1
|
Huang H, Zeng J, Yu X, Du H, Wen C, Mao Y, Tang H, Kuang X, Liu W, Yu H, Liu H, Li B, Long C, Yan J, Shen H. Establishing chronic models of age-related macular degeneration via long-term iron ion overload. Am J Physiol Cell Physiol 2024; 326:C1367-C1383. [PMID: 38406826 DOI: 10.1152/ajpcell.00532.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 01/26/2024] [Accepted: 02/14/2024] [Indexed: 02/27/2024]
Abstract
Age-related macular degeneration (AMD) is characterized by the degenerative senescence in the retinal pigment epithelium (RPE) and photoreceptors, which is accompanied by the accumulation of iron ions in the aging retina. However, current models of acute oxidative stress are still insufficient to simulate the gradual progression of AMD. To address this, we established chronic injury models by exposing the aRPE-19 cells, 661W cells, and mouse retina to iron ion overload over time. Investigations at the levels of cell biology and molecular biology were performed. It was demonstrated that long-term treatment of excessive iron ions induced senescence-like morphological changes, decreased cell proliferation, and impaired mitochondrial function, contributing to apoptosis. Activation of the mitogen-activated protein kinase (MAPK) pathway and the downstream molecules were confirmed both in the aRPE-19 and 661W cells. Furthermore, iron ion overload resulted in dry AMD-like lesions and decreased visual function in the mouse retina. These findings suggest that chronic exposure to overloading iron ions plays a significant role in the pathogenesis of retinopathy and provide a potential model for future studies on AMD.NEW & NOTEWORTHY To explore the possibility of constructing reliable research carriers on age-related macular degeneration (AMD), iron ion overload was applied to establish models in vitro and in vivo. Subsequent investigations into cellular physiology and molecular biology confirmed the presence of senescence in these models. Through this study, we hope to provide a better option of feasible methods for future researches into AMD.
Collapse
Affiliation(s)
- Hao Huang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Department of Ophthalmology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, People's Republic of China
| | - Jingshu Zeng
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xinyue Yu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Han Du
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Chaojuan Wen
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Yan Mao
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Han Tang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Xielan Kuang
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Wei Liu
- Department of Ophthalmology, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, People's Republic of China
| | - Huan Yu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Huijun Liu
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Eye Fundus Department, Affiliated Aier Eye Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Bowen Li
- Eye Center of Xiangya Hospital, Central South University, Changsha, People's Republic of China
| | - Chongde Long
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Jianhua Yan
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| | - Huangxuan Shen
- State Key Laboratory of Ophthalmology, Guangdong Provincial Key Laboratory of Ophthalmology and Visual Science, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
- Biobank of Eye, State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, People's Republic of China
| |
Collapse
|
2
|
Federman N, Gordon EM, Chawla SP, Hall FL. Editorial: Celebrating the 200th mendel's anniversary: gene-targeted diagnostics and therapies for cancer. FRONTIERS IN MOLECULAR MEDICINE 2024; 4:1366963. [PMID: 39086436 PMCID: PMC11285536 DOI: 10.3389/fmmed.2024.1366963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 01/15/2024] [Indexed: 08/02/2024]
Affiliation(s)
- Noah Federman
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Erlinda M. Gordon
- Cancer Center of Southern California, Santa Monica, CA, United States
- Aveni Foundation, Santa Monica, CA, United States
| | - Sant P. Chawla
- Cancer Center of Southern California, Santa Monica, CA, United States
- Counterpoint Biomedica LLC, Santa Monica, CA, United States
| | - Frederick L. Hall
- Counterpoint Biomedica LLC, Santa Monica, CA, United States
- Delta Next-Gene, LLC, Santa Monica, CA, United States
| |
Collapse
|
3
|
Fujiwara M, Ferdousi F, Isoda H. Investigation into Molecular Brain Aging in Senescence-Accelerated Mouse (SAM) Model Employing Whole Transcriptomic Analysis in Search of Potential Molecular Targets for Therapeutic Interventions. Int J Mol Sci 2023; 24:13867. [PMID: 37762170 PMCID: PMC10530366 DOI: 10.3390/ijms241813867] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/06/2023] [Accepted: 09/07/2023] [Indexed: 09/29/2023] Open
Abstract
With the progression of an aging society, cognitive aging has emerged as a pressing concern necessitating attention. The senescence-accelerated mouse-prone 8 (SAMP8) model has proven instrumental in investigating the early stages of cognitive aging. Through an extensive examination of molecular changes in the brain cortex, utilizing integrated whole-genome transcriptomics, our principal aim was to uncover potential molecular targets with therapeutic applications and relevance to drug screening. Our investigation encompassed four distinct conditions, comparing the same strain at different time points (1 year vs. 16 weeks) and the same time point across different strains (SAMP8 vs. SAMR1), namely: physiological aging, accelerated aging, early events in accelerated aging, and late events in accelerated aging. Focusing on key functional alterations associated with aging in the brain, including neurogenesis, synapse dynamics, neurometabolism, and neuroinflammation, we identified candidate genes linked to these processes. Furthermore, employing protein-protein interaction (PPI) analysis, we identified pivotal hub genes involved in interactions within these functional domains. Additionally, gene-set perturbation analysis allowed us to uncover potential upstream genes or transcription factors that exhibited activation or inhibition across the four conditions. In summary, our comprehensive analysis of the SAMP8 mouse brain through whole-genome transcriptomics not only deepens our understanding of age-related changes but also lays the groundwork for a predictive model to facilitate drug screening for cognitive aging.
Collapse
Affiliation(s)
- Michitaka Fujiwara
- Graduate School of Environmental Science Program, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8577, Japan
- Open Innovation Laboratory for Food and Medicinal Resource Engineering, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| | - Farhana Ferdousi
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| | - Hiroko Isoda
- Open Innovation Laboratory for Food and Medicinal Resource Engineering, National Institute of Advanced Industrial Science and Technology (AIST), 1-1-1 Tennodai, Tsukuba 305-8572, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
- Alliance for Research on the Mediterranean and North Africa (ARENA), University of Tsukuba, 1-1-1 Tennodai, Tsukuba 305-8572, Japan
| |
Collapse
|
4
|
Nong JS, Zhou X, Liu JQ, Luo JZ, Huang JM, Xie HX, Yang KJ, Wang J, Ye XP, Peng T. Nucleoporin 107 is a prognostic biomarker in hepatocellular carcinoma associated with immune infiltration. Cancer Med 2023; 12:10990-11009. [PMID: 36952458 DOI: 10.1002/cam4.5807] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/25/2023] Open
Abstract
OBJECTIVE To assess the diagnostic value and clinical significance of nucleoporin 107 (NUP107) in hepatocellular carcinoma (HCC), and explore the possible mechanisms. METHODS The transcriptomic and clinical data of HCC patients were retrieved from The Cancer Genome Atlas (TCGA) and GEO databases. Tissue specimens were collected from HCC patients in the Guangxi area. According to the expression levels and prognostic characteristics of NUP107, ROC curves and nomogram models were constructed using the R package. RESULTS NUP107 was highly expressed in 26 human cancers including HCC, and was associated with advanced HCC staging and worse prognosis. NUP107 showed satisfactory ability to predict the prognosis of HCC patients (AUC >0.8). Results of gene set enrichment analysis (GSEA) further showed that NUP107 was mainly associated with cell cycle-related pathways such as the cell cycle, DNA replication, G2M checkpoint, E2F target, and mitotic spindle. In addition, NUP107 was also associated with immune infiltration in HCC and showed significant positive correlation with immune checkpoints (PD-L1 and TIM-3).
Collapse
Affiliation(s)
- Ju-Sen Nong
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Xin Zhou
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Jun-Qi Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Jian-Zhu Luo
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Jia-Mi Huang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Hai-Xiang Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Ke-Jian Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Jing Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Xin-Ping Ye
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, People's Republic of China
- Key Laboratory of Early Prevention & Treatment for Regional High Frequency Tumor, Ministry of Education, Guangxi Medical University, Nanning, People's Republic of China
| |
Collapse
|
5
|
Gordon EM, Hall FL. The advent of a pan-collagenous CLOVIS POINT for pathotropic targeting and cancer gene therapy, a retrospective. FRONTIERS IN MOLECULAR MEDICINE 2023; 3:1125928. [PMID: 39086682 PMCID: PMC11285703 DOI: 10.3389/fmmed.2023.1125928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 02/13/2023] [Indexed: 08/02/2024]
Abstract
The 'Clovis Point'-an enabling prehistoric gain-of-function in stone-age tool technologies which empowered the Paleoindian-Americans to hunt, to strike-deep, and to kill designated target megafauna more efficiently-was created biochemically by molecular-genetic bio-engineering. This Biomedical "Clovis Point" was crafted by adapting a broad-spectrum Pan-Collagen Binding Domain (Pan-Coll/CBD) found within the immature pre-pro-peptide segment of Von Willebrand Factor into a constructive series of advanced medical applications. Developed experimentally, preclinically, and clinically into a cutting-edge Biotechnology Platform, the Clovis Point is suitable for 1) solid-state binding of growth factors on collagenous scaffolds for improved orthopedic wound healing, 2) promoting regeneration of injured/diseased tissues; and 3) autologous stem cell capture, expansion, and gene-based therapies. Subsequent adaptations of the high-affinity Pan-Coll/CBD (exposed-collagen-seeking/surveillance function) for intravenous administration in humans, enabled the physiological delivery, aka Pathotropic Targeting to diseased tissues via the modified envelopes of gene vectors; enabling 4) precision tumor-targeting for cancer gene therapy and 5) adoptive/localized immunotherapies, demonstrating improved long-term survival value-thus pioneering a proximal and accessible cell cycle control point for cancer management-empowering modern medical oncologists to address persistent problems of chemotherapy resistance, recurrence, and occult progression of metastatic disease. Recent engineering adaptations have advanced the clinical utility to include the targeted delivery of small molecule APIs: including taxanes, mAbs, and RNA-based therapeutics.
Collapse
Affiliation(s)
- Erlinda M. Gordon
- Counterpoint Biomedica LLC, Santa Monica, CA, United States
- Delta Next-Gene, LLC, Santa Monica, CA, United States
| | - Frederick L. Hall
- Counterpoint Biomedica LLC, Santa Monica, CA, United States
- Delta Next-Gene, LLC, Santa Monica, CA, United States
| |
Collapse
|
6
|
Lin Y, Zhang Z, Li Y, Chen Y, Su M, Zhao W. LncRNA DIRC1 is a novel prognostic biomarker and correlated with immune infiltrates in stomach adenocarcinoma. Medicine (Baltimore) 2022; 101:e31839. [PMID: 36401393 PMCID: PMC9678523 DOI: 10.1097/md.0000000000031839] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
The potential application value of Long non-coding RNA disrupted in renal carcinoma 1 (DIRC1) has not yet been explored, the purpose of this study was to explore the relationship between DIRC1 and stomach adenocarcinoma (STAD) based on the cancer genome atlas database. Wilcoxon rank sum test, Chi-square test, Fisher test and logistic regression were used to evaluate relationships between clinical-pathologic features and DIRC1 expression. Receiver operating characteristic (ROC) curves were used to describe binary classifier value of DIRC1 using area under curve (AUC) score. Kaplan-Meier method was used to assess the impact of DIRC1 on prognosis and the impact of DIRC1-related hub genes on prognosis. Gene oncology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used to predict the function of differentially expressed genes associated with DIRC1. Gene set enrichment analysis (GSEA) was used to predict biological states or processes associated with DIRC1. Immune infiltration analysis was performed to identify the significantly involved functions of DIRC1. Protein-protein interaction (PPI) networks were established and 10 hub genes identified with Cytoscape software. Real time-polymerase chain reaction (RT-PCR) was used to detect the expression of DIRC1 in Gastric Cancer patients and healthy people. Increased DIRC1 expression in STAD was associated with T stage (P = .004), race (P = .045), histologic grade (P = .029) and anatomic neoplasm subdivision (P = .034). ROC curve suggested the significant diagnostic ability of DIRC1 (AUC = 0.779). High DIRC1 expression predicted a poorer Overall survival (P = .004, hazard ratio: 1.63; 95% confidence interval: 1.17-2.27; P = .034). GO and KEGG analysis demonstrated that DIRC1 is related to epidermis, collagen-containing extracellular matrix, receptor-ligand activity, protein digestion and absorption, etc. GSEA demonstrated that E2F target, G2M checkpoint, Myc target, interferon γ reaction were differentially enriched in the high DIRC1 expression phenotype. SsGSEA and Spearman correlation revealed the relationships between DIRC1 and macrophages, dendritic cells, and Th1 cells were the strongest. Coregulatory proteins were included in the PPI network, higher expressions of 4 hub genes were associated with worse prognosis in STAD. RT-PCR showed that the expression of DIRC1 in the serum of Gastric Cancer patients was higher than healthy people (P = .027). DIRC1 expression was significantly correlated with poor survival and immune infiltrations in STAD, and it may be a promising prognostic biomarker in STAD.
Collapse
Affiliation(s)
- Yuning Lin
- Xiamen Key Laboratory of Biomarker Translational Medicine, Medical Laboratory of Xiamen Humanity Hospital Fujian Medical University, Xiamen, China
| | - Zhongying Zhang
- Xiamen Key Laboratory of Biomarker Translational Medicine, Medical Laboratory of Xiamen Humanity Hospital Fujian Medical University, Xiamen, China
- * Correspondence: Zhongying Zhang, Xiamen Key Laboratory of Biomarker Translational Medicine, Medical Laboratory of Xiamen Humanity Hospital Fujian Medical University, No.3777 Xianyue Road, Xiamen, Fujian Province 361009, China (e-mail: )
| | - Ying Li
- Ultrasonography Department, Women and Children’s Hospital, School of Medicine, Xiamen University, Xiamen, China
| | - Yongquan Chen
- Xiamen Key Laboratory of Biomarker Translational Medicine, Medical Laboratory of Xiamen Humanity Hospital Fujian Medical University, Xiamen, China
| | - Meiying Su
- Xiamen Key Laboratory of Biomarker Translational Medicine, Medical Laboratory of Xiamen Humanity Hospital Fujian Medical University, Xiamen, China
| | - Wenzhen Zhao
- Xiamen Key Laboratory of Biomarker Translational Medicine, Medical Laboratory of Xiamen Humanity Hospital Fujian Medical University, Xiamen, China
| |
Collapse
|
7
|
Liu J, Jia J, Wang S, Zhang J, Xian S, Zheng Z, Deng L, Feng Y, Zhang Y, Zhang J. Prognostic Ability of Enhancer RNAs in Metastasis of Non-Small Cell Lung Cancer. Molecules 2022; 27:molecules27134108. [PMID: 35807355 PMCID: PMC9268450 DOI: 10.3390/molecules27134108] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Revised: 06/15/2022] [Accepted: 06/23/2022] [Indexed: 02/01/2023] Open
Abstract
(1) Background: Non-small cell lung cancer (NSCLC) is the most common lung cancer. Enhancer RNA (eRNA) has potential utility in the diagnosis, prognosis and treatment of cancer, but the role of eRNAs in NSCLC metastasis is not clear; (2) Methods: Differentially expressed transcription factors (DETFs), enhancer RNAs (DEEs), and target genes (DETGs) between primary NSCLC and metastatic NSCLC were identified. Prognostic DEEs (PDEEs) were screened by Cox regression analyses and a predicting model for metastatic NSCLC was constructed. We identified DEE interactions with DETFs, DETGs, reverse phase protein arrays (RPPA) protein chips, immunocytes, and pathways to construct a regulation network using Pearson correlation. Finally, the mechanisms and clinical significance were explained using multi-dimensional validation unambiguously; (3) Results: A total of 255 DEEs were identified, and 24 PDEEs were selected into the multivariate Cox regression model (AUC = 0.699). Additionally, the NSCLC metastasis-specific regulation network was constructed, and six key PDEEs were defined (ANXA8L1, CASTOR2, CYP4B1, GTF2H2C, PSMF1 and TNS4); (4) Conclusions: This study focused on the exploration of the prognostic value of eRNAs in the metastasis of NSCLC. Finally, six eRNAs were identified as potential markers for the prediction of metastasis of NSCLC.
Collapse
Affiliation(s)
- Jun Liu
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (J.L.); (J.J.)
- School of Medicine, Tongji University, Shanghai 200092, China; (S.W.); (J.Z.); (S.X.); (Z.Z.)
| | - Jingyi Jia
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (J.L.); (J.J.)
- School of Medicine, Tongji University, Shanghai 200092, China; (S.W.); (J.Z.); (S.X.); (Z.Z.)
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Clinical Research Center for Infectious Diseases (Tuberculosis), Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Siqiao Wang
- School of Medicine, Tongji University, Shanghai 200092, China; (S.W.); (J.Z.); (S.X.); (Z.Z.)
| | - Junfang Zhang
- School of Medicine, Tongji University, Shanghai 200092, China; (S.W.); (J.Z.); (S.X.); (Z.Z.)
| | - Shuyuan Xian
- School of Medicine, Tongji University, Shanghai 200092, China; (S.W.); (J.Z.); (S.X.); (Z.Z.)
| | - Zixuan Zheng
- School of Medicine, Tongji University, Shanghai 200092, China; (S.W.); (J.Z.); (S.X.); (Z.Z.)
| | - Lin Deng
- Normal College, Qingdao University, Qingdao 266071, China;
| | - Yonghong Feng
- School of Medicine, Tongji University, Shanghai 200092, China; (S.W.); (J.Z.); (S.X.); (Z.Z.)
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Shanghai Clinical Research Center for Infectious Diseases (Tuberculosis), Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Correspondence: (Y.F.); (Y.Z.); (J.Z.)
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
- Correspondence: (Y.F.); (Y.Z.); (J.Z.)
| | - Jie Zhang
- Department of Anesthesiology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China; (J.L.); (J.J.)
- School of Medicine, Tongji University, Shanghai 200092, China; (S.W.); (J.Z.); (S.X.); (Z.Z.)
- Correspondence: (Y.F.); (Y.Z.); (J.Z.)
| |
Collapse
|
8
|
Adler SO, Spiesser TW, Uschner F, Münzner U, Hahn J, Krantz M, Klipp E. A yeast cell cycle model integrating stress, signaling, and physiology. FEMS Yeast Res 2022; 22:6592118. [PMID: 35617157 PMCID: PMC9246278 DOI: 10.1093/femsyr/foac026] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/22/2022] [Accepted: 05/23/2022] [Indexed: 11/25/2022] Open
Abstract
The cell division cycle in eukaryotic cells is a series of highly coordinated molecular interactions that ensure that cell growth, duplication of genetic material, and actual cell division are precisely orchestrated to give rise to two viable progeny cells. Moreover, the cell cycle machinery is responsible for incorporating information about external cues or internal processes that the cell must keep track of to ensure a coordinated, timely progression of all related processes. This is most pronounced in multicellular organisms, but also a cardinal feature in model organisms such as baker's yeast. The complex and integrative behavior is difficult to grasp and requires mathematical modeling to fully understand the quantitative interplay of the single components within the entire system. Here, we present a self-oscillating mathematical model of the yeast cell cycle that comprises all major cyclins and their main regulators. Furthermore, it accounts for the regulation of the cell cycle machinery by a series of external stimuli such as mating pheromones and changes in osmotic pressure or nutrient quality. We demonstrate how the external perturbations modify the dynamics of cell cycle components and how the cell cycle resumes after adaptation to or relief from stress.
Collapse
Affiliation(s)
- Stephan O Adler
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Thomas W Spiesser
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Friedemann Uschner
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany.,Institute for Medical Informatics and Biometry, Technische Universität Dresden, Fetscherstr. 74, 01307 Dresden, Sachsen, Germany
| | - Ulrike Münzner
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany.,Laboratory of Cell Systems, Institute for Protein Research, Osaka University, 3-2 Yamadaoka, 565-0871, Suita, Osaka, Japan
| | - Jens Hahn
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Marcus Krantz
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| | - Edda Klipp
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Invalidenstr. 42, 10115 Berlin, Germany
| |
Collapse
|
9
|
Lázari LC, Wolf IR, Schnepper AP, Valente GT. LncRNAs of Saccharomyces cerevisiae bypass the cell cycle arrest imposed by ethanol stress. PLoS Comput Biol 2022; 18:e1010081. [PMID: 35587936 PMCID: PMC9232138 DOI: 10.1371/journal.pcbi.1010081] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 06/24/2022] [Accepted: 04/05/2022] [Indexed: 11/19/2022] Open
Abstract
Ethanol alters many subsystems of Saccharomyces cerevisiae, including the cell cycle. Two ethanol-responsive lncRNAs in yeast interact with cell cycle proteins, and here, we investigated the role of these RNAs in cell cycle. Our network dynamic modeling showed that higher and lower ethanol-tolerant strains undergo cell cycle arrest in mitosis and G1 phases, respectively, during ethanol stress. The higher population rebound of the lower ethanol-tolerant phenotype after stress relief responds to the late phase arrest. We found that the lncRNA lnc9136 of SEY6210 (a lower ethanol-tolerant strain) induces cells to skip mitosis arrest. Simulating an overexpression of lnc9136 and analyzing CRISPR–Cas9 mutants lacking this lncRNA suggest that lnc9136 induces a regular cell cycle even under ethanol stress, indirectly regulating Swe1p and Clb1/2 by binding to Gin4p and Hsl1p. Notably, lnc10883 of BY4742 (a higher ethanol-tolerant strain) does not prevent G1 arrest in this strain under ethanol stress. However, lnc19883 circumvents DNA and spindle damage checkpoints, maintaining a functional cell cycle by interacting with Mec1p or Bub1p even in the presence of DNA/spindle damage. Overall, we present the first evidence of direct roles for lncRNAs in regulating yeast cell cycle proteins, the dynamics of this system in different ethanol-tolerant phenotypes, and a new yeast cell cycle model. Ethanol is a cell stressor in yeast that dampen ethanol production. LncRNAs are RNAs that control many cellular processes. Computational simulations allow us to study the dynamism of cell systems. Therefore, we built a computational model of the yeast cell cycle to investigate how cells respond to ethanol stress. Simulations showed that ethanol stress or spindle damage arrests the cell cycle. Furthermore, the performance of higher and lower ethanol-tolerant strains in poststress recovery growth seems to be related to the cell cycle phase in which cells are stalled. However, two lncRNAs maintain the activity of the cell cycle even in yeast cells under these stresses by repressing specific cell cycle proteins. Finally, this model facilitates analyses of the yeast cell cycle for applied or basic science purposes.
Collapse
Affiliation(s)
- Lucas Cardoso Lázari
- Department of Parasitology, Institute of Biomedical Sciences, Sāo Paulo University (USP), Sao Paulo, Brazil
- Department of Bioprocess and Biotechnology, School of Agriculture, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Ivan Rodrigo Wolf
- Department of Bioprocess and Biotechnology, School of Agriculture, Sao Paulo State University (UNESP), Botucatu, Brazil
- Department of Structural and Functional Biology, Institute of Bioscience at Botucatu, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Amanda Piveta Schnepper
- Department of Bioprocess and Biotechnology, School of Agriculture, Sao Paulo State University (UNESP), Botucatu, Brazil
| | - Guilherme Targino Valente
- Department of Bioprocess and Biotechnology, School of Agriculture, Sao Paulo State University (UNESP), Botucatu, Brazil
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
- * E-mail: ,
| |
Collapse
|
10
|
Chen Y, Chen C, Song D, Liu T, Cheng O. Dexmedetomidine protects SH-SY5Y cells against MPP + -induced declining of mitochondrial membrane potential and cell cycle deficits. Eur J Neurosci 2021; 54:4141-4153. [PMID: 33905578 DOI: 10.1111/ejn.15252] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 11/29/2022]
Abstract
Dexmedetomidine (Dex), an adrenergic α2 receptor agonist, is commonly used in deep-brain stimulation surgery for Parkinson's disease (PD). However, there is evidence that the use of anaesthetics may accelerate the progression of neurodegenerative diseases. The effect of Dex on PD remains unclear. Here, we cultured the all-trans-retinoicacid (ATRA) differentiated SH-SY5Y cells in vitro and then treated with MPP+ (1.5mM) with or without Dex (10nM) or Dex combined with Atipamezole (Ati,100nM, adrenergic α2 receptor inhibitor). The ratio of apoptotic cells, mitochondrial membrane potential (Δψm), reactive oxygen species (ROS), cell cycle and apoptotic markers (Cleaved caspase-3, 9) were analysed by flow cytometry and immunofluorescence. We found that the levels of apoptotic ratio and cleaved caspase-3, 9 increased, ROS accumulated, and mitochondrial membrane potential decreased after MPP+treatment, while these changes were partially reversed by Dex. Dex also prevented MPP+ induced cell arrest by increasing G1 phase cells, decreasing S phase cells, and decreasing the expression of cyclinD1 and Cdk4. Moreover the effects of Dex were partially reversed by Ati. These findings reveal that Dex attenuated MPP+ -induced apoptosis of SH-SY5Y cells by preventing the loss of Δψm, reducing ROS, and regulating the cell cycle. Our findings indicated that Dex is more likely to be a potential drug for the treatment of PD.
Collapse
Affiliation(s)
- Yaohua Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Cheng Chen
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Song
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tingting Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Laboratory Research Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Oumei Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
11
|
Joshi SN, Murphy EA, Olaniyi P, Bryant RJ. The multiple effects of aspirin in prostate cancer patients. Cancer Treat Res Commun 2020; 26:100267. [PMID: 33360326 DOI: 10.1016/j.ctarc.2020.100267] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/02/2020] [Accepted: 12/07/2020] [Indexed: 01/31/2023]
Abstract
Aspirin is a commonly used medication with anti-inflammatory and analgesic properties, and it is widely used to reduce the risk of ischaemic heart disease-related events and/or cerebrovascular accidents. However, there is also evidence from epidemiological and interventional studies to suggest that regular aspirin use can reduce the risk of prostate cancer development and progression, and can reduce the risk of disease recurrence following anti-prostate cancer therapy. Aspirin use in African-American men is associated with a reduced incidence of advanced PCa and reduced disease recurrence, and there is evidence from other studies of an association between regular aspirin use and decreased PCa-related mortality. The cyclooxygenase-2 enzyme inhibited by Aspirin and other NSAIDs, and which catalyses prostaglandin synthesis and mediates inflammation, is overexpressed in prostate cancer, therefore inhibition of cyclooxygenase-2 may have direct, and indirect, therapeutic effects. This review explores the evidence suggesting that aspirin use can modify prostate cancer biology and disease characteristics, and explores the potential mechanisms underpinning the observed associations between aspirin use and modification of prostate cancer risk. It also summarises the potential for adjuvant aspirin use to combine with other therapeutic approaches such as radical surgery and radiotherapy.
Collapse
Affiliation(s)
- S N Joshi
- Medical Sciences Divisional Office, University of Oxford, Level 3, John Radcliffe Hospital, Oxford OX3 9DU, United Kingdom
| | - E A Murphy
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford OX3 7DQ, United Kingdom
| | - P Olaniyi
- Department of Urology, Ipswich Hospital, East Suffolk and North Essex NHS Foundation Trust, Heath Road, Ipswich IP4 5PD, United Kingdom
| | - R J Bryant
- Department of Urology, Ipswich Hospital, East Suffolk and North Essex NHS Foundation Trust, Heath Road, Ipswich IP4 5PD, United Kingdom; Department of Urology, Churchill Hospital, Oxford University Hospitals NHS Foundation Trust, Oxford OX3 7LE, United Kingdom.
| |
Collapse
|
12
|
Investigation of the Possible Role of RAD9 in Post-Diapaused Embryonic Development of the Brine Shrimp Artemia sinica. Genes (Basel) 2019; 10:genes10100768. [PMID: 31574972 PMCID: PMC6826366 DOI: 10.3390/genes10100768] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 09/25/2019] [Accepted: 09/26/2019] [Indexed: 11/17/2022] Open
Abstract
Background: The cell cycle checkpoint protein RAD9 is a vital cell cycle regulator in eukaryotic cells. RAD9 is involved in diverse cellular functions by oligomer or monomer. However, the specific mechanism of its activity remains unknown in crustaceans, especially in embryonic diapause resumption of the brine shrimp Artemia sinica. Methods and Results: In the present article, a 1238 bp full-length cDNA of As–RAD9 gene, encoding 376 amino acids, was obtained from A. sinica. The expression pattern of As–RAD9 was analyzed by qPCR and Western blot. The mRNA expression level climbs to the top at the 10 h stage of embryo development, while the protein expression pattern is generally consistent with qPCR results. Moreover, the As–RADd9 related signaling proteins, As–RAD1, As–HUS1, As–RAD17, and As–CHK1, were also detected. Immunofluorescence assay showed that the location of As–RAD9 did not show tissue or organ specificity, and the intracellular expression was concentrated in the cytoplasm more than in the nucleus. We also explored the amount of As–RAD9 under the stresses of cold and high salinity, and the results indicate that As–RAD9 is a stress-related factor, though the mechanisms may be different in response to different stresses. Knocking down of the As–RAD9 gene led to embryonic development delay in A. sinica. Conclusions: All these results reveal that As–RAD9 is necessary for post-diapaused embryonic development in A. sinica.
Collapse
|
13
|
Fu Y, Niu Y, Pan B, Liu Y, Zhang B, Li X, Yang A, Nie J, Wang R, Yang J. OGG1 methylation mediated the effects of cell cycle and oxidative DNA damage related to PAHs exposure in Chinese coke oven workers. CHEMOSPHERE 2019; 224:48-57. [PMID: 30807913 DOI: 10.1016/j.chemosphere.2019.02.114] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2018] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 06/09/2023]
Abstract
Previous publications have indicated that polycyclic aromatic hydrocarbons (PAHs) exposures are associated with increased DNA damage and abnormal cell cycle arrest; however, the details of mechanisms remain largely unknown. This study aimed to quantify the associations of 8-oxoguanine DNA glycosylase (OGG1) methylation with urinary PAHs metabolites, DNA damage and cell cycle arrest, and further to assess the role of OGG1 methylation in mediating the association of urinary PAHs metabolites with DNA damage and cell cycle arrest. Urinary biomarkers of PAHs exposure and a marker of oxidative DNA damage (8-hydroxy-2'-deoxyguanosin, 8-OHdG) were measured by high performance liquid chromatography. Cell cycle of lymphocyte was analysed with flow cytometry and OGG1 methylation in venous blood was measured by pyrosequencing. After adjusting for covariates, urinary 1-OHP levels were positively associated with lymphocyte S phase arrest and oxidative DNA damage, while were negatively associated with G0/G1 phase arrest. OGG1 methylation was not only positively correlated with urinary 1-OHP in a dose-responsive manner (P trend = 0.008) but was also associated with G0/G1 phase arrest (OR: 0.63, 95% CI: 0.41-0.97), S phase arrest (OR: 1.55, 95% CI: 1.01-2.40) and oxidative DNA damage (OR: 1.71, 95% CI: 1.02-2.86). Mediation analysis estimated that OGG1 methylation mediated about 20% of associations between urinary 1-OHP levels and cell cycle arrest and oxidative DNA damage, respectively (all P < 0.05). Our findings suggested that urinary 1-OHP concentrations were associated with cell cycle arrest and oxidative DNA damage by a mechanism partly involving OGG1 methylation.
Collapse
Affiliation(s)
- Ye Fu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yingying Niu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Baolong Pan
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Yanli Liu
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Bin Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Xuejing Li
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Aimin Yang
- School of Public Health, The University of Hong Kong, Hong Kong SAR, China
| | - Jisheng Nie
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Ruisheng Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China
| | - Jin Yang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, Taiyuan 030001, Shanxi, China.
| |
Collapse
|
14
|
Labrie M, Kim TB, Ju Z, Lee S, Zhao W, Fang Y, Lu Y, Chen K, Ramirez P, Frumovitz M, Meyer L, Fleming ND, Sood AK, Coleman RL, Mills GB, Westin SN. Adaptive responses in a PARP inhibitor window of opportunity trial illustrate limited functional interlesional heterogeneity and potential combination therapy options. Oncotarget 2019; 10:3533-3546. [PMID: 31191824 PMCID: PMC6544405 DOI: 10.18632/oncotarget.26947] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022] Open
Abstract
Poly (ADP-ribose) polymerase inhibitor (PARPi)-based combination therapies are demonstrating efficacy in patients, however, identifying the right combination for the right patient remains a critical challenge. Thus, it is urgent to develop approaches able to identify patients likely to benefit from specific combination therapies. Several groups, including ours, have demonstrated that targeting adaptive responses induced by PARPi increases depth and duration of response. In this study, we instituted a talazoparib (PARPi) monotherapy window of opportunity trial to identify informative adaptive responses in high grade serous ovarian cancer patients (HGSOC). Patients were treated for 7 to 14 days with PARPi monotherapy prior to surgery with tissue samples from multiple sites being collected pre- and post-treatment in each patient. Analysis of these samples demonstrated that individual patients displayed different adaptive responses with limited interlesional heterogeneity. Ability of combination therapies designed to interdict adaptive responses to decrease viability was validated using model systems. Thus, assessment of adaptive responses to PARPi provides an opportunity for patient-specific selection of combination therapies designed to interdict patient-specific adaptive responses to maximize patient benefit.
Collapse
Affiliation(s)
- Marilyne Labrie
- Knight Cancer Institute and Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Tae-Beom Kim
- Department of Bioinformatics and Computational Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Zhenlin Ju
- Department of Bioinformatics and Computational Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Sanghoon Lee
- Department of Systems Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Zhao
- Department of Systems Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Yong Fang
- Knight Cancer Institute and Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA
| | - Yiling Lu
- Department of Systems Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Pedro Ramirez
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Frumovitz
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Larissa Meyer
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Nicole D Fleming
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Anil K Sood
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Robert L Coleman
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Gordon B Mills
- Knight Cancer Institute and Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, OR, USA.,Department of Systems Biology, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Shannon N Westin
- Department of Gynecologic Oncology and Reproductive Medicine, University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
15
|
Nam HK, Vesela I, Siismets E, Hatch NE. Tissue nonspecific alkaline phosphatase promotes calvarial progenitor cell cycle progression and cytokinesis via Erk1,2. Bone 2019; 120:125-136. [PMID: 30342227 PMCID: PMC6360114 DOI: 10.1016/j.bone.2018.10.013] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/09/2018] [Accepted: 10/14/2018] [Indexed: 01/09/2023]
Abstract
Bone growth is dependent upon the presence of self-renewing progenitor cell populations. While the contribution of Tissue Nonspecific Alkaline Phosphatase (TNAP) enzyme activity in promoting bone mineralization when expressed in differentiated bone forming cells is well understood, little is known regarding the role of TNAP in bone progenitor cells. We previously found diminished proliferation in the calvarial MC3T3E1 cell line upon suppression of TNAP by shRNA, and in calvarial cells and tissues of TNAP-/- mice. These findings indicate that TNAP promotes cell proliferation. Here we investigate how TNAP mediates this effect. Results show that TNAP is essential for calvarial progenitor cell cycle progression and cytokinesis, and that these effects are mediated by inorganic phosphate and Erk1/2. Levels of active Erk1/2 are significantly diminished in TNAP deficient cranial cells and tissues even in the presence of inorganic phosphate. Moreover, in the absence of TNAP, FGFR2 expression levels are high and FGF2 rescues phospho-Erk1/2 levels and cell cycle abnormalities to a significantly greater extent than inorganic phosphate. Based upon the data we propose a model in which TNAP stimulates Erk1/2 activity via both phosphate dependent and independent mechanisms to promote cell cycle progression and cytokinesis in calvarial bone progenitor cells. Concomitantly, TNAP feeds back to inhibit FGFR2 expression. These results identify a novel mechanism by which TNAP promotes calvarial progenitor cell renewal and indicate that converging pathways exist downstream of FGF signaling and TNAP activity to control craniofacial skeletal development.
Collapse
Affiliation(s)
- Hwa Kyung Nam
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, 1011 N University Avenue, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | - Iva Vesela
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, 1011 N University Avenue, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | - Erica Siismets
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, 1011 N University Avenue, University of Michigan, Ann Arbor, MI 48109-1078, USA
| | - Nan E Hatch
- Department of Orthodontics and Pediatric Dentistry, School of Dentistry, 1011 N University Avenue, University of Michigan, Ann Arbor, MI 48109-1078, USA.
| |
Collapse
|
16
|
Zhong Y, Jiang L, Lin H, Li X, Long X, Zhou Y, Li B, Li Z. Overexpression of KIF18A promotes cell proliferation, inhibits apoptosis, and independently predicts unfavorable prognosis in lung adenocarcinoma. IUBMB Life 2019; 71:942-955. [PMID: 30817091 DOI: 10.1002/iub.2030] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/07/2019] [Accepted: 02/09/2019] [Indexed: 01/29/2023]
Affiliation(s)
- Yonglong Zhong
- Medical College, Guangxi University; Nanning Guangxi Zhuang Autonomous Region China
- Department of Thoracic Cardiovascular Surgery; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning Guangxi Zhuang Autonomous Region China
| | - Lingyu Jiang
- Intensive Care Unit; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning China
| | - Hui Lin
- Medical College, Guangxi University; Nanning Guangxi Zhuang Autonomous Region China
- Department of Thoracic Cardiovascular Surgery; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning Guangxi Zhuang Autonomous Region China
| | - Xiangwei Li
- Department of Thoracic Cardiovascular Surgery; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning Guangxi Zhuang Autonomous Region China
| | - Xiaomao Long
- Department of Thoracic Cardiovascular Surgery; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning Guangxi Zhuang Autonomous Region China
| | - Yifan Zhou
- Department of Thoracic Cardiovascular Surgery; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning Guangxi Zhuang Autonomous Region China
| | - Baijun Li
- Department of Thoracic Cardiovascular Surgery; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning Guangxi Zhuang Autonomous Region China
| | - Zongrong Li
- Department of Thoracic Cardiovascular Surgery; The People's Hospital of Guangxi Zhuang Autonomous Region; Nanning Guangxi Zhuang Autonomous Region China
| |
Collapse
|
17
|
Silva IT, Munkert J, Nolte E, Schneider NFZ, Rocha SC, Ramos ACP, Kreis W, Braga FC, de Pádua RM, Taranto AG, Cortes V, Barbosa LA, Wach S, Taubert H, Simões CMO. Cytotoxicity of AMANTADIG - a semisynthetic digitoxigenin derivative - alone and in combination with docetaxel in human hormone-refractory prostate cancer cells and its effect on Na +/K +-ATPase inhibition. Biomed Pharmacother 2018; 107:464-474. [PMID: 30107342 DOI: 10.1016/j.biopha.2018.08.028] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2018] [Revised: 07/26/2018] [Accepted: 08/06/2018] [Indexed: 01/29/2023] Open
Abstract
Cardiac glycosides (CGs) are natural compounds used to treat congestive heart failure. They have garnered attention as a potential cancer treatment option, especially because they bind to Na+/K+-ATPase as a target and activate intracellular signaling pathways leading to a variety of cellular responses. In this study we evaluated AMANTADIG, a semisynthetic cardenolide derivative, for its cytotoxic activity in two human androgen-insensitive prostate carcinoma cell lines, and the potential synergistic effects with docetaxel. AMANTADIG induced cytotoxic effects in both cell lines, and a combination with docetaxel showed a moderate and strong synergism in DU145 and PC-3 cells, respectively, at concentrations considerably lower than their IC50 values. Cell cycle analyses showed that AMANTADIG and its synergistic combination induced G2/M arrest of DU145 and PC-3 cells by modulating Cyclin B1, CDK1, p21 and, mainly, survivin expression, a promising target in cancer therapy. Furthermore, AMANTADIG presented reduced toxicity toward non-cancerous cell type (PBMC), and computational docking studies disclosed high-affinity binding to the Na+/K+-ATPase α subunit, a result that was experimentally confirmed by Na+/K+-ATPase inhibition assays. Hence, AMANTADIG inhibited Na+/K+-ATPase activity in PC-3 cells, as well as in purified pig kidney at nanomolar range. Altogether, these data highlight the potent effects of AMANTADIG in combination with docetaxel and offer important insights for the development of more effective and selective therapies against prostate cancer.
Collapse
Affiliation(s)
- Izabella Thaís Silva
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil; Departamento de Ciências Farmacêuticas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Jennifer Munkert
- Department of Biology, Chair of Pharmaceutical Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Elke Nolte
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | | | - Sayonarah Carvalho Rocha
- Laboratório de Bioquímica Celular, Faculdade de Bioquímica, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, Brazil
| | - Ana Carolina Pacheco Ramos
- Laboratório de Bioquímica Celular, Faculdade de Bioquímica, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, Brazil
| | - Wolfgang Kreis
- Department of Biology, Chair of Pharmaceutical Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Fernão Castro Braga
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Rodrigo Maia de Pádua
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Alex G Taranto
- Laboratório de Química Farmacêutica Medicinal, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, Brazil
| | - Vanessa Cortes
- Laboratório de Bioquímica Celular, Faculdade de Bioquímica, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, Brazil
| | - Leandro Augusto Barbosa
- Laboratório de Bioquímica Celular, Faculdade de Bioquímica, Universidade Federal de São João del Rei, Campus Centro-Oeste Dona Lindu, Divinópolis, Brazil
| | - Sven Wach
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | - Helge Taubert
- Department of Urology and Pediatric Urology, University Hospital Erlangen, Erlangen, Germany
| | | |
Collapse
|
18
|
Gordon EM, Ravicz JR, Liu S, Chawla SP, Hall FL. Cell cycle checkpoint control: The cyclin G1/Mdm2/p53 axis emerges as a strategic target for broad-spectrum cancer gene therapy - A review of molecular mechanisms for oncologists. Mol Clin Oncol 2018; 9:115-134. [PMID: 30101008 PMCID: PMC6083405 DOI: 10.3892/mco.2018.1657] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 06/14/2018] [Indexed: 12/13/2022] Open
Abstract
Basic research in genetics, biochemistry and cell biology has identified the executive enzymes and protein kinase activities that regulate the cell division cycle of all eukaryotic organisms, thereby elucidating the importance of site-specific protein phosphorylation events that govern cell cycle progression. Research in cancer genomics and virology has provided meaningful links to mammalian checkpoint control elements with the characterization of growth-promoting proto-oncogenes encoding c-Myc, Mdm2, cyclins A, D1 and G1, and opposing tumor suppressor proteins, such as p53, pRb, p16INK4A and p21WAF1, which are commonly dysregulated in cancer. While progress has been made in identifying numerous enzymes and molecular interactions associated with cell cycle checkpoint control, the marked complexity, particularly the functional redundancy, of these cell cycle control enzymes in mammalian systems, presents a major challenge in discerning an optimal locus for therapeutic intervention in the clinical management of cancer. Recent advances in genetic engineering, functional genomics and clinical oncology converged in identifying cyclin G1 (CCNG1 gene) as a pivotal component of a commanding cyclin G1/Mdm2/p53 axis and a strategic locus for re-establishing cell cycle control by means of therapeutic gene transfer. The purpose of the present study is to provide a focused review of cycle checkpoint control as a practicum for clinical oncologists with an interest in applied molecular medicine. The aim is to present a unifying model that: i) clarifies the function of cyclin G1 in establishing proliferative competence, overriding p53 checkpoints and advancing cell cycle progression; ii) is supported by studies of inhibitory microRNAs linking CCNG1 expression to the mechanisms of carcinogenesis and viral subversion; and iii) provides a mechanistic basis for understanding the broad-spectrum anticancer activity and single-agent efficacy observed with dominant-negative cyclin G1, whose cytocidal mechanism of action triggers programmed cell death. Clinically, the utility of companion diagnostics for cyclin G1 pathways is anticipated in the staging, prognosis and treatment of cancers, including the potential for rational combinatorial therapies.
Collapse
Affiliation(s)
- Erlinda M Gordon
- Cancer Center of Southern California/Sarcoma Oncology Center, Santa Monica, CA 90403, USA.,Aveni Foundation, Santa Monica, CA 90405, USA.,DELTA Next-Gen, LLC, Santa Monica, CA 90405, USA
| | - Joshua R Ravicz
- Cancer Center of Southern California/Sarcoma Oncology Center, Santa Monica, CA 90403, USA
| | - Seiya Liu
- Department of Cell Biology, Harvard University, Cambridge, MA 02138, USA
| | - Sant P Chawla
- Cancer Center of Southern California/Sarcoma Oncology Center, Santa Monica, CA 90403, USA
| | - Frederick L Hall
- Aveni Foundation, Santa Monica, CA 90405, USA.,DELTA Next-Gen, LLC, Santa Monica, CA 90405, USA
| |
Collapse
|
19
|
Mohamed MS, Abdelhamid AO, Almutairi FM, Ali AG, Bishr MK. Induction of apoptosis by pyrazolo[3,4-d]pyridazine derivative in lung cancer cells via disruption of Bcl-2/Bax expression balance. Bioorg Med Chem 2017; 26:623-629. [PMID: 29290491 DOI: 10.1016/j.bmc.2017.12.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 12/12/2017] [Accepted: 12/19/2017] [Indexed: 01/08/2023]
Abstract
In the rapidly expanding era of cancer target therapy, regulators of apoptosis are emerging as attractive therapeutic targets. X-linked inhibitor of apoptosis (XIAP) is of specific interest owing to its characteristic overexpression in a wide variety of neoplasms, with a resultant survival advantage for tumor cells and treatment resistance. In this study, we examined three pyrazolo [3,4-d] pyridazine derivatives (PPDs) through molecular modeling and studied their modes of interaction with XIAP-BIR3 domain. PPD-1, which possessed the highest binding affinity with XIAP, was tested on A549 (lung cancer cell line); HCT-116 (colorectal carcinoma cell line); HEPG2 (liver carcinoma cell line), HFB4 (normal human skin melanocyte cell line) and WI-38 (human embryonic lung fibroblasts). In comparison to cisplatin as a positive control, PPD-1 yielded remarkable cytotoxicity on all cancer cell lines, with the highest anti-tumor activity on A549 and a favorable therapeutic ratio. Flow cytometry studies concluded that PPD-1 treatment induces Sub G1 and G2/M cell cycle arrest and apoptosis. The percentage of apoptotic cells in PPD-1 treated A549 cells was considerably higher than that in untreated cells (10.06% vs 0.57%, respectively). To further investigate the mechanism of induction of apoptosis by PPD-1, Real time-PCR was used to quantify the expression levels of key apoptotic regulators. Significant overexpression of the effector capsase-3, pro-apoptotic bax and tumor suppressor gene p53 were noted as compared to untreated cells (7.19 folds, 7.28 folds, and 5.08 folds, respectively). Moreover, PPD-1 inhibited the expression of the anti-apoptotic bcl-2 gene to 0.22 folds. These findings demonstrate that PPD-1 treatment disrupts the Bcl-2/BAX balance in lung cancer cell lines, leading to apoptosis induction possibly through intrinsic mitochondria-dependent pathway. These novel insights elucidate the mechanism of PPD-1 cytotoxicity in lung cancer cell lines and offer a promising therapeutic approach that needs further study.
Collapse
Affiliation(s)
- Mervat S Mohamed
- Department of Biochemistry, Faculty of Science, University of Tabuk, Saudi Arabia; Department of Chemistry, Biochemistry Speciality, Faculty of Science, Cairo University, Egypt.
| | - Abdou O Abdelhamid
- Department of Chemistry, Organic Chemistry Speciality, Faculty of Science, Cairo University, Egypt
| | - Fahad M Almutairi
- Department of Biochemistry, Faculty of Science, University of Tabuk, Saudi Arabia
| | - Ayat G Ali
- Department of Biochemistry, El Sahel Teaching Hospital, Cairo, Egypt
| | - Mai K Bishr
- Department of Radiotherapy, Children's Cancer Hospital Egypt (CCHE), Cairo, Egypt
| |
Collapse
|
20
|
Zhao RY. Yeast for virus research. MICROBIAL CELL (GRAZ, AUSTRIA) 2017; 4:311-330. [PMID: 29082230 PMCID: PMC5657823 DOI: 10.15698/mic2017.10.592] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 08/27/2017] [Indexed: 12/25/2022]
Abstract
Budding yeast (Saccharomyces cerevisiae) and fission yeast (Schizosaccharomyces pombe) are two popular model organisms for virus research. They are natural hosts for viruses as they carry their own indigenous viruses. Both yeasts have been used for studies of plant, animal and human viruses. Many positive sense (+) RNA viruses and some DNA viruses replicate with various levels in yeasts, thus allowing study of those viral activities during viral life cycle. Yeasts are single cell eukaryotic organisms. Hence, many of the fundamental cellular functions such as cell cycle regulation or programed cell death are highly conserved from yeasts to higher eukaryotes. Therefore, they are particularly suited to study the impact of those viral activities on related cellular activities during virus-host interactions. Yeasts present many unique advantages in virus research over high eukaryotes. Yeast cells are easy to maintain in the laboratory with relative short doubling time. They are non-biohazardous, genetically amendable with small genomes that permit genome-wide analysis of virologic and cellular functions. In this review, similarities and differences of these two yeasts are described. Studies of virologic activities such as viral translation, viral replication and genome-wide study of virus-cell interactions in yeasts are highlighted. Impacts of viral proteins on basic cellular functions such as cell cycle regulation and programed cell death are discussed. Potential applications of using yeasts as hosts to carry out functional analysis of small viral genome and to develop high throughput drug screening platform for the discovery of antiviral drugs are presented.
Collapse
Affiliation(s)
- Richard Yuqi Zhao
- Department of Pathology, Department of Microbiology and Immunology, Institute of Global Health, and Institute of Human Virology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
21
|
Host Cell S Phase Restricts Legionella pneumophila Intracellular Replication by Destabilizing the Membrane-Bound Replication Compartment. mBio 2017; 8:mBio.02345-16. [PMID: 28830950 PMCID: PMC5565972 DOI: 10.1128/mbio.02345-16] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
Legionella pneumophila grows within cells ranging from environmental amoebae to human macrophages. In spite of this conserved strategy of pathogenesis, identification of host factors that restrict L. pneumophila intracellular replication has not been extended outside components of the mammalian innate immune response. We performed a double-stranded RNA (dsRNA) screen against more than 50% of the Drosophila melanogaster annotated open reading frames (ORFs) to identify host cell factors that restrict L. pneumophila. The majority of analyzed dsRNAs that stimulated L. pneumophila intracellular replication were directed against host proteins involved in protein synthesis or cell cycle control. Consistent with disruption of the cell cycle stimulating intracellular replication, proteins involved in translation initiation also resulted in G1 arrest. Stimulation of replication was dependent on the stage of cell cycle arrest, as dsRNAs causing arrest during S phase had an inhibitory effect on intracellular replication. The inhibitory effects of S phase arrest could be recapitulated in a human cell line, indicating that cell cycle control of L. pneumophila replication is evolutionarily conserved. Synchronized HeLa cell populations in S phase and challenged with L. pneumophila failed to progress through the cell cycle and were depressed for supporting intracellular replication. Poor bacterial replication in S phase was associated with loss of the vacuole membrane barrier, resulting in exposure of bacteria to the cytosol and their eventual degradation. These results are consistent with the model that S phase is inhibitory for L. pneumophila intracellular survival as a consequence of failure to maintain the integrity of the membrane surrounding intracellular bacteria. Legionella pneumophila has the ability to replicate within human macrophages and amoebal hosts. Here, we report that the host cell cycle influences L. pneumophila intracellular replication. Our data demonstrate that the G1 and G2/M phases of the host cell cycle are permissive for bacterial replication, while S phase is toxic for the bacterium. L. pneumophila replicates poorly within host cells present in S phase. The inability of L. pneumophila to replicate relies on its failure to control the integrity of its vacuole, leading to cytosolic exposure of the bacteria and eventual degradation. The data presented here argue that growth-arrested host cells that are encountered by L. pneumophila in either the environment or within human hosts are ideal targets for intracellular replication because their transit through S phase is blocked.
Collapse
|
22
|
Yu Z, Ruter DL, Kushner EJ, Bautch VL. Excess centrosomes induce p53-dependent senescence without DNA damage in endothelial cells. FASEB J 2017. [PMID: 28626028 DOI: 10.1096/fj.201601320r] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Tumor blood vessels support tumor growth and progression. Centrosomes are microtubule organization centers in cells, and often up to 30% of tumor endothelial cells (ECs) acquire excess (>2) centrosomes. Although excess centrosomes can lead to aneuploidy and chromosome instability in tumor cells, how untransformed ECs respond to excess centrosomes is poorly understood. We found that the frequency of primary human ECs with excess centrosomes was quickly reduced in a p53-dependent manner. Excess centrosomes in ECs were associated with p53 phosphorylation at Ser33, increased p21 levels, and decreased cell proliferation and expression of senescence markers, but independent of DNA damage and apoptosis. Aspects of the senescence-associated phenotype were also observed in mouse ECs that were isolated from tumors with excess centrosomes. Primary ECs with excess centrosomes in vascular sprouts also had elevated Ser33 p53 phosphorylation and expressed senescence markers. Our work demonstrates that nontransformed ECs respond differently to excess centrosomes than do most tumor cells-they undergo senescence in vascular sprouts and vessels, which suggests that pathologic outcomes of centrosome overduplication depend on the transformation status of cells.-Yu, Z., Ruter, D. L., Kushner, E. J., Bautch, V. L. Excess centrosomes induce p53-dependent senescence without DNA damage in endothelial cells.
Collapse
Affiliation(s)
- Zhixian Yu
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Dana L Ruter
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Erich J Kushner
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| | - Victoria L Bautch
- Department of Biology, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
23
|
Merigliano C, Marzio A, Renda F, Somma MP, Gatti M, Vernì F. A Role for the Twins Protein Phosphatase (PP2A-B55) in the Maintenance of Drosophila Genome Integrity. Genetics 2017; 205:1151-1167. [PMID: 28040742 PMCID: PMC5340330 DOI: 10.1534/genetics.116.192781] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 12/21/2016] [Indexed: 01/14/2023] Open
Abstract
The protein phosphatase 2A (PP2A) is a conserved heterotrimeric enzyme that regulates several cellular processes including the DNA damage response and mitosis. Consistent with these functions, PP2A is mutated in many types of cancer and acts as a tumor suppressor. In mammalian cells, PP2A inhibition results in DNA double strand breaks (DSBs) and chromosome aberrations (CABs). However, the mechanisms through which PP2A prevents DNA damage are still unclear. Here, we focus on the role of the Drosophila twins (tws) gene in the maintenance of chromosome integrity; tws encodes the B regulatory subunit (B/B55) of PP2A. Mutations in tws cause high frequencies of CABs (0.5 CABs/cell) in Drosophila larval brain cells and lead to an abnormal persistence of γ-H2Av repair foci. However, mutations that disrupt the PP4 phosphatase activity impair foci dissolution but do not cause CABs, suggesting that a delayed foci regression is not clastogenic. We also show that Tws is required for activation of the G2/M DNA damage checkpoint while PP4 is required for checkpoint recovery, a result that points to a conserved function of these phosphatases from flies to humans. Mutations in the ATM-coding gene tefu are strictly epistatic to tws mutations for the CAB phenotype, suggesting that failure to dephosphorylate an ATM substrate(s) impairs DNA DSBs repair. In addition, mutations in the Ku70 gene, which do not cause CABs, completely suppress CAB formation in tws Ku70 double mutants. These results suggest the hypothesis that an improperly phosphorylated Ku70 protein can lead to DNA damage and CABs.
Collapse
Affiliation(s)
- Chiara Merigliano
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
| | - Antonio Marzio
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
| | - Fioranna Renda
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
| | - Maria Patrizia Somma
- Istituto di Biologia e Patologia Molecolari del Consiglio Nazionale delle Ricerche, Sapienza, Università di Roma, 00185, Italy
| | - Maurizio Gatti
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
- Istituto di Biologia e Patologia Molecolari del Consiglio Nazionale delle Ricerche, Sapienza, Università di Roma, 00185, Italy
| | - Fiammetta Vernì
- Dipartimento di Biologia e Biotecnologie "C. Darwin," Sapienza, Università di Roma, 00185, Italy
| |
Collapse
|
24
|
Jeon YJ, Bang W, Cho JH, Lee RH, Kim SH, Kim MS, Park SM, Shin JC, Chung HJ, Oh KB, Seo JM, Ko S, Shim JH, Chae JI. Kahweol induces apoptosis by suppressing BTF3 expression through the ERK signaling pathway in non-small cell lung cancer cells. Int J Oncol 2016; 49:2294-2302. [DOI: 10.3892/ijo.2016.3727] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 08/04/2016] [Indexed: 11/06/2022] Open
|
25
|
CDK-1 Inhibition in G2 Stabilizes Kinetochore-Microtubules in the following Mitosis. PLoS One 2016; 11:e0157491. [PMID: 27281342 PMCID: PMC4900577 DOI: 10.1371/journal.pone.0157491] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 05/30/2016] [Indexed: 11/18/2022] Open
Abstract
Cell proliferation is driven by cyclical activation of cyclin-dependent kinases (CDKs), which produce distinct biochemical cell cycle phases. Mitosis (M phase) is orchestrated by CDK-1, complexed with mitotic cyclins. During M phase, chromosomes are segregated by a bipolar array of microtubules called the mitotic spindle. The essential bipolarity of the mitotic spindle is established by the kinesin-5 Eg5, but factors influencing the maintenance of spindle bipolarity are not fully understood. Here, we describe an unexpected link between inhibiting CDK-1 before mitosis and bipolar spindle maintenance. Spindles in human RPE-1 cells normally collapse to monopolar structures when Eg5 is inhibited at metaphase. However, we found that inhibition of CDK-1 in the G2 phase of the cell cycle improved the ability of RPE-1 cells to maintain spindle bipolarity without Eg5 activity in the mitosis immediately after release from CDK-1 inhibition. This improved bipolarity maintenance correlated with an increase in the stability of kinetochore-microtubules, the subset of microtubules that link chromosomes to the spindle. The improvement in bipolarity maintenance after CDK-1 inhibition in G2 required both the kinesin-12 Kif15 and increased stability of kinetochore-microtubules. Consistent with increased kinetochore-microtubule stability, we find that inhibition of CDK-1 in G2 impairs mitotic fidelity by increasing the incidence of lagging chromosomes in anaphase. These results suggest that inhibition of CDK-1 in G2 causes unpredicted effects in mitosis, even after CDK-1 inhibition is relieved.
Collapse
|
26
|
Piña FJ, Fleming T, Pogliano K, Niwa M. Reticulons Regulate the ER Inheritance Block during ER Stress. Dev Cell 2016; 37:279-88. [PMID: 27117666 DOI: 10.1016/j.devcel.2016.03.025] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 03/07/2016] [Accepted: 03/28/2016] [Indexed: 01/05/2023]
Abstract
Segregation of functional organelles during the cell cycle is crucial to generate healthy daughter cells. In Saccharomyces cerevisiae, ER stress causes an ER inheritance block to ensure cells inherit a functional ER. Here, we report that formation of tubular ER in the mother cell, the first step in ER inheritance, depends on functional symmetry between the cortical ER (cER) and perinuclear ER (pnER). ER stress induces functional asymmetry, blocking tubular ER formation and ER inheritance. Using fluorescence recovery after photobleaching, we show that the ER chaperone Kar2/BiP fused to GFP and an ER membrane reporter, Hmg1-GFP, behave differently in the cER and pnER. The functional asymmetry and tubular ER formation depend on Reticulons/Yop1, which maintain ER structure. LUNAPARK1 deletion in rtn1Δrtn2Δyop1Δ cells restores the pnER/cER functional asymmetry, tubular ER generation, and ER inheritance blocks. Thus, Reticulon/Yop1-dependent changes in ER structure are linked to ER inheritance during the yeast cell cycle.
Collapse
Affiliation(s)
- Francisco Javier Piña
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, NSB#1, Room 5328, La Jolla, CA 92093-0377, USA
| | - Tinya Fleming
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, NSB#1, Room 4113, La Jolla, CA 92093-0377, USA
| | - Kit Pogliano
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, NSB#1, Room 4113, La Jolla, CA 92093-0377, USA
| | - Maho Niwa
- Section of Molecular Biology, Division of Biological Sciences, University of California, San Diego, NSB#1, Room 5328, La Jolla, CA 92093-0377, USA.
| |
Collapse
|
27
|
Yang J, Chen W, Fan Y, Zhang H, Wang W, Zhang H. Ubiquitin Protein Ligase Ring2 Is Involved in S-phase Checkpoint and DNA Damage in Cells Exposed to Benzo[a]pyrene. J Biochem Mol Toxicol 2016; 30:481-488. [PMID: 27095601 DOI: 10.1002/jbt.21811] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 03/11/2016] [Accepted: 03/22/2016] [Indexed: 01/08/2023]
Abstract
Previous studies in our laboratory demonstrated that Ring2 may affect DNA damage and repair through pathways other than through regulating the expression of the nucleotide excision repair protein. In a series of experiments using wild-type cell (16HBE and WI38) and small interfering RNA (siRNA) Ring2 cells exposed to benzo[a]pyrene (BaP), we evaluated the cell cycle and DNA damage. The benzo(a)pyrene-7,8-dihydrodiol-9,10-epoxide (BPDE-DNA) adduct assay demonstrated that in vitro exposure to BaP increased DNA damage in a time- and dose-dependent manner in wild-type and siRNA Ring2 cells. Analysis of covariance showed that a decrease of Ring2 caused DNA hypersensitivity to BaP. Flow cytometry results and proliferating cell nuclear antigen levels indicated that inhibition of Ring2 attenuated the effect of BaP on S-phase arrest. Taken together, these data implied that the lower proportion of cells in the S phase induced by inhibition of Ring2 may play an important role in DNA hypersensitivity to BaP.
Collapse
Affiliation(s)
- Jin Yang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, 030001, Taiyuan, People's Republic of China.
| | - Wentao Chen
- Department of Occupational Health, School of Public Health, Shanxi Medical University, 030001, Taiyuan, People's Republic of China
| | - Yanfeng Fan
- Department of Occupational Health, School of Public Health, Shanxi Medical University, 030001, Taiyuan, People's Republic of China
| | - Huitao Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, 030001, Taiyuan, People's Republic of China
| | - Wubin Wang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, 030001, Taiyuan, People's Republic of China
| | - Hongjie Zhang
- Department of Occupational Health, School of Public Health, Shanxi Medical University, 030001, Taiyuan, People's Republic of China
| |
Collapse
|
28
|
den Hollander P, Rawls K, Tsimelzon A, Shepherd J, Mazumdar A, Hill J, Fuqua SAW, Chang JC, Osborne CK, Hilsenbeck SG, Mills GB, Brown PH. Phosphatase PTP4A3 Promotes Triple-Negative Breast Cancer Growth and Predicts Poor Patient Survival. Cancer Res 2016; 76:1942-53. [PMID: 26921331 PMCID: PMC4873402 DOI: 10.1158/0008-5472.can-14-0673] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2014] [Accepted: 10/15/2015] [Indexed: 12/21/2022]
Abstract
Triple-negative breast cancer (TNBC) has the worst prognosis of all breast cancers, and women diagnosed with TNBC currently lack targeted treatment options. To identify novel targets for TNBC, we evaluated phosphatase expression in breast tumors and characterized their contributions to in vitro and in vivo growth of TNBC. Using Affymetrix microarray analysis of 102 breast cancers, we identified 146 phosphatases that were significantly differentially expressed in TNBC compared with estrogen receptor (ER)-positive tumors. Of these, 19 phosphatases were upregulated (0.66-fold; FDR = 0.05) in TNBC compared with ER-positive breast cancers. We knocked down 17 overexpressed phosphatases in four triple-negative and four ER-positive breast cancer lines using specific siRNAs and found that depletion of six of these phosphatases significantly reduced growth and anchorage-independent growth of TNBC cells to a greater extent than ER-positive cell lines. Further analysis of the phosphatase PTP4A3 (also known as PRL-3) demonstrated its requirement for G1-S cell-cycle progression in all breast cancer cells, but PTP4A3 regulated apoptosis selectively in TNBC cells. In addition, PTP4A3 inhibition reduced the growth of TNBC tumors in vivo Moreover, in silico analysis revealed the PTP4A3 gene to be amplified in 29% of basal-like breast cancers, and high expression of PTP4A3 could serve as an independent prognostic indicator for worse overall survival. Collectively, these studies define the importance of phosphatase overexpression in TNBC and lay the foundation for the development of new targeted therapies directed against phosphatases or their respective signaling pathways for TNBC patients. Cancer Res; 76(7); 1942-53. ©2016 AACR.
Collapse
Affiliation(s)
- Petra den Hollander
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Kathryn Rawls
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anna Tsimelzon
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jonathan Shepherd
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas
| | - Abhijit Mazumdar
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jamal Hill
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Suzanne A W Fuqua
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Jenny C Chang
- Methodist Cancer Center, The Methodist Hospital Research Institute, Houston, Texas
| | - C Kent Osborne
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Susan G Hilsenbeck
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, Texas
| | - Gordon B Mills
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Powel H Brown
- Department of Clinical Cancer Prevention, The University of Texas MD Anderson Cancer Center, Houston, Texas. Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
29
|
Abstract
Neurons are usually regarded as postmitotic cells that undergo apoptosis in response to cell cycle reactivation. Nevertheless, recent evidence indicates the existence of a defined developmental program that induces DNA replication in specific populations of neurons, which remain in a tetraploid state for the rest of their adult life. Similarly, de novo neuronal tetraploidization has also been described in the adult brain as an early hallmark of neurodegeneration. The aim of this review is to integrate these recent developments in the context of cell cycle regulation and apoptotic cell death in neurons. We conclude that a variety of mechanisms exists in neuronal cells for G1/S and G2/M checkpoint regulation. These mechanisms, which are connected with the apoptotic machinery, can be modulated by environmental signals and the neuronal phenotype itself, thus resulting in a variety of outcomes ranging from cell death at the G1/S checkpoint to full proliferation of differentiated neurons.
Collapse
Key Words
- AD, Alzheimer disease
- BDNF, brain-derived neurotrophic factor
- BrdU, 5-bromo-2′-deoxyuridine
- CKI, Cdk-inhibitor
- CNS, central nervous system
- Cdk, cyclin-dependent kinase
- Cip/Kip, cyclin inhibitor protein/kinase inhibitor protein
- G0, quiescent state
- G1, growth phase 1
- G2, growth phase 2
- Ink, inhibitor of kinase
- Mcm2, minichromosome maintenance 2
- PCNA, proliferating cell nuclear antigen
- PD, Parkinson disease
- RGCs, retinal ganglion cells
- Rb, Retinoblastoma
- S-phase
- S-phase, synthesis phase.
- apoptosis
- cell cycle re-entry
- mitosis
- neuron
- p38MAPK, p38 mitogen-activated protein kinase
- p75NTR, neurotrophin receptor p75
- tetraploid
Collapse
Affiliation(s)
- José M Frade
- a Department of Molecular, Cellular and Developmental Neurobiology; Instituto Cajal; Consejo Superior de Investigaciones Científicas (IC-CSIC) ; Madrid , Spain
| | | |
Collapse
|
30
|
Vinogradov AE. Accelerated pathway evolution in mouse-like rodents involves cell cycle control. Mamm Genome 2015; 26:609-18. [PMID: 26424469 DOI: 10.1007/s00335-015-9605-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 09/23/2015] [Indexed: 10/23/2022]
Abstract
Rodents include both the cancer-susceptible short-lived mouse and the two unrelated cancer-resistant long-lived mole-rats. In this work, their genomes were analyzed with the goal to reveal pathways enriched in genes, which are more similar between the mole-rats than between the mouse and the naked mole-rat. The pathways related to cell cycle control were prominent. They include external signal transduction and all cell cycle stages. There are several stem cell pathways among them. The other enriched pathways involve ubiquitin-dependent protein degradation, immunity, mRNA splicing, and apoptosis. The ubiquitin-dependent protein degradation is a core of network of enriched pathways. However, this phenomenon is not specific for the mouse and the mole-rats. The other muroid species show features similar to the mouse, whereas the non-muroid rodents and the human show features similar to the mole-rats. The higher ratio of non-synonymous to synonymous nucleotide substitutions (dN/dS) indicates the accelerated evolution of revealed pathways in the muroid rodents (except the blind mole-rat). Paradoxically, the dN/dS averaged over the whole genome is lower in the muroids, i.e., the purifying selection is generally stronger in them. In practical sense, these data suggest caveat for using muroid rodents (mouse, rat, and hamsters) as biomedical models of human conditions involving cell cycle and show the network of pathways where muroid genes are most different (compared with non-muroid) from human genes. The guinea pig is emphasized as a more suitable rodent model for biomedical research involving cell cycle.
Collapse
|
31
|
Piña FJ, Niwa M. The ER Stress Surveillance (ERSU) pathway regulates daughter cell ER protein aggregate inheritance. eLife 2015; 4. [PMID: 26327697 PMCID: PMC4555637 DOI: 10.7554/elife.06970] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 08/04/2015] [Indexed: 12/11/2022] Open
Abstract
Stress induced by cytoplasmic protein aggregates can have deleterious consequences for the cell, contributing to neurodegeneration and other diseases. Protein aggregates are also formed within the endoplasmic reticulum (ER), although the fate of ER protein aggregates, specifically during cell division, is not well understood. By simultaneous visualization of both the ER itself and ER protein aggregates, we found that ER protein aggregates that induce ER stress are retained in the mother cell by activation of the ER Stress Surveillance (ERSU) pathway, which prevents inheritance of stressed ER. In contrast, under conditions of normal ER inheritance, ER protein aggregates can enter the daughter cell. Thus, whereas cytoplasmic protein aggregates are retained in the mother cell to protect the functional capacity of daughter cells, the fate of ER protein aggregates is determined by whether or not they activate the ERSU pathway to impede transmission of the cortical ER during the cell cycle.
Collapse
Affiliation(s)
- Francisco J Piña
- Division of Biological Sciences, Section of Molecular Biology, Univeristy of California, San Diego, San Diego, United States
| | - Maho Niwa
- Division of Biological Sciences, Section of Molecular Biology, Univeristy of California, San Diego, San Diego, United States
| |
Collapse
|
32
|
Abstract
Cell division relies on coordinated regulation of the cell cycle. A process including a well-defined series of strictly regulated molecular mechanisms involving cyclin-dependent kinases, retinoblastoma protein, and polo-like kinases. Dysfunctions in cell cycle regulation are associated with disease such as cancer, diabetes, and neurodegeneration. Compartmentalization of cellular signaling is a common strategy used to ensure the accuracy and efficiency of cellular responses. Compartmentalization of intracellular signaling is maintained by scaffolding proteins, such as A-kinase anchoring proteins (AKAPs). AKAPs are characterized by their ability to anchor the regulatory subunits of protein kinase A (PKA), and thereby achieve guidance to different cellular locations via various targeting domains. Next to PKA, AKAPs also associate with several other signaling elements including receptors, ion channels, protein kinases, phosphatases, small GTPases, and phosphodiesterases. Taking the amount of possible AKAP signaling complexes and their diverse localization into account, it is rational to believe that such AKAP-based complexes regulate several critical cellular events of the cell cycle. In fact, several AKAPs are assigned as tumor suppressors due to their vital roles in cell cycle regulation. Here, we first briefly discuss the most important players of cell cycle progression. After that, we will review our recent knowledge of AKAPs linked to the regulation and progression of the cell cycle, with special focus on AKAP12, AKAP8, and Ezrin. At last, we will discuss this specific AKAP subset in relation to diseases with focus on a diverse subset of cancer.
Collapse
Affiliation(s)
- B Han
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands. .,Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands.
| | - W J Poppinga
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| | - M Schmidt
- Department of Molecular Pharmacology, University of Groningen, Groningen, The Netherlands.,Groningen Research Institute for Asthma and COPD, GRIAC, Groningen, The Netherlands
| |
Collapse
|
33
|
Levine MT, Vander Wende HM, Malik HS. Mitotic fidelity requires transgenerational action of a testis-restricted HP1. eLife 2015; 4:e07378. [PMID: 26151671 PMCID: PMC4491702 DOI: 10.7554/elife.07378] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2015] [Accepted: 06/08/2015] [Indexed: 01/02/2023] Open
Abstract
Sperm-packaged DNA must undergo extensive reorganization to ensure its timely participation in embryonic mitosis. Whereas maternal control over this remodeling is well described, paternal contributions are virtually unknown. In this study, we show that Drosophila melanogaster males lacking Heterochromatin Protein 1E (HP1E) sire inviable embryos that undergo catastrophic mitosis. In these embryos, the paternal genome fails to condense and resolve into sister chromatids in synchrony with the maternal genome. This delay leads to a failure of paternal chromosomes, particularly the heterochromatin-rich sex chromosomes, to separate on the first mitotic spindle. Remarkably, HP1E is not inherited on mature sperm chromatin. Instead, HP1E primes paternal chromosomes during spermatogenesis to ensure faithful segregation post-fertilization. This transgenerational effect suggests that maternal control is necessary but not sufficient for transforming sperm DNA into a mitotically competent pronucleus. Instead, paternal action during spermiogenesis exerts post-fertilization control to ensure faithful chromosome segregation in the embryo. DOI:http://dx.doi.org/10.7554/eLife.07378.001 The genetic information of cells is packaged into structures called chromosomes, which are made up of long strands of DNA that are wrapped around proteins to form a structure called chromatin. The cells of most animals contain two copies of every chromosome, but the egg and sperm cells contain only one copy. This means that when an egg fuses with a sperm cell during fertilization, the resulting ‘zygote’ will contain two copies of each chromosome—one inherited from the mother, and one from the father. These chromosomes duplicate and divide many times within the developing embryo in a process known as mitosis. The first division of the zygote is particularly complicated, as the egg and sperm chromosomes must go through extensive—and yet different—chromatin reorganization processes. For instance, paternal DNA is inherited via sperm, where specialized sperm proteins package the DNA more tightly than in the maternal DNA, which is packaged by histone proteins used throughout development. For paternal DNA to participate in mitosis in the embryo, it must first undergo a transition to a histone-packaged state. Despite these differences, both maternal and paternal chromosomes must undergo mitosis at the same time if the zygote is to successfully divide. Although it is known that the egg cell contributes essential proteins that are incorporated into the sperm chromatin to help it reorganize, the importance of paternal proteins in coordinating this process remains poorly understood. Many members of a family of proteins called Heterochromatin Protein 1 (or HP1 for short) have previously been shown to control chromatin organization in plants and animals. In 2012, researchers found that several HP1 proteins are found only in the testes of the fruit fly species Drosophila melanogaster. They predicted that these proteins might help control the reorganization of the paternal chromosomes following fertilization. Levine et al.—including researchers involved in the 2012 study—have now used genetic and cell-based techniques to show that one member of the HP1 family (called HP1E) ensures that the paternal chromosomes are ready for cell division at the same time as the maternal chromosomes. Male flies that are unable to produce this protein do not have any offspring because, while these flies' sperm can fertilize eggs, the resulting zygotes cannot divide as normal. Further experiments revealed that HP1E is not inherited through the chromatin of mature sperm, but instead influences the structure of the chromosomes during the final stages of the development of the sperm cells in the fly testes. This study shows that both maternal and paternal proteins are needed to control how the paternal chromosomes reorganize in fruit fly embryos. Although difficult to discover and decipher, this work re-emphasizes the importance of paternal epigenetic contributions—changes that alter how DNA is read, without changing the DNA sequence itself—for ensuring the viability of resulting offspring. Future work will reveal both the molecular mechanism of this epigenetic transfer of information, as well as why certain Drosophila species are able to naturally overcome the loss of the essential HP1E protein. DOI:http://dx.doi.org/10.7554/eLife.07378.002
Collapse
Affiliation(s)
- Mia T Levine
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Helen M Vander Wende
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| | - Harmit S Malik
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, Seattle, United States
| |
Collapse
|
34
|
Sobol A, Galluzzo P, Weber MJ, Alani S, Bocchetta M. Depletion of Amyloid Precursor Protein (APP) causes G0 arrest in non-small cell lung cancer (NSCLC) cells. J Cell Physiol 2015; 230:1332-41. [PMID: 25502341 DOI: 10.1002/jcp.24875] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 12/05/2014] [Indexed: 01/24/2023]
Abstract
We recently reported that Amyloid Precursor Protein (APP) regulates global protein synthesis in a variety of human dividing cells, including non-small cell lung cancer (NSCLC) cells. More specifically, APP depletion causes an increase of both cap- and IRES-dependent translation. Since growth and proliferation are tightly coupled processes, here, we asked what effects artificial downregulation of APP could have elicited in NSCLC cells proliferation. APP depletion caused a G0/G1 arrest through destabilization of the cyclin-C protein and reduced pRb phosphorylation at residues Ser802/811. siRNA to cyclin-C mirrored the cell cycle distribution observed when silencing APP. Cells arrested in G0/G1 (and with augmented global protein synthesis) increased their size and underwent a necrotic cell death due to cell membrane permeabilization. These phenotypes were reversed by overexpression of the APP C-terminal domain, indicating a novel role for APP in regulating early cell cycle entry decisions. It is seems that APP moderates the rate of protein synthesis before the cell clears growth factors- and nutrients-dependent checkpoint in mid G1. Our results raise questions on how such processes interact in the context of (at least) dividing NSCLC cells. The data presented here suggest that APP, although required for G0/G1 transitions, moderates the rate of protein synthesis before the cell fully commits to cell cycle progression following mechanisms, which seem additional to concurrent signals deriving from the PI3-K/Akt/mTORC-1 axis. APP appears to play a central role in regulating cell cycle entry with the rate of protein synthesis; and its loss-of-function causes cell size abnormalities and death.
Collapse
Affiliation(s)
- Anna Sobol
- Department of Pathology, Loyola University Chicago Medical Center, Oncology Institute, Maywood, Illinois
| | | | | | | | | |
Collapse
|
35
|
Ovejero-Benito MC, Frade JM. p27(Kip1) participates in the regulation of endoreplication in differentiating chick retinal ganglion cells. Cell Cycle 2015; 14:2311-22. [PMID: 25946375 PMCID: PMC4614947 DOI: 10.1080/15384101.2015.1044175] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Nuclear DNA duplication in the absence of cell division (i.e. endoreplication) leads to somatic polyploidy in eukaryotic cells. In contrast to some invertebrate neurons, whose nuclei may contain up to 200,000-fold the normal haploid DNA amount (C), polyploid neurons in higher vertebrates show only 4C DNA content. To explore the mechanism that prevents extra rounds of DNA synthesis in these latter cells we focused on the chick retina, where a population of tetraploid retinal ganglion cells (RGCs) has been described. We show that differentiating chick RGCs that express the neurotrophic receptors p75 and TrkB while lacking retinoblastoma protein, a feature of tetraploid RGCs, also express p27Kip1. Two different short hairpin RNAs (shRNA) that significantly downregulate p27Kip1 expression facilitated DNA synthesis and increased ploidy in isolated chick RGCs. Moreover, this forced DNA synthesis could not be prevented by Cdk4/6 inhibition, thus suggesting that it is triggered by a mechanism similar to endoreplication. In contrast, p27Kip1 deficiency in mouse RGCs does not lead to increased ploidy despite previous observations have shown ectopic DNA synthesis in RGCs from p27Kip1−/− mice. This suggests that a differential mechanism is used for the regulation of neuronal endoreplication in mammalian versus avian RGCs.
Collapse
Affiliation(s)
- María C Ovejero-Benito
- a Department of Molecular , Cellular, and Developmental Neurobiology; Cajal Institute; IC-CSIC ; Madrid , Spain
| | | |
Collapse
|
36
|
Breast cancer cell line MCF7 escapes from G1/S arrest induced by proteasome inhibition through a GSK-3β dependent mechanism. Sci Rep 2015; 5:10027. [PMID: 25941117 PMCID: PMC4419540 DOI: 10.1038/srep10027] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Accepted: 03/24/2015] [Indexed: 12/18/2022] Open
Abstract
Targeting the ubiquitin proteasome pathway has emerged as a rational approach in the treatment of human cancers. Autophagy has been described as a cytoprotective mechanism to increase tumor cell survival under stress conditions. Here, we have focused on the role of proteasome inhibition in cell cycle progression and the role of autophagy in the proliferation recovery. The study was performed in the breast cancer cell line MCF7 compared to the normal mammary cell line MCF10A. We found that the proteasome inhibitor MG132 induced G1/S arrest in MCF10A, but G2/M arrest in MCF7 cells. The effect of MG132 on MCF7 was reproduced on MCF10A cells in the presence of the glycogen synthase kinase 3β (GSK-3β) inhibitor VII. Similarly, MCF7 cells overexpressing constitutively active GSK-3β behaved like MCF10A cells. On the other hand, MCF10A cells remained arrested after MG132 removal while MCF7 recovered the proliferative capacity. Importantly, this recovery was abolished in the presence of the autophagy inhibitor 3-methyladenine (3-MA). Thus, our results support the relevance of GSK-3β and autophagy as two targets for controlling cell cycle progression and proliferative capacity in MCF7, highlighting the co-treatment of breast cancer cells with 3-MA to synergize the effect of the proteasome inhibition.
Collapse
|
37
|
Spiesser TW, Kühn C, Krantz M, Klipp E. Bud-Localization of CLB2 mRNA Can Constitute a Growth Rate Dependent Daughter Sizer. PLoS Comput Biol 2015; 11:e1004223. [PMID: 25910075 PMCID: PMC4429581 DOI: 10.1371/journal.pcbi.1004223] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 03/03/2015] [Indexed: 11/19/2022] Open
Abstract
Maintenance of cellular size is a fundamental systems level process that requires balancing of cell growth with proliferation. This is achieved via the cell division cycle, which is driven by the sequential accumulation and destruction of cyclins. The regulatory network around these cyclins, particularly in G1, has been interpreted as a size control network in budding yeast, and cell size as being decisive for the START transition. However, it is not clear why disruptions in the G1 network may lead to altered size rather than loss of size control, or why the S-G2-M duration also depends on nutrients. With a mathematical population model comprised of individually growing cells, we show that cyclin translation would suffice to explain the observed growth rate dependence of cell volume at START. Moreover, we assess the impact of the observed bud-localisation of the G2 cyclin CLB2 mRNA, and find that localised cyclin translation could provide an efficient mechanism for measuring the biosynthetic capacity in specific compartments: The mother in G1, and the growing bud in G2. Hence, iteration of the same principle can ensure that the mother cell is strong enough to grow a bud, and that the bud is strong enough for independent life. Cell sizes emerge in the model, which predicts that a single CDK-cyclin pair per growth phase suffices for size control in budding yeast, despite the necessity of the cell cycle network around the cyclins to integrate other cues. Size control seems to be exerted twice, where the G2/M control affects bud size through bud-localized translation of CLB2 mRNA, explaining the dependence of the S-G2-M duration on nutrients. Taken together, our findings suggest that cell size is an emergent rather than a regulatory property of the network linking growth and proliferation.
Collapse
Affiliation(s)
- Thomas W. Spiesser
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Berlin, Germany
- * E-mail: (TWS); (EK)
| | - Clemens Kühn
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Marcus Krantz
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Edda Klipp
- Theoretical Biophysics, Humboldt-Universität zu Berlin, Berlin, Germany
- * E-mail: (TWS); (EK)
| |
Collapse
|
38
|
Yu Y, Duan J, Geng W, Li Q, Jiang L, Li Y, Yu Y, Sun Z. Aberrant cytokinesis and cell fusion result in multinucleation in HepG2 cells exposed to silica nanoparticles. Chem Res Toxicol 2015; 28:490-500. [PMID: 25625797 DOI: 10.1021/tx500473h] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The multinucleation effect of silica nanoparticles (SiNPs) had been determined in our previous studies, but the relative mechanisms of multinucleation and how the multinucleated cells are generated were still not clear. This extensional study was conducted to investigate the mechanisms underlying the formation of multinucleated cells after SiNPs exposure. We first investigated cellular multinucleation, then performed time-lapse confocal imaging to certify whether the multinucleated cells resulted from cell fusion or abnormal cell division. Our results confirmed for the first time that there are three patterns contributing to the SiNPs-induced multinucleation in HepG2 cells: cell fusion, karyokinesis without cytokinesis, and cytokinesis followed by fusion. The chromosomal passenger complex (CPC) deficiency and cell cycle arrest in G1/S and G2/M checkpoints may be responsible for the cell aberrant cytokinesis. The activated MAPK/ERK1/2 signaling and decreased mitosis related proteins might be the underlying mechanism of cell cycle arrest and thus multinucleation. In summary, we confirmed the hypothesis that aberrant cytokinesis and cell fusion resulted in multinucleation in HepG2 cells after SiNPs exposure. Since cell fusion and multinucleation were involved in genetic instability and tumor development, this study suggests the potential ability of SiNPs to induce cellular genetic instability. These findings raise concerns with regard to human health hazards and environmental risks with SiNPs exposure.
Collapse
Affiliation(s)
- Yongbo Yu
- School of Public Health, Capital Medical University , Beijing 100069, P.R. China
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Morphogenesis checkpoint kinase Swe1 is the executor of lipolysis-dependent cell-cycle progression. Proc Natl Acad Sci U S A 2015; 112:E1077-85. [PMID: 25713391 DOI: 10.1073/pnas.1423175112] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Cell growth and division requires the precise duplication of cellular DNA content but also of membranes and organelles. Knowledge about the cell-cycle-dependent regulation of membrane and storage lipid homeostasis is only rudimentary. Previous work from our laboratory has shown that the breakdown of triacylglycerols (TGs) is regulated in a cell-cycle-dependent manner, by activation of the Tgl4 lipase by the major cyclin-dependent kinase Cdc28. The lipases Tgl3 and Tgl4 are required for efficient cell-cycle progression during the G1/S (Gap1/replication phase) transition, at the onset of bud formation, and their absence leads to a cell-cycle delay. We now show that defective lipolysis activates the Swe1 morphogenesis checkpoint kinase that halts cell-cycle progression by phosphorylation of Cdc28 at tyrosine residue 19. Saturated long-chain fatty acids and phytosphingosine supplementation rescue the cell-cycle delay in the Tgl3/Tgl4 lipase-deficient strain, suggesting that Swe1 activity responds to imbalanced sphingolipid metabolism, in the absence of TG degradation. We propose a model by which TG-derived sphingolipids are required to activate the protein phosphatase 2A (PP2A(Cdc55)) to attenuate Swe1 phosphorylation and its inhibitory effect on Cdc28 at the G1/S transition of the cell cycle.
Collapse
|
40
|
Zou Y, Hu M, Lee J, Nambiar SM, Garcia V, Bao Q, Chan JY, Dai G. Nrf2 is essential for timely M phase entry of replicating hepatocytes during liver regeneration. Am J Physiol Gastrointest Liver Physiol 2015; 308:G262-8. [PMID: 25524062 PMCID: PMC4329475 DOI: 10.1152/ajpgi.00332.2014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates various cellular activities, including redox balance, detoxification, metabolism, autophagy, proliferation, and apoptosis. Several studies have demonstrated that Nrf2 regulates hepatocyte proliferation during liver regeneration. The aim of this study was to investigate how Nrf2 modulates the cell cycle of replicating hepatocytes in regenerating livers. Wild-type and Nrf2 null mice were subjected to 2/3 partial hepatectomy (PH) and killed at multiple time points for various analyses. Nrf2 null mice exhibited delayed liver regrowth, although the lost liver mass was eventually restored 7 days after PH. Nrf2 deficiency did not affect the number of hepatocytes entering the cell cycle but did delay hepatocyte mitosis. Mechanistically, the lack of Nrf2 resulted in increased mRNA and protein levels of hepatic cyclin A2 when the remaining hepatocytes were replicating in response to PH. Moreover, Nrf2 deficiency in regenerating livers caused dysregulation of Wee1, Cdc2, and cyclin B1 mRNA and protein expression, leading to decreased Cdc2 activity. Thus, Nrf2 is required for timely M phase entry of replicating hepatocytes by ensuring proper regulation of cyclin A2 and the Wee1/Cdc2/cyclin B1 pathway during liver regeneration.
Collapse
Affiliation(s)
- Yuhong Zou
- 1Department of Biology, School of Science, Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana;
| | - Min Hu
- 1Department of Biology, School of Science, Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana; ,2Department of Pathology, Anhui University of Traditional Chinese Medicine, Hefei, China; and
| | - Joonyong Lee
- 1Department of Biology, School of Science, Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana;
| | - Shashank Manohar Nambiar
- 1Department of Biology, School of Science, Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana;
| | - Veronica Garcia
- 1Department of Biology, School of Science, Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana;
| | - Qi Bao
- 1Department of Biology, School of Science, Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana;
| | - Jefferson Y. Chan
- 3Pathology and Laboratory Medicine, School of Medicine, University of California, Irvine, California
| | - Guoli Dai
- Department of Biology, School of Science, Center for Regenerative Biology and Medicine, Indiana University-Purdue University Indianapolis, Indianapolis, Indiana;
| |
Collapse
|
41
|
Xu LL, Luo WS, Tan N, Xu Q, Xu B, Zhu-Ge FY. Total flavonoids of litchi suppress proliferation of LX2 human hepatic stellate cells by upregulating p27 expression. Shijie Huaren Xiaohua Zazhi 2015; 23:539-546. [DOI: 10.11569/wcjd.v23.i4.539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect of total flavonoids of litchi (TFL) on the proliferation of LX2 human hepatic stellate cells and to explore the underlying mechanism.
METHODS: LX2 cells treated with different concentrations (7.8125, 15.6250, 31.2500, 62.5000, and 125.0000 µg/mL) of TFL were examined for cell growth inhibition using CCK-8. Flow cytometry was used to analyze the changes in cell cycle distribution of LX2 cells. The expression of p27 mRNA and protein in LX2 cells was determined by real-time quantitative PCR and Western blot, respectively.
RESULTS: Exposure to TFL caused significant dose- and time-dependent inhibition of LX2 cell proliferation. TFL induced S-phase cell cycle arrest as shown by flow cytometric analysis. In addition, expression of p27 mRNA and protein in LX2 cells was upregulated in the treatment groups.
CONCLUSION: TFL treatment inhibits LX2 cell proliferation and arrests cells at S phase, and the mechanism may be associated with the upregulation of p27 expression.
Collapse
|
42
|
Zhang Y, Zhu CC, Sun SC. Cyclin B3: an anaphase onset controller in meiosis. Cell Cycle 2015; 14:3013. [PMID: 26496167 PMCID: PMC4825606 DOI: 10.1080/15384101.2015.1084201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Accepted: 07/22/2015] [Indexed: 10/23/2022] Open
Affiliation(s)
- Yu Zhang
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing, China
| | - Cheng-Cheng Zhu
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing, China
| | - Shao-Chen Sun
- College of Animal Science and Technology; Nanjing Agricultural University; Nanjing, China
| |
Collapse
|
43
|
Abstract
The budding yeast Saccharomyces cerevisiae is a powerful model organism for studying fundamental aspects of eukaryotic cell biology. This Primer article presents a brief historical perspective on the emergence of this organism as a premier experimental system over the course of the past century. An overview of the central features of the S. cerevisiae genome, including the nature of its genetic elements and general organization, is also provided. Some of the most common experimental tools and resources available to yeast geneticists are presented in a way designed to engage and challenge undergraduate and graduate students eager to learn more about the experimental amenability of budding yeast. Finally, a discussion of several major discoveries derived from yeast studies highlights the far-reaching impact that the yeast system has had and will continue to have on our understanding of a variety of cellular processes relevant to all eukaryotes, including humans.
Collapse
|
44
|
Jossé R, Martin SE, Guha R, Ormanoglu P, Pfister TD, Reaper PM, Barnes CS, Jones J, Charlton P, Pollard JR, Morris J, Doroshow JH, Pommier Y. ATR inhibitors VE-821 and VX-970 sensitize cancer cells to topoisomerase i inhibitors by disabling DNA replication initiation and fork elongation responses. Cancer Res 2014; 74:6968-79. [PMID: 25269479 PMCID: PMC4252598 DOI: 10.1158/0008-5472.can-13-3369] [Citation(s) in RCA: 118] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Camptothecin and its derivatives, topotecan and irinotecan, are specific topoisomerase I (Top1) inhibitors and potent anticancer drugs killing cancer cells by producing replication-associated DNA double-strand breaks, and the indenoisoquinoline LMP-400 (indotecan) is a novel Top1 inhibitor in clinical trial. To develop novel drug combinations, we conducted a synthetic lethal siRNA screen using a library that targets nearly 7,000 human genes. Depletion of ATR, the main transducer of replication stress, came as a top candidate gene for camptothecin synthetic lethality. Validation studies using ATR siRNA and the ATR inhibitor VE-821 confirmed marked antiproliferative synergy with camptothecin and even greater synergy with LMP-400. Single-cell analyses and DNA fiber combing assays showed that VE-821 abrogates the S-phase replication elongation checkpoint and the replication origin-firing checkpoint induced by camptothecin and LMP-400. As expected, the combination of Top1 inhibitors with VE-821 inhibited the phosphorylation of ATR and Chk1; however, it strongly induced γH2AX. In cells treated with the combination, the γH2AX pattern changed over time from the well-defined Top1-induced damage foci to an intense peripheral and diffuse nuclear staining, which could be used as response biomarker. Finally, the clinical derivative of VE-821, VX-970, enhanced the in vivo tumor response to irinotecan without additional toxicity. A key implication of our work is the mechanistic rationale and proof of principle it provides to evaluate the combination of Top1 inhibitors with ATR inhibitors in clinical trials.
Collapse
Affiliation(s)
- Rozenn Jossé
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland
| | - Scott E Martin
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, Maryland
| | - Rajarshi Guha
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, Maryland
| | - Pinar Ormanoglu
- Division of Pre-Clinical Innovation, National Center for Advancing Translational Sciences (NCATS), NIH, Rockville, Maryland
| | - Thomas D Pfister
- Laboratory of Human Toxicology and Pharmacology, Applied/Developmental Research Directorate, Leidos Biomedical Research, Inc, National Laboratory for Cancer Research, Frederick, Maryland
| | - Philip M Reaper
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, Oxfordshire, United Kingdom
| | | | - Julie Jones
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, Oxfordshire, United Kingdom
| | - Peter Charlton
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, Oxfordshire, United Kingdom
| | - John R Pollard
- Vertex Pharmaceuticals (Europe) Ltd, Abingdon, Oxfordshire, United Kingdom
| | - Joel Morris
- Drug Synthesis and Chemistry Branch, Division of Cancer Treatment, Division of Cancer Treatment and Diagnosis (DTP-DCTD), NCI-NIH, Bethesda, Maryland
| | - James H Doroshow
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland. Drug Synthesis and Chemistry Branch, Division of Cancer Treatment, Division of Cancer Treatment and Diagnosis (DTP-DCTD), NCI-NIH, Bethesda, Maryland
| | - Yves Pommier
- Developmental Therapeutics Branch and Laboratory of Molecular Pharmacology, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland.
| |
Collapse
|
45
|
The chromatin remodeling factor CHD5 is a transcriptional repressor of WEE1. PLoS One 2014; 9:e108066. [PMID: 25247294 PMCID: PMC4172601 DOI: 10.1371/journal.pone.0108066] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2014] [Accepted: 08/26/2014] [Indexed: 11/19/2022] Open
Abstract
Loss of the chromatin remodeling ATPase CHD5 has been linked to the progression of neuroblastoma tumors, yet the underlying mechanisms behind the tumor suppressor role of CHD5 are unknown. In this study, we purified the human CHD5 complex and found that CHD5 is a component of the full NuRD transcriptional repressor complex, which also contains methyl-CpG binding proteins and histone deacetylases. The CHD5/NuRD complex appears mutually exclusive with the related CHD4/NuRD complex as overexpression of CHD5 results in loss of the CHD4 protein in cells. Following a search for genes that are regulated by CHD5 in neuroblastoma cells, we found that CHD5 binds to and represses the G2/M checkpoint gene WEE1. Reintroduction of CHD5 into neuroblastoma cells represses WEE1 expression, demonstrating that CHD5 can function as a repressor in cells. A catalytically inactive mutant version of CHD5 is able to associate with a NuRD cofactor but fails to repress transcription. Our study shows that CHD5 is a NuRD-associated transcriptional repressor and identifies WEE1 as one of the CHD5-regulated genes that may link CHD5 to tumor suppression.
Collapse
|
46
|
Matthews LM, Evans JP. α-endosulfine (ENSA) regulates exit from prophase I arrest in mouse oocytes. Cell Cycle 2014; 13:1639-49. [PMID: 24675883 DOI: 10.4161/cc.28606] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Mammalian oocytes in ovarian follicles are arrested in meiosis at prophase I. This arrest is maintained until ovulation, upon which the oocyte exits from this arrest, progresses through meiosis I and to metaphase of meiosis II. The progression from prophase I to metaphase II, known as meiotic maturation, is mediated by signals that coordinate these transitions in the life of the oocyte. ENSA (α-endosulfine) and ARPP19 (cAMP-regulated phosphoprotein-19) have emerged as regulators of M-phase, with function in inhibition of protein phosphatase 2A (PP2A) activity. Inhibition of PP2A maintains the phosphorylated state of CDK1 substrates, thus allowing progression into and/or maintenance of an M-phase state. We show here ENSA in mouse oocytes plays a key role in the progression from prophase I arrest into M-phase of meiosis I. The majority of ENSA-deficient oocytes fail to exit from prophase I arrest. This function of ENSA in oocytes is dependent on PP2A, and specifically on the regulatory subunit PPP2R2D (also known as B55δ). Treatment of ENSA-deficient oocytes with Okadaic acid to inhibit PP2A rescues the defect in meiotic progression, with Okadaic acid-treated, ENSA-deficient oocytes being able to exit from prophase I arrest. Similarly, oocytes deficient in both ENSA and PPP2R2D are able to exit from prophase I arrest to an extent similar to wild-type oocytes. These data are evidence of a role for ENSA in regulating meiotic maturation in mammalian oocytes, and also have potential relevance to human oocyte biology, as mouse and human have genes encoding both Arpp19 and Ensa.
Collapse
Affiliation(s)
- Lauren M Matthews
- Department of Biochemistry and Molecular Biology; Bloomberg School of Public Health; Johns Hopkins University; Baltimore, MD USA
| | - Janice P Evans
- Department of Biochemistry and Molecular Biology; Bloomberg School of Public Health; Johns Hopkins University; Baltimore, MD USA
| |
Collapse
|