Overexpressions of both DDX5 and TCF12 were associated with clinicopathological features and poor prognosis of OS patients. siRNA based knockdown of DDX5 inhibited the proliferation of MG63 cells as demonstrated by an in vitro MTS assay and 5-ethynyl-2-deoxyuridine DNA proliferation detection, and promoted apoptosis of MG63 cells measured by flow cytometry. In addition, DDX5 knockdown inhibited the MG63 cell migration and invasion on transwell assays. Further experiments showed that DDX5 knockdown not only inhibited the expression of TCF12 but also decreased the mRNA and protein levels of Cyclin E1, an important regulator of G1–S phase progression, suggesting that DDX5 was required for the entry of cells into S phase. Overexpression of TCF12 reversed the cell proliferation, migration and invasion in MG63 cells induced by DDX5 knockdown accompanied by the upregulation of Cyclin E1. Additionally, we observed that DDX5 interacted with TCF12 in both OS tissues and MG63 cells by Co-immunoprecipitation assays. Taken together, our study revealed that DDX5 interacts with TCF12 and promotes the progression of OS by stimulating cell cycle progression. Our results suggest that DDX5 and TCF12 could be potential biomarkers for the diagnosis and treatment of OS.
Collapse
Affiliation(s)
- Yanchun Chen
- Department of Histology and Embryology, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Qiaozhen Wang
- Department of Human Anatomy, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Qing Wang
- Department of Human Anatomy, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Jinmeng Liu
- Department of Histology and Embryology, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Xin Jiang
- Department of Histology and Embryology, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yawen Zhang
- Department of Histology and Embryology, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Yongxin Liu
- Department of Anesthesiology, Weifang Medical University, Weifang, China
| | - Fenghua Zhou
- Department of Pathology, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Huancai Liu
- Department of Joint Surgery, Affiliated Hospital, Weifang Medical University, Weifang, China
| |
Collapse
|
34
|
Sonntag R, Giebeler N, Nevzorova YA, Bangen JM, Fahrenkamp D, Lambertz D, Haas U, Hu W, Gassler N, Cubero FJ, Müller-Newen G, Abdallah AT, Weiskirchen R, Ticconi F, Costa IG, Barbacid M, Trautwein C, Liedtke C. Cyclin E1 and cyclin-dependent kinase 2 are critical for initiation, but not for progression of hepatocellular carcinoma. Proc Natl Acad Sci U S A 2018; 115:9282-9287. [PMID: 30150405 PMCID: PMC6140539 DOI: 10.1073/pnas.1807155115] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
E-type cyclins E1 (CcnE1) and E2 (CcnE2) are regulatory subunits of cyclin-dependent kinase 2 (Cdk2) and thought to control the transition of quiescent cells into the cell cycle. Initial findings indicated that CcnE1 and CcnE2 have largely overlapping functions for cancer development in several tumor entities including hepatocellular carcinoma (HCC). In the present study, we dissected the differential contributions of CcnE1, CcnE2, and Cdk2 for initiation and progression of HCC in mice and patients. To this end, we tested the HCC susceptibility in mice with constitutive deficiency for CcnE1 or CcnE2 as well as in mice lacking Cdk2 in hepatocytes. Genetic inactivation of CcnE1 largely prevented development of liver cancer in mice in two established HCC models, while ablation of CcnE2 had no effect on hepatocarcinogenesis. Importantly, CcnE1-driven HCC initiation was dependent on Cdk2. However, isolated primary hepatoma cells typically acquired independence on CcnE1 and Cdk2 with increasing progression in vitro, which was associated with a gene signature involving secondary induction of CcnE2 and up-regulation of cell cycle and DNA repair pathways. Importantly, a similar expression profile was also found in HCC patients with elevated CcnE2 expression and poor survival. In general, overall survival in HCC patients was synergistically affected by expression of CcnE1 and CcnE2, but not through Cdk2. Our study suggests that HCC initiation specifically depends on CcnE1 and Cdk2, while HCC progression requires expression of any E-cyclin, but no Cdk2.
Collapse
Affiliation(s)
- Roland Sonntag
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Nives Giebeler
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Yulia A Nevzorova
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Jörg-Martin Bangen
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Dirk Fahrenkamp
- Institute of Biochemistry and Molecular Biology, University Hospital RWTH, 52074 Aachen, Germany
| | - Daniela Lambertz
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Ute Haas
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Wei Hu
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Nikolaus Gassler
- Institute of Pathology, University Hospital RWTH, 52074 Aachen, Germany
| | - Francisco Javier Cubero
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Gerhard Müller-Newen
- Institute of Biochemistry and Molecular Biology, University Hospital RWTH, 52074 Aachen, Germany
| | - Ali T Abdallah
- Interdisciplinary Center for Clinical Research, University Hospital RWTH, 52074 Aachen, Germany
| | - Ralf Weiskirchen
- Institute of Molecular Pathobiochemistry, Experimental Gene Therapy and Clinical Chemistry, University Hospital RWTH, 52074 Aachen, Germany
| | - Fabio Ticconi
- Institute for Computational Genomics, RWTH Aachen University, 52074 Aachen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen University, 52074 Aachen, Germany
| | - Mariano Barbacid
- Molecular Oncology, Centro Nacional de Investigaciones Oncológicas (CNIO), 28029 Madrid, Spain
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany
| | - Christian Liedtke
- Department of Internal Medicine III, University Hospital Rheinisch-Westfälische Technische Hochschule (RWTH), 52074 Aachen, Germany;
| |
Collapse
|