1
|
Luo H, Huang K, Cheng M, Long X, Zhu X, Wu M. The HNF4A-CHPF pathway promotes proliferation and invasion through interactions with MAD1L1 in glioma. Aging (Albany NY) 2023; 15:11052-11066. [PMID: 37851364 PMCID: PMC10637790 DOI: 10.18632/aging.205076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/22/2023] [Indexed: 10/19/2023]
Abstract
Chondroitin polymerizing factor (CHPF) is an important glycosyltransferases that participates in the biosynthesis of chondroitin sulfate (CS). Our previous study showed that silencing CHPF expression inhibited glioma cell proliferation in vitro, but the molecular mechanisms by which CHPF contributes to development of glioma have not been characterized. In this study, we found that CHPF was up-regulated in glioma tissues and was positively correlated with malignant clinical pathological characteristics of patients with glioma. Silencing CHPF expression inhibited proliferation, colony formation, migration, and cell cycle of glioma cells. Moreover, silencing CHPF suppressed glioma malignance in vivo. Immunoprecipitation, co-immunoprecipitation, GST pulldown, and liquid chromatography-mass spectrometry (LC-MS/MS) assays were used to verify the interaction between CHPF and Mitotic arrest deficient 1-like 1 (MAD1L1). In addition, Chromatin Immunoprecipitation (ChIP)-PCR analysis showed that HNF4A bound to the CHPF promoter region, which indicated that the transcription factor hepatocyte nuclear factor 4A (HNF4A) could regulate the expression of CHPF in glioma cells.
Collapse
Affiliation(s)
- Haitao Luo
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi Province, China
| | - Kai Huang
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi Province, China
| | - Mengqi Cheng
- Department of Health Management Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaoyan Long
- Science Research Center, East China Institute of Digital Medical Engineering, Shangrao, Jiangxi Province, China
| | - Xingen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi Province, China
| | - Miaojing Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
- Institute of Neuroscience, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
2
|
Chan Y, Liu Y, Kong Y, Xu W, Zeng X, Li H, Guo Y, Tang X, Zhang J, Zhu B. Maternal genetic polymorphisms in the major mitotic checkpoint genes MAD1L1 and MAD2L1 associated with the risk of survival in abnormal chromosomal fetuses. Front Genet 2023; 14:1105184. [PMID: 37007941 PMCID: PMC10061030 DOI: 10.3389/fgene.2023.1105184] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Accepted: 02/27/2023] [Indexed: 03/18/2023] Open
Abstract
Background: The genetic etiology of fetal chromosome abnormalities remains unknown, which brings about an enormous burden for patients, families, and society. The spindle assembly checkpoint (SAC) controls the normal procedure of chromosome disjunction and may take part in the process.Objective: The aim of this study was to explore the association between polymorphisms of MAD1L1 rs1801368 and MAD2L1 rs1283639804, involved in SAC and fetal chromosome abnormalities.Methods: The case–control study collected 563 cases and 813 health controls to test the genotypes of MAD1L1 rs1801368 and MAD2L1 rs1283639804 polymorphisms by polymerase chain reaction–restrictive fragment length polymorphism methods (PCR-RFLP).Results:MAD1L1 rs1801368 polymorphism was associated with fetal chromosome abnormalities alone or combined to lower homocysteine (HCY) levels (alone: dominant: OR: 1.75, 95%CI: 1.19–2.57, and p = 0.005; CT vs. CC: OR = 0.73, 95%CI: 0.57–0.94, and p = 0.016; lower HCY: C vs. T: OR = 0.74, 95%CI: 0.57–0.95, and p = 0.02; dominant: OR = 1.75, 95%CI: 0.79–1.92, and p = 0.005). No significant differences were found in other genetic models or subgroups (p > 0.05, respectively). MAD2L1 rs1283639804 polymorphism revealed a sole genotype in the studied population. HCY is significantly associated with fetal chromosome abnormalities in younger groups (OR: 1.78, 95%CI: 1.28–2.47, and p = 0.001).Conclusion: The results implied that the polymorphism of MAD1L1 rs1801368 may become the susceptibility factor to fetal chromosome abnormalities alone or combined to lower HCY levels but not to MAD2L1 rs1283639804 polymorphism. In addition, HCY significantly affects fetal chromosomal abnormalities in younger women.
Collapse
Affiliation(s)
- Ying Chan
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yize Liu
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Yamin Kong
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Weiming Xu
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xiaohong Zeng
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Haichun Li
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Yan Guo
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Xinhua Tang
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Jinman Zhang
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Baosheng Zhu
- Department of Medical Genetics, NHC Key Laboratory of Preconception Health Birth in Western China, Yunnan Provincial Key Laboratory for Birth Defects and Genetic Diseases, First People’s Hospital of Yunnan Province, Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
- School of Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- *Correspondence: Baosheng Zhu,
| |
Collapse
|
3
|
Mohapatra S, Winkle M, Ton AN, Nguyen D, Calin GA. The Role of Non-Coding RNAs in Chromosomal Instability in Cancer. J Pharmacol Exp Ther 2023; 384:10-19. [PMID: 36167417 PMCID: PMC9827503 DOI: 10.1124/jpet.122.001357] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/22/2022] [Accepted: 08/17/2022] [Indexed: 01/12/2023] Open
Abstract
Chromosomal instability (CIN) is characterized by an increased frequency of changes in chromosome structure or number and is regarded as a hallmark of cancer. CIN plays a prevalent role in tumorigenesis and cancer progression by assisting the cancer cells' phenotypic adaptation to stress, which have been tightly linked to therapy resistance and metastasis. Both CIN-inducing and CIN-repressing agents are being clinically tested for the treatment of cancer to increase CIN levels to unsustainable levels leading to cell death or to decrease CIN levels to limit the development of drug resistance, respectively. Non-coding RNAs (ncRNAs) including microRNAs and long ncRNAs (lncRNAs) have been fundamentally implicated in CIN. The miR-22, miR-26a, miR-28, and miR-186 target important checkpoint proteins involved in mediating chromosomal stability and their expression modulation has been directly related to CIN occurrence. lncRNAs derived from telomeric, centrosomal, and enhancer regions play an important role in mediating genome stability, while specific lncRNA transcripts including genomic instability inducing RNA called Ginir, P53-responsive lncRNA termed as GUARDIN, colon cancer-associated transcript 2, PCAT2, and ncRNA activated by DNA damage called NORAD have been shown to act within CIN-associated pathways. In this review, we discuss how these ncRNAs either maintain or disrupt the stability of chromosomes and how these mechanisms could be exploited for novel therapeutic approaches targeting CIN in cancer patients. SIGNIFICANCE STATEMENT: Chromosomal instability increases tumor heterogeneity and thereby assists the phenotypic adaptation of cancer cells, causing therapy resistance and metastasis. Several microRNAs and long non-coding RNAs that have been causally linked to chromosomal instability could represent novel therapeutic targets. Understanding the role of non-coding RNAs in regulating different genes involved in driving chromosomal instability will give insights into how non-coding RNAs can be utilized toward modifying chemotherapeutic regimens in different cancers.
Collapse
Affiliation(s)
- Swati Mohapatra
- Department of Translational Molecular Pathology (S.M., M.W., A.N.T., G.A.C.), UT Health Graduate School of Biomedical Sciences (S.M.), Program in Molecular Genetic Technology, School of Health Professions (A.N.T.), and Center for RNA Interference and Non-Coding RNAs (G.A.C.), The University of Texas MD Anderson Cancer Center, Houston, Texas; and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (D.N.)
| | - Melanie Winkle
- Department of Translational Molecular Pathology (S.M., M.W., A.N.T., G.A.C.), UT Health Graduate School of Biomedical Sciences (S.M.), Program in Molecular Genetic Technology, School of Health Professions (A.N.T.), and Center for RNA Interference and Non-Coding RNAs (G.A.C.), The University of Texas MD Anderson Cancer Center, Houston, Texas; and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (D.N.)
| | - Anh N Ton
- Department of Translational Molecular Pathology (S.M., M.W., A.N.T., G.A.C.), UT Health Graduate School of Biomedical Sciences (S.M.), Program in Molecular Genetic Technology, School of Health Professions (A.N.T.), and Center for RNA Interference and Non-Coding RNAs (G.A.C.), The University of Texas MD Anderson Cancer Center, Houston, Texas; and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (D.N.)
| | - Dien Nguyen
- Department of Translational Molecular Pathology (S.M., M.W., A.N.T., G.A.C.), UT Health Graduate School of Biomedical Sciences (S.M.), Program in Molecular Genetic Technology, School of Health Professions (A.N.T.), and Center for RNA Interference and Non-Coding RNAs (G.A.C.), The University of Texas MD Anderson Cancer Center, Houston, Texas; and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (D.N.)
| | - George A Calin
- Department of Translational Molecular Pathology (S.M., M.W., A.N.T., G.A.C.), UT Health Graduate School of Biomedical Sciences (S.M.), Program in Molecular Genetic Technology, School of Health Professions (A.N.T.), and Center for RNA Interference and Non-Coding RNAs (G.A.C.), The University of Texas MD Anderson Cancer Center, Houston, Texas; and Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts (D.N.)
| |
Collapse
|
4
|
Esposito E, Weidemann DE, Rogers JM, Morton CM, Baybay EK, Chen J, Hauf S. Mitotic checkpoint gene expression is tuned by codon usage bias. EMBO J 2022; 41:e107896. [PMID: 35811551 PMCID: PMC9340482 DOI: 10.15252/embj.2021107896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 05/30/2022] [Accepted: 06/06/2022] [Indexed: 11/09/2022] Open
Abstract
The mitotic checkpoint (also called spindle assembly checkpoint, SAC) is a signaling pathway that safeguards proper chromosome segregation. Correct functioning of the SAC depends on adequate protein concentrations and appropriate stoichiometries between SAC proteins. Yet very little is known about the regulation of SAC gene expression. Here, we show in the fission yeast Schizosaccharomyces pombe that a combination of short mRNA half-lives and long protein half-lives supports stable SAC protein levels. For the SAC genes mad2+ and mad3+ , their short mRNA half-lives are caused, in part, by a high frequency of nonoptimal codons. In contrast, mad1+ mRNA has a short half-life despite a higher frequency of optimal codons, and despite the lack of known RNA-destabilizing motifs. Hence, different SAC genes employ different strategies of expression. We further show that Mad1 homodimers form co-translationally, which may necessitate a certain codon usage pattern. Taken together, we propose that the codon usage of SAC genes is fine-tuned to ensure proper SAC function. Our work shines light on gene expression features that promote spindle assembly checkpoint function and suggests that synonymous mutations may weaken the checkpoint.
Collapse
Affiliation(s)
- Eric Esposito
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Douglas E Weidemann
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Jessie M Rogers
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Claire M Morton
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Erod Keaton Baybay
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Jing Chen
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| | - Silke Hauf
- Department of Biological SciencesVirginia TechBlacksburgVAUSA
- Fralin Life Sciences InstituteVirginia TechBlacksburgVAUSA
| |
Collapse
|
5
|
Wang Y, Yu T, Han Y, He Y, Song Y, Guo L, An L, Yang C, Wang F. Phosphorylation of MAD2 at Ser195 Promotes Spindle Checkpoint Defects and Sensitizes Cancer Cells to Radiotherapy in ATM Deficient Cells. Front Cell Dev Biol 2022; 10:817831. [PMID: 35309941 PMCID: PMC8924061 DOI: 10.3389/fcell.2022.817831] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 01/18/2022] [Indexed: 11/13/2022] Open
Abstract
The spindle assembly checkpoint (SAC) is a critical monitoring device in mitosis for the maintenance of genomic stability. Specifically, the SAC complex comprises several factors, including Mad1, Mad2, and Bub1. Ataxia-telangiectasia mutated (ATM) kinase, the crucial regulator in DNA damage response (DDR), also plays a critical role in mitosis by regulating Mad1 dimerization and SAC. Here, we further demonstrated that ATM negatively regulates the phosphorylation of Mad2, another critical component of the SAC, which is also involved in DDR. Mechanistically, we found that phosphorylation of Mad2 is aberrantly increased in ATM-deficient cells. Point-mutation analysis further revealed that Serine 195 mainly mediated Mad2 phosphorylation upon ATM ablation. Functionally, the phosphorylation of Mad2 causes decreased DNA damage repair capacity and is related to the resistance to cancer cell radiotherapy. Altogether, this study unveils the key regulatory role of Mad2 phosphorylation in checkpoint defects and DNA damage repair in ATM-deficient cells.
Collapse
Affiliation(s)
- Yang Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Tianyu Yu
- Department of General Surgery, Pudong New Area Gongli Hospital Affiliated to Naval Military Medical University, Naval Military Medical University, Shanghai, China
| | - Yi Han
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yazhi He
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yiran Song
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Leiming Guo
- Department of R&D, Shanghai Creative Immune Therapeutics Co., Ltd, Shanghai, China
| | - Liwei An
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Chunying Yang
- Central Laboratory, Shanghai Putuo District People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Feng Wang
- Department of Gastroenterology, Shanghai 10th People's Hospital, Tongji University School of Medicine, Shanghai, China.,Central Laboratory, Shanghai Putuo District People's Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
6
|
Dishman AF, Peterson FC, Volkman BF. Specific binding-induced modulation of the XCL1 metamorphic equilibrium. Biopolymers 2021; 112:e23402. [PMID: 32986858 PMCID: PMC8004533 DOI: 10.1002/bip.23402] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Revised: 08/27/2020] [Accepted: 09/10/2020] [Indexed: 01/25/2023]
Abstract
The metamorphic protein XCL1 switches between two distinct native structures with different functions in the human immune system. This structural interconversion requires complete rearrangement of all hydrogen bonding networks, yet fold-switching occurs spontaneously and reversibly in solution. One structure occupies the canonical α-β chemokine fold and binds XCL1's cognate G-protein coupled receptor, while the other structure occupies a dimeric, all-β fold that binds glycosaminoglycans and has antimicrobial activity. Both of these functions are important for the biologic role of XCL1 in the immune system, and each structure is approximately equally populated under near-physiologic conditions. Recent work has begun to illuminate XCL1's role in combatting infection and cancer. However, without a way to control XCL1's dynamic structural interconversion, it is difficult to study the role of XCL1 fold-switching in human health and disease. Thus, a molecular tool that can regulate the fractional population of the two XCL1 structures is needed. Here, we find by heparin affinity chromatography and NMR that an engineered XCL1 variant called CC5 can trigger a dose-dependent shift in XCL1's metamorphic equilibrium such that the receptor binding structure is depleted, and the antimicrobial structure is more heavily populated. This shift likely occurs due to formation of XCL1-CC5 heterodimers in which both protomers occupy the β-sheet structure. These findings lay the groundwork for future studies seeking to understand the functional role of XCL1 metamorphosis, as well as studies screening for a drug-like molecule that can therapeutically target XCL1 by tuning its metamorphic equilibrium. Moreover, the proof of concept presented here suggests that protein metamorphosis is druggable, opening numerous avenues for controlling biological function of metamorphic proteins by altering the population of their multiple native states.
Collapse
Affiliation(s)
- Acacia F. Dishman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
- Medical Scientist Training Program, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Francis C. Peterson
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| | - Brian F. Volkman
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, Wisconsin, USA
| |
Collapse
|
7
|
Teye EK, Lu S, Chen F, Yang W, Abraham T, Stairs DB, Wang HG, Yochum GS, Brodsky RA, Pu JJ. PIGN spatiotemporally regulates the spindle assembly checkpoint proteins in leukemia transformation and progression. Sci Rep 2021; 11:19022. [PMID: 34561473 PMCID: PMC8463542 DOI: 10.1038/s41598-021-98218-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 09/06/2021] [Indexed: 12/19/2022] Open
Abstract
Phosphatidylinositol glycan anchor biosynthesis class N (PIGN) has been linked to the suppression of chromosomal instability. The spindle assembly checkpoint complex is responsible for proper chromosome segregation during mitosis to prevent chromosomal instability. In this study, the novel role of PIGN as a regulator of the spindle assembly checkpoint was unveiled in leukemic patient cells and cell lines. Transient downregulation or ablation of PIGN resulted in impaired mitotic checkpoint activation due to the dysregulated expression of spindle assembly checkpoint-related proteins including MAD1, MAD2, BUBR1, and MPS1. Moreover, ectopic overexpression of PIGN restored the expression of MAD2. PIGN regulated the spindle assembly checkpoint by forming a complex with the spindle assembly checkpoint proteins MAD1, MAD2, and the mitotic kinase MPS1. Thus, PIGN could play a vital role in the spindle assembly checkpoint to suppress chromosomal instability associated with leukemic transformation and progression.
Collapse
Affiliation(s)
- Emmanuel K Teye
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Shasha Lu
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fangyuan Chen
- Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wenrui Yang
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA.,Institute of Hematology, Peking Union Medical College, Tianjin, China
| | - Thomas Abraham
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Douglas B Stairs
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Hong-Gang Wang
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Gregory S Yochum
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Robert A Brodsky
- Division of Hematology, Johns Hopkins Medicine, Baltimore, MD, USA
| | - Jeffrey J Pu
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA, USA. .,University of Arizona Cancer Center, 1515 N Campbell Avenue, #1968C, Tucson, AZ, 85724, USA.
| |
Collapse
|
8
|
Genetics of autosomal mosaic chromosomal alteration (mCA). J Hum Genet 2021; 66:879-885. [PMID: 34321609 DOI: 10.1038/s10038-021-00964-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 07/14/2021] [Accepted: 07/15/2021] [Indexed: 12/11/2022]
Abstract
Mosaic chromosomal alterations (mCAs) are frequently observed in cancer cells and are regarded as one of the common features of cancers. Strikingly, accumulating studies demonstrated that mCAs are also prevalent in elderly individuals without cancer, implying mCA could be a feature of aging and not necessarily a cancerous state. However, the genetic basis of mCA has been mostly unknown. Recent studies of autosomal mCA based on biobank-scale datasets, including UK Biobank and Biobank Japan, provided a glimpse into the underlying genetic mechanism. In this concise review, we briefly introduced mCA, its link with cancer and aging, and the emerging genetic mechanisms of this phenomenon. We highlighted the following aspects: (1) the interplay between somatic and inherited germline mutations in generating mosaicism; (2) monogenic and polygenic architectures of mCA; and (3) population-specific profiles of mCA. We provided a future perspective emphasizing the need to understand the connection between mCA and other characteristics of aging, in particular, the epigenetic and immunologic features.
Collapse
|
9
|
Schuyler SC, Chen HY. Using Budding Yeast to Identify Molecules That Block Cancer Cell 'Mitotic Slippage' Only in the Presence of Mitotic Poisons. Int J Mol Sci 2021; 22:ijms22157985. [PMID: 34360748 PMCID: PMC8347345 DOI: 10.3390/ijms22157985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/20/2021] [Accepted: 07/23/2021] [Indexed: 11/16/2022] Open
Abstract
Research on the budding yeast Saccharomyces cerevisiae has yielded fundamental discoveries on highly conserved biological pathways and yeast remains the best-studied eukaryotic cell in the world. Studies on the mitotic cell cycle and the discovery of cell cycle checkpoints in budding yeast has led to a detailed, although incomplete, understanding of eukaryotic cell cycle progression. In multicellular eukaryotic organisms, uncontrolled aberrant cell division is the defining feature of cancer. Some of the most successful classes of anti-cancer chemotherapeutic agents are mitotic poisons. Mitotic poisons are thought to function by inducing a mitotic spindle checkpoint-dependent cell cycle arrest, via the assembly of the highly conserved mitotic checkpoint complex (MCC), leading to apoptosis. Even in the presence of mitotic poisons, some cancer cells continue cell division via 'mitotic slippage', which may correlate with a cancer becoming refractory to mitotic poison chemotherapeutic treatments. In this review, knowledge about budding yeast cell cycle control is explored to suggest novel potential drug targets, namely, specific regions in the highly conserved anaphase-promoting complex/cyclosome (APC/C) subunits Apc1 and/or Apc5, and in a specific N-terminal region in the APC/C co-factor cell division cycle 20 (Cdc20), which may yield molecules which block 'mitotic slippage' only in the presence of mitotic poisons.
Collapse
Affiliation(s)
- Scott C. Schuyler
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan;
- Division of Head and Neck Surgery, Department of Otolaryngology, Chang Gung Memorial Hospital, Kwei-Shan, Taoyuan 333, Taiwan
- Correspondence: ; Tel.: +886-03-211-8800 (ext. 3596)
| | - Hsin-Yu Chen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Taoyuan 333, Taiwan;
| |
Collapse
|
10
|
Schuyler SC, Wang LI, Ding YS, Lee YC, Chen HY. Deletion of Budding Yeast MAD2 Suppresses Clone-to-Clone Differences in Artificial Linear Chromosome Copy Numbers and Gives Rise to Higher Retention Rates. Microorganisms 2020; 8:microorganisms8101495. [PMID: 33003307 PMCID: PMC7599710 DOI: 10.3390/microorganisms8101495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 09/22/2020] [Accepted: 09/25/2020] [Indexed: 11/16/2022] Open
Abstract
Our goal was to investigate the changes in artificial short-linear chromosome average copy numbers per cell arising from partial or full loss of Mitotic Arrest-Deficient 2 (MAD2) spindle checkpoint function in budding yeast Saccharomyces cerevisiae. Average artificial linear chromosome copy numbers in a population of cells, as measured by quantitative polymerase chain reactions (qPCR), and retention rates, as measured by fluctuation analyses, were performed on a total of 62 individual wild type and mad2∆ mutant haploid and diploid clones. Wild type cells, both haploids and diploids, displayed phenotypically unique clone-to-clone differences: one group of 15 clones displayed low-copy numbers per cell and high retention rates, were 1 clone was found to have undergone a genomic integration event, and the second group of 15 clones displayed high copy numbers per cell and low retention rates, with the latter values being consistent with the previously published results where only a single clone had been measured. These chromosome states were observed to be unstable when propagated for 10 days under selection, where high copy-low retention rate clones evolved into low copy-high retention rate clones, but no evidence for integration events was observed. By contrast, mad2∆ haploid and mad2∆/mad2∆ diploids displayed a suppression of the clone-to-clone differences, where 20 out of 21 clones had mid-level artificial linear chromosome copy numbers per cell, but maintained elevated chromosome retention rates. The elevated levels in retention rates in mad2∆ and mad2∆/mad2∆ cells were also maintained even in the absence of selection during growth over 3 days. MAD2/mad2∆ heterozygous diploids displayed multiple clonal groups: 4 with low copy numbers, 5 with mid-level copy numbers, and 1 with a high copy number of artificial linear chromosomes, but all 10 clones uniformly displayed low retention rates. Our observations reveal that MAD2 function contributes to the ability of yeast cells to maintain a high number of artificial linear chromosomes per cell in some clones, but, counter-intuitively, mad2∆ suppresses clone-to-clone differences and leads to an improvement in artificial linear chromosome retention rates yielding a more uniform and stable clonal population with mid-level chromosome copy numbers per cell.
Collapse
Affiliation(s)
- Scott C. Schuyler
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan; (L.-I.W.); (Y.-S.D.); (Y.-C.L.); (H.-Y.C.)
- Division of Head and Neck Surgery, Department of Otolaryngology, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan 333, Taiwan
- Correspondence: ; Tel.: +886-3-211-8800-x3596
| | - Lin-Ing Wang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan; (L.-I.W.); (Y.-S.D.); (Y.-C.L.); (H.-Y.C.)
| | - Yi-Shan Ding
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan; (L.-I.W.); (Y.-S.D.); (Y.-C.L.); (H.-Y.C.)
| | - Yi-Chieh Lee
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan; (L.-I.W.); (Y.-S.D.); (Y.-C.L.); (H.-Y.C.)
| | - Hsin-Yu Chen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan 333, Taiwan; (L.-I.W.); (Y.-S.D.); (Y.-C.L.); (H.-Y.C.)
| |
Collapse
|
11
|
Voutsadakis IA. Clinical Implications of Chromosomal Instability (CIN) and Kinetochore Abnormalities in Breast Cancers. Mol Diagn Ther 2020; 23:707-721. [PMID: 31372940 DOI: 10.1007/s40291-019-00420-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Genetic instability is a defining property of cancer cells and is the basis of various lesions including point mutations, copy number alterations and translocations. Chromosomal instability (CIN) is part of the genetic instability of cancer and consists of copy number alterations in whole or parts of cancer cell chromosomes. CIN is observed in differing degrees in most cancers. In breast cancer, CIN is commonly part of the genomic landscape of the disease and has a higher incidence in aggressive sub-types. Tumor suppressors that are commonly mutated or disabled in cancer, such as p53 and pRB, play roles in protection against CIN, and as a result, their dysfunction contributes to the establishment or tolerance of CIN. Several structural and regulatory proteins of the centromeres and kinetochore, the complex structure that is responsible for the correct distribution of genetic material in the daughter cells during mitosis, are direct or, mostly, indirect transcription targets of p53 and pRB. Thus, despite the absence of structural defects in genes encoding for centromere and kinetochore components, dysfunction of these tumor suppressors may have profound implications for the correct function of the mitotic apparatus contributing to CIN. CIN and its prognostic and therapeutic implications in breast cancer are discussed in this article.
Collapse
Affiliation(s)
- Ioannis A Voutsadakis
- Algoma District Cancer Program, Sault Area Hospital, 750 Great Northern Road, Sault Ste Marie, ON, P6B 0A8, Canada. .,Section of Internal Medicine, Division of Clinical Sciences, Northern Ontario School of Medicine, Sudbury, ON, Canada.
| |
Collapse
|
12
|
Sun Z, Lu J, Wu M, Li M, Bai L, Shi Z, Hao L, Wu Y. Deficiency of PTEN leads to aberrant chromosome segregation through downregulation of MAD2. Mol Med Rep 2019; 20:4235-4243. [PMID: 31545428 PMCID: PMC6797992 DOI: 10.3892/mmr.2019.10668] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 08/20/2019] [Indexed: 11/07/2022] Open
Abstract
Proper spindle formation and accurate chromosome segregation are essential for ensuring mitotic fidelity. Phosphatase and tensin homolog (PTEN) is a multifunctional protein, which is able to maintain the stability of the genome and chromosomes. The present study described an essential role of PTEN in regulating chromosome segregation to prevent gross genomic instability via regulation of mitotic arrest deficient 2 (MAD2). PTEN knockdown induced cell cycle arrest and abnormal chromosome segregation, which manifested as the formation of anaphase bridges, lagging chromosomes and premature chromatid separation. In addition, MAD2 was identified as a potential target of PTEN. Furthermore, the present study revealed that PTEN knockdown resulted in MAD2 degradation via the ubiquitin-proteasomal pathway, while restoration of MAD2 expression partially ameliorated the mitotic defects induced by PTEN loss. The results from the present study proposed a novel mechanism by which PTEN maintains chromosome stability.
Collapse
Affiliation(s)
- Zhuo Sun
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Jinqi Lu
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Muyu Wu
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Mingyan Li
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Lu Bai
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| | - Zhenduo Shi
- Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Lin Hao
- Department of Urology, Xuzhou Central Hospital, Xuzhou, Jiangsu 221009, P.R. China
| | - Yongping Wu
- Department of Pathology, Laboratory of Clinical and Experimental Pathology, Xuzhou Medical University, Xuzhou, Jiangsu 221004, P.R. China
| |
Collapse
|
13
|
Zhou Q, Pham KTM, Hu H, Kurasawa Y, Li Z. A kinetochore-based ATM/ATR-independent DNA damage checkpoint maintains genomic integrity in trypanosomes. Nucleic Acids Res 2019; 47:7973-7988. [PMID: 31147720 PMCID: PMC6736141 DOI: 10.1093/nar/gkz476] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/23/2019] [Accepted: 05/17/2019] [Indexed: 02/02/2023] Open
Abstract
DNA damage-induced cell cycle checkpoints serve as surveillance mechanisms to maintain genomic stability, and are regulated by ATM/ATR-mediated signaling pathways that are conserved from yeast to humans. Trypanosoma brucei, an early divergent microbial eukaryote, lacks key components of the conventional DNA damage-induced G2/M cell cycle checkpoint and the spindle assembly checkpoint, and nothing is known about how T. brucei controls its cell cycle checkpoints. Here we discover a kinetochore-based, DNA damage-induced metaphase checkpoint in T. brucei. MMS-induced DNA damage triggers a metaphase arrest by modulating the abundance of the outer kinetochore protein KKIP5 in an Aurora B kinase- and kinetochore-dependent, but ATM/ATR-independent manner. Overexpression of KKIP5 arrests cells at metaphase through stabilizing the mitotic cyclin CYC6 and the cohesin subunit SCC1, mimicking DNA damage-induced metaphase arrest, whereas depletion of KKIP5 alleviates the DNA damage-induced metaphase arrest and causes chromosome mis-segregation and aneuploidy. These findings suggest that trypanosomes employ a novel DNA damage-induced metaphase checkpoint to maintain genomic integrity.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, TX 77030, USA
| | - Kieu T M Pham
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, TX 77030, USA
| | - Huiqing Hu
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, TX 77030, USA
| | - Yasuhiro Kurasawa
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, TX 77030, USA
| | - Ziyin Li
- Department of Microbiology and Molecular Genetics, McGovern Medical School, University of Texas Health Science Center at Houston, TX 77030, USA
| |
Collapse
|
14
|
Gay S, Piccini D, Bruhn C, Ricciardi S, Soffientini P, Carotenuto W, Biffo S, Foiani M. A Mad2-Mediated Translational Regulatory Mechanism Promoting S-Phase Cyclin Synthesis Controls Origin Firing and Survival to Replication Stress. Mol Cell 2019; 70:628-638.e5. [PMID: 29775579 PMCID: PMC5972228 DOI: 10.1016/j.molcel.2018.04.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 02/26/2018] [Accepted: 04/23/2018] [Indexed: 11/03/2022]
Abstract
Cell survival to replication stress depends on the activation of the Mec1ATR-Rad53 checkpoint response that protects the integrity of stalled forks and controls the origin firing program. Here we found that Mad2, a member of the spindle assembly checkpoint (SAC), contributes to efficient origin firing and to cell survival in response to replication stress. We show that Rad53 and Mad2 promote S-phase cyclin expression through different mechanisms: while Rad53 influences Clb5,6 degradation, Mad2 promotes their protein synthesis. We found that Mad2 co-sediments with polysomes and modulates the association of the translation inhibitor Caf204E-BP with the translation machinery and the initiation factor eIF4E. This Mad2-dependent translational regulatory process does not depend on other SAC proteins. Altogether our observations indicate that Mad2 has an additional function outside of mitosis to control DNA synthesis and collaborates with the Mec1-Rad53 regulatory axis to allow cell survival in response to replication stress.
Collapse
Affiliation(s)
- Sophie Gay
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy.
| | - Daniele Piccini
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Christopher Bruhn
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Sara Ricciardi
- Fondazione Istituto Nazionale Genetica Molecolare, Via Francesco Sforza, 32, 20122 Milan, Italy; Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy
| | - Paolo Soffientini
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Walter Carotenuto
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Stefano Biffo
- Fondazione Istituto Nazionale Genetica Molecolare, Via Francesco Sforza, 32, 20122 Milan, Italy; Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy
| | - Marco Foiani
- IFOM, The FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy; Università degli Studi di Milano, Via Celoria 26, 20133 Milan, Italy.
| |
Collapse
|
15
|
Schuyler SC, Wu YFO, Chen HY, Ding YS, Lin CJ, Chu YT, Chen TC, Liao L, Tsai WW, Huang A, Wang LI, Liao TW, Jhuo JH, Cheng V. Peptide inhibitors of the anaphase promoting-complex that cause sensitivity to microtubule poison. PLoS One 2018; 13:e0198930. [PMID: 29883473 PMCID: PMC5993284 DOI: 10.1371/journal.pone.0198930] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 05/29/2018] [Indexed: 02/01/2023] Open
Abstract
There is an interest in identifying Anaphase Promoting-Complex/Cyclosome (APC/C) inhibitors that lead to sensitivity to microtubule poisons as a strategy for targeting cancer cells. Using budding yeast Saccharomyces cerevisiae, peptides derived from the Mitotic Arrest Deficient 2 (Mad2)-binding motif of Cell Division Cycle 20 (Cdc20) were observed to inhibit both Cdc20- and CDC20 Homology 1 (Cdh1)-dependent APC/C activity. Over expression of peptides in vivo led to sensitivity to a microtubule poison and, in a recovery from a microtubule poison arrest, delayed degradation of yeast Securin protein Precocious Dissociation of Sisters 1 (Pds1). Peptides with mutations in the Cdc20 activating KILR-motif still bound APC/C, but lost the ability to inhibit APC/C in vitro and lost the ability to induce sensitivity to a microtubule poison in vivo. Thus, an APC/C binding and activation motif that promotes mitotic progression, namely the Cdc20 KILR-motif, can also function as an APC/C inhibitor when present in excess. Another activator for mitotic progression after recovery from microtubule poison is p31comet, where a yeast predicted open-reading frame YBR296C-A encoding a 39 amino acid predicted protein was identified by homology to p31comet, and named Tiny Yeast Comet 1 (TYC1). Tyc1 over expression resulted in sensitivity to microtubule poison. Tyc1 inhibited both APC/CCdc20 and APC/CCdh1 activities in vitro and bound to APC/C. A homologous peptide derived from human p31comet bound to and inhibited yeast APC/C demonstrating evolutionary retention of these biochemical activities. Cdc20 Mad2-binding motif peptides and Tyc1 disrupted the ability of the co-factors Cdc20 and Cdh1 to bind to APC/C, and co-over expression of both together in vivo resulted in an increased sensitivity to microtubule poison. We hypothesize that Cdc20 Mad2-binding motif peptides, Tyc1 and human hp31 peptide can serve as novel molecular tools for investigating APC/C inhibition that leads to sensitivity to microtubule poison in vivo.
Collapse
Affiliation(s)
- Scott C. Schuyler
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
- Division of Colorectal Surgery, Department of Surgery, Chang Gung Memorial Hospital, Kwei-Shan, Tao-Yuan, Taiwan
- * E-mail:
| | - Yueh-Fu Olivia Wu
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Hsin-Yu Chen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Yi-Shan Ding
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Chia-Jung Lin
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Yu-Ting Chu
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Ting-Chun Chen
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Louis Liao
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Wei-Wei Tsai
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Anna Huang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Lin-Ing Wang
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Ting-Wei Liao
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Jia-Hua Jhuo
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| | - Vivien Cheng
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, Taiwan
| |
Collapse
|
16
|
Bond MJ, Bleiler M, Harrison LE, Scocchera EW, Nakanishi M, G-Dayanan N, Keshipeddy S, Rosenberg DW, Wright DL, Giardina C. Spindle Assembly Disruption and Cancer Cell Apoptosis with a CLTC-Binding Compound. Mol Cancer Res 2018; 16:1361-1372. [PMID: 29769406 DOI: 10.1158/1541-7786.mcr-18-0178] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/05/2018] [Accepted: 04/25/2018] [Indexed: 11/16/2022]
Abstract
AK3 compounds are mitotic arrest agents that induce high levels of γH2AX during mitosis and apoptosis following release from arrest. We synthesized a potent AK3 derivative, AK306, that induced arrest and apoptosis of the HCT116 colon cancer cell line with an EC50 of approximately 50 nmol/L. AK306 was active on a broad spectrum of cancer cell lines with total growth inhibition values ranging from approximately 25 nmol/L to 25 μmol/L. Using biotin and BODIPY-linked derivatives of AK306, binding to clathrin heavy chain (CLTC/CHC) was observed, a protein with roles in endocytosis and mitosis. AK306 inhibited mitosis and endocytosis, while disrupting CHC cellular localization. Cells arrested in mitosis by AK306 showed the formation of multiple microtubule-organizing centers consisting of pericentrin, γ-tubulin, and Aurora A foci, without apparent centrosome amplification. Cells released from AK306 arrest were unable to form bipolar spindles, unlike nocodazole-released cells that reformed spindles and completed division. Like AK306, CHC siRNA knockdown disrupted spindle formation and activated p53. A short-term (3-day) treatment of tumor-bearing APC-mutant mice with AK306 increased apoptosis in tumors, but not normal mucosa. These findings indicate that targeting the mitotic CHC complex can selectively induce apoptosis and may have therapeutic value.Implication: Disruption of clathrin with a small-molecule inhibitor, AK306, selectively induces apoptosis in cancer cells by disrupting bipolar spindle formation. Mol Cancer Res; 16(9); 1361-72. ©2018 AACR.
Collapse
Affiliation(s)
- Michael J Bond
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut.,Department of Pharmacology, Yale University, New Haven, Connecticut
| | - Marina Bleiler
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Lauren E Harrison
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut
| | - Eric W Scocchera
- Department of Medicinal Chemistry, University of Connecticut, Storrs, Connecticut
| | - Masako Nakanishi
- Center for Molecular Oncology, UConn Health, Farmington, Connecticut
| | - Narendran G-Dayanan
- Department of Medicinal Chemistry, University of Connecticut, Storrs, Connecticut
| | - Santosh Keshipeddy
- Department of Medicinal Chemistry, University of Connecticut, Storrs, Connecticut
| | | | - Dennis L Wright
- Department of Medicinal Chemistry, University of Connecticut, Storrs, Connecticut
| | - Charles Giardina
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut.
| |
Collapse
|
17
|
Byrne T, Nelson L, Beirne JP, Sharpe D, Quinn JE, McCluggage WG, Robson T, Furlong F. BRCA1 and MAD2 Are Coexpressed and Are Prognostic Indicators in Tubo-ovarian High-Grade Serous Carcinoma. Int J Gynecol Cancer 2018; 28:472-478. [PMID: 29465507 DOI: 10.1097/igc.0000000000001214] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
OBJECTIVES The aim of this study was to investigate the relationship between BRCA1 and mitotic arrest deficiency protein 2 (MAD2) protein expression, as determined by immunohistochemistry, and clinical outcomes in epithelial ovarian carcinoma (EOC). METHODS A tissue microarray consisting of 94 formalin-fixed paraffin-embedded EOC with fully matched clinicopathological data were immunohistochemically stained with anti-BRCA1 and anti-MAD2 antibodies. The cores were scored in a semiquantitative manner evaluating nuclear staining intensity and extent. Coexpression of BRCA1 and MAD2 was evaluated, and patient survival analyses were undertaken. RESULTS Coexpression of BRCA1 and MAD2 was assessed in 94 EOC samples, and survival analysis was performed on 65 high-grade serous carcinomas (HGSCs). There was a significant positive correlation between BRCA1 and MAD2 expression in this patient cohort (P < 0.0001). Both low BRCA1 and low MAD2 are independently associated with overall survival because of HGSC. Low coexpression of BRCA1 and MAD2 was also significantly associated with overall survival and was driven by BRCA1 expression. CONCLUSION BRCA1 and MAD2 expressions are strongly correlated in EOC, but BRCA1 expression remains the stronger prognostic factor in HGSC.
Collapse
|
18
|
Guo X, Ni J, Dai X, Zhou T, Yang G, Xue J, Wang X. Biphasic regulation of spindle assembly checkpoint by low and high concentrations of resveratrol leads to the opposite effect on chromosomal instability. MUTATION RESEARCH. GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2018; 825:19-30. [PMID: 29307372 DOI: 10.1016/j.mrgentox.2017.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 10/02/2017] [Accepted: 11/07/2017] [Indexed: 01/14/2023]
Abstract
Resveratrol (RSV) is a naturally occurring polyphenolic phytoalexin possessing numerous health-promoting effects. Chromosomal instability (CIN), usually results from defective spindle assembly checkpoint (SAC), is a major contributor to many diseases. While it's recently recognized that RSV exhibits a nonlinear dose response for disease prevention, whether it's the case for its role in CIN remains unknown. Here, we investigated the potential of a broad range of RSV concentrations (0.01-100μM) on CIN and the underlying mechanisms in human normal colon epithelial NCM460 cells. CIN was measured by cytokinesis-block micronucleus assay; mitotic fidelity was determined by aberrant mitosis analysis; SAC activity was assessed by nocodazole-challenge assay, and the expression of SAC genes was examined by RT-qPCR. We found that 0.1μM RSV significantly reduced CIN (P<0.01), while 100μM RSV significantly induced it (P<0.05). Mitotic infidelity was significantly prevented by 0.1μM RSV but promoted by 100μM RSV (P<0.05 for both). Moreover, the function of SAC was sustained and impaired by 0.1μM and 100μM RSV, respectively. Several SAC genes, including Aurora-B, Aurora-C, Plk-1 and CENP-E, were significantly up-regulated and down-regulated by 0.1μM and 100μM RSV, respectively (P<0.05). In conclusion, RSV exhibited a biphasic dose-dependent effect on CIN that was exerted via the regulation of mitotic fidelity through the SAC network. The health implications of these findings were summarized.
Collapse
Affiliation(s)
- Xihan Guo
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, 650500, China; School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China
| | - Juan Ni
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, 650500, China
| | - Xueqin Dai
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, 650500, China
| | - Tao Zhou
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, 650500, China
| | - Guofang Yang
- China Gene Health Management Group, Ltd., Shanghai, 200433, China
| | - Jinglun Xue
- China Gene Health Management Group, Ltd., Shanghai, 200433, China
| | - Xu Wang
- School of Life Sciences, The Engineering Research Center of Sustainable Development and Utilization of Biomass Energy, Yunnan Normal University, Kunming, Yunnan, 650500, China; School of Life Sciences, Yunnan University, Kunming, Yunnan, 650091, China.
| |
Collapse
|
19
|
Chan SH, Ngeow J. Germline mutation contribution to chromosomal instability. Endocr Relat Cancer 2017; 24:T33-T46. [PMID: 28808044 DOI: 10.1530/erc-17-0062] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 05/18/2017] [Indexed: 12/29/2022]
Abstract
Genomic instability is a feature of cancer that fuels oncogenesis through increased frequency of genetic disruption, leading to loss of genomic integrity and promoting clonal evolution as well as tumor transformation. A form of genomic instability prevalent across cancer types is chromosomal instability, which involves karyotypic changes including chromosome copy number alterations as well as gross structural abnormalities such as transversions and translocations. Defects in cellular mechanisms that are in place to govern fidelity of chromosomal segregation, DNA repair and ultimately genomic integrity are known to contribute to chromosomal instability. In this review, we discuss the association of germline mutations in these pathways with chromosomal instability in the background of related cancer predisposition syndromes. We will also reflect on the impact of genetic predisposition to clinical management of patients and how we can exploit this vulnerability to promote catastrophic genomic instability as a therapeutic strategy.
Collapse
Affiliation(s)
- Sock Hoai Chan
- Division of Medical OncologyCancer Genetics Service, National Cancer Centre Singapore, Singapore
| | - Joanne Ngeow
- Division of Medical OncologyCancer Genetics Service, National Cancer Centre Singapore, Singapore
- Oncology Academic Clinical ProgramDuke-NUS Medical School Singapore, Singapore
| |
Collapse
|
20
|
Zhang P, Li Z, Wang N, Duan G, Wang W, Feng Y, Zhao Y, Wang L, Zhu H, Zhang Q, Liu X, Wu W, Wu Y, Yao W, Wang J, Wu Y, Feng F. Coal tar pitch extract could induce chromosomal instability of human bronchial epithelial cells mediated by spindle checkpoint-related proteins. Oncotarget 2017; 8:56506-56517. [PMID: 28915607 PMCID: PMC5593578 DOI: 10.18632/oncotarget.17025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2016] [Accepted: 03/21/2017] [Indexed: 11/25/2022] Open
Abstract
Coal tar pitch (CTP) is a byproduct of coal tar distillation. The workers working with coal tar or in aluminum smelters, potrooms and carbon plants have the opportunities of exposing to coal tar pitch volatiles. Coal tar pitches from which polycyclic aromatic hydrocarbons (PAHs) originate have been shown to exhibit lung carcinogenicity in humans. Chromosomal instability (CIN) is a mechanism in carcinogenesis, however, whether CIN is involved in coal tar pitch-induced lung cancer remains elusive. In this present study, human bronchial epithelial cells (BEAS-2B) were first exposed to coal tar pitch extracts (CTPE) to induce a malignant transformation model. Then, the occurrence of severe chromosomal changes detected using G band, R band and multiplex fluorescence in situ hybridization (M-FISH) staining were examined. It was shown that more clones of transformed BEAS-2B cells at passage 30 following stimulation with CTPE were formed in the soft agar compared with the vehicle control. Moreover, the expression of the spindle checkpoint-related proteins, mitotic arrest defective 2 (Mad2), budding uninhibited in benzimidazole 1 (Bub1), and anaphase-promoting complex (APC), indicators of abnormal chromosomes and carcinogenesis, reduced in CTPE-treated BEAS-2B cells at Passage 30 compared with the vehicle control using real-time PCR and immunohistochemistry. In summary, exposure of BEAS-2B cells to CTPE may induce chromosomal instability through spindle checkpoint-related proteins.
Collapse
Affiliation(s)
- Peng Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China.,The Affiliated Cancer Hospital of Zhengzhou University (Henan Cancer Hospital), Zhengzhou, Henan, China
| | - Zhitao Li
- Medical College, Henan University of Science and Technology, Luoyang, Henan, China
| | - Na Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Guangcai Duan
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Wei Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Yanming Feng
- Medical College, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yong Zhao
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Lixia Wang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Hansong Zhu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Qiao Zhang
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Xiaozhuan Liu
- Medical College, Henan University of Science and Technology, Luoyang, Henan, China
| | - Weidong Wu
- School of Public Health, Xinxiang Medical University, Xinxiang, Henan, China
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Wu Yao
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Wang
- Department of Pulmonary, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yiming Wu
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| | - Feifei Feng
- College of Public Health, Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
21
|
Huang T, Yang L, Wang G, Ding G, Peng B, Wen Y, Wang Z. Inhibition of Skp2 sensitizes lung cancer cells to paclitaxel. Onco Targets Ther 2017; 10:439-446. [PMID: 28176922 PMCID: PMC5261849 DOI: 10.2147/ott.s125789] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
S-phase kinase-associated protein 2 (Skp2) is an E3 ubiquitin ligase and plays an important role in the control of cell cycle progression. Skp2 is upregulated in several cancers, including lung cancers, but the role of Skp2 in the tumorigenesis and anticancer drug resistance in human lung cancer remains to be determined. We report here that Skp2 positively regulated mitotic arrest deficient 2 (MAD2) expression and that inhibition of Skp2 sensitizes human lung cancer cells to paclitaxel. Knockdown of Skp2 by small interfering RNA (siRNA) decreased Mad2 messenger RNA (mRNA) and protein levels in A549 and NCI-H1975 cells, accompanied with upregulation of p27 but decrease of the phosphorylation of retinoblastoma (Rb). In contrast, ectopic overexpression of Skp2 increased Mad2 mRNA and protein levels and phosphorylation of Rb, while it decreased p27. Pharmacological inhibition of CDK1/2 by flavopiridol or E2F1 with HLM006474 led to downregulation of Mad2 expression and prevented the increase of Mad2 expression by Skp2. Most importantly, pharmacological inhibition of Skp2 sensitized A549 and NCI-H1299 cells to paclitaxel. Our results demonstrated that SKP2 positively regulates the gene expression of MAD2 through p27-CDKs-E2F1 signaling pathway and that inhibition of Skp2 sensitizes A549 and NCI-H1299 cells to paclitaxel, suggesting that small molecule inhibitors of Skp2 are potential agents for the treatment of lung cancer with upregulation of Skp2.
Collapse
Affiliation(s)
- Tonghai Huang
- Department of Thoracic Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Lin Yang
- Department of Thoracic Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Guangsuo Wang
- Department of Thoracic Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Guanggui Ding
- Department of Thoracic Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Bin Peng
- Department of Thoracic Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Yuxin Wen
- Department of Thoracic Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| | - Zheng Wang
- Department of Thoracic Surgery, Shenzhen People's Hospital, Shenzhen, Guangdong Province, People's Republic of China
| |
Collapse
|
22
|
Meng Z, Chen G, Chen J, Yang B, Yu M, Feng L, Jiang Z, Guo W, Tian W. Tumorigenicity analysis of heterogeneous dental stem cells and its self-modification for chromosome instability. Cell Cycle 2016; 14:3396-407. [PMID: 26322910 DOI: 10.1080/15384101.2015.1036204] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Heterogeneity demonstrates that stem cells are constituted by several sub-clones in various differentiation states. The heterogeneous state is maintained by cross-talk among sub-clones, thereby ensuring stem cell adaption. In this study, we investigated the roles of heterogeneity on genetic stability. Three sub-clones (DF2, DF8 and DF18) were isolated from heterogeneous dental stem cells (DSCs), and were proved to be chromosome instability (CIN) after long term expansion. Cell apoptosis were not detected in sub-clones, which exhibited strong tumorigenesis tendency, coupled with weak expression of p53 and aberrant ultra-structure. However, 3 sub-clones did not overexpress tumor related markers or induce tumorigenesis in vivo. The mixed-culture study suggested that 3-clone-mixed culturing cells (DF1) presented apparent decrease in the ratio of aneuploidy. The screening experiment further proved that 3 sub-clones functioned separately in this modification procedure but only mixed culturing all 3 sub-clones, simulated heterogeneous microenvironment, could achieve complete modification. Additionally, osteogenesis capability of 3 sub-clones was partially influenced by CIN while DSCs still kept stronger osteogenesis than sub-clones. These results suggested aberrant sub-clones isolated from heterogeneous DSCs were not tumorigenesis and could modify CIN by cross-talk among themselves, indicating that the heterogeneity played a key role in maintaining genetic stability and differentiation capability in dental stem cells.
Collapse
Affiliation(s)
- Zhaosong Meng
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,c Department of Oral and Maxillofacial Surgery ; West China School of Stomatology; Sichuan University ; Chengdu , China
| | - Guoqing Chen
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China
| | - Jinlong Chen
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,c Department of Oral and Maxillofacial Surgery ; West China School of Stomatology; Sichuan University ; Chengdu , China
| | - Bo Yang
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,c Department of Oral and Maxillofacial Surgery ; West China School of Stomatology; Sichuan University ; Chengdu , China
| | - Mei Yu
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China
| | - Lian Feng
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China
| | - Zongting Jiang
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China
| | - Weihua Guo
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,d Department of Pedodontics ; West China School of Stomatology; Sichuan University ; Chengdu , China
| | - Weidong Tian
- a National Engineering Laboratory for Oral Regenerative Medicine; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,b State Key Laboratory of Oral Diseases; West China Hospital of Stomatology; Sichuan University ; Chengdu , China.,c Department of Oral and Maxillofacial Surgery ; West China School of Stomatology; Sichuan University ; Chengdu , China
| |
Collapse
|
23
|
Shandilya J, Medler KF, Roberts SGE. Regulation of AURORA B function by mitotic checkpoint protein MAD2. Cell Cycle 2016; 15:2196-2201. [PMID: 27341405 DOI: 10.1080/15384101.2016.1200773] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Cell cycle checkpoint signaling stringently regulates chromosome segregation during cell division. MAD2 is one of the key components of the spindle and mitotic checkpoint complex that regulates the fidelity of cell division along with MAD1, CDC20, BUBR1, BUB3 and MAD3. MAD2 ablation leads to erroneous attachment of kinetochore-spindle fibers and defective chromosome separation. A potential role for MAD2 in the regulation of events beyond the spindle and mitotic checkpoints is not clear. Together with active spindle assembly checkpoint signaling, AURORA B kinase activity is essential for chromosome condensation as cells enter mitosis. AURORA B phosphorylates histone H3 at serine 10 and serine 28 to facilitate the formation of condensed metaphase chromosomes. In the absence of functional AURORA B cells escape mitosis despite the presence of misaligned chromosomes. In this study we report that silencing of MAD2 results in a drastic reduction of metaphase-specific histone H3 phosphorylation at serine 10 and serine 28. We demonstrate that this is due to mislocalization of AURORA B in the absence of MAD2. Conversely, overexpression of MAD2 concentrated the localization of AURORA B at the metaphase plate and caused hyper-phosphorylation of histone H3. We find that MAD1 plays a minor role in influencing the MAD2-dependent regulation of AURORA B suggesting that the effects of MAD2 on AURORA B are independent of the spindle checkpoint complex. Our findings reveal that, in addition to its role in checkpoint signaling, MAD2 ensures chromosome stability through the regulation of AURORA B.
Collapse
Affiliation(s)
- Jayasha Shandilya
- a Department of Biological Sciences , University at Buffalo , Buffalo , NY , USA
| | - Kathryn F Medler
- a Department of Biological Sciences , University at Buffalo , Buffalo , NY , USA
| | - Stefan G E Roberts
- a Department of Biological Sciences , University at Buffalo , Buffalo , NY , USA.,b School of Cellular and Molecular Medicine, University of Bristol , Bristol , UK
| |
Collapse
|
24
|
Pryzhkova MV, Jordan PW. Conditional mutation of Smc5 in mouse embryonic stem cells perturbs condensin localization and mitotic progression. J Cell Sci 2016; 129:1619-34. [PMID: 26919979 PMCID: PMC4852767 DOI: 10.1242/jcs.179036] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2015] [Accepted: 02/22/2016] [Indexed: 12/11/2022] Open
Abstract
Correct duplication of stem cell genetic material and its appropriate segregation into daughter cells are requisites for tissue, organ and organism homeostasis. Disruption of stem cell genomic integrity can lead to developmental abnormalities and cancer. Roles of the Smc5/6 structural maintenance of chromosomes complex in pluripotent stem cell genome maintenance have not been investigated, despite its important roles in DNA synthesis, DNA repair and chromosome segregation as evaluated in other model systems. Using mouse embryonic stem cells (mESCs) with a conditional knockout allele of Smc5, we showed that Smc5 protein depletion resulted in destabilization of the Smc5/6 complex, accumulation of cells in G2 phase of the cell cycle and apoptosis. Detailed assessment of mitotic mESCs revealed abnormal condensin distribution and perturbed chromosome segregation, accompanied by irregular spindle morphology, lagging chromosomes and DNA bridges. Mutation of Smc5 resulted in retention of Aurora B kinase and enrichment of condensin on chromosome arms. Furthermore, we observed reduced levels of Polo-like kinase 1 at kinetochores during mitosis. Our study reveals crucial requirements of the Smc5/6 complex during cell cycle progression and for stem cell genome maintenance.
Collapse
Affiliation(s)
- Marina V Pryzhkova
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| | - Philip W Jordan
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205, USA
| |
Collapse
|
25
|
HSET overexpression fuels tumor progression via centrosome clustering-independent mechanisms in breast cancer patients. Oncotarget 2016; 6:6076-91. [PMID: 25788277 PMCID: PMC4467423 DOI: 10.18632/oncotarget.3475] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2014] [Accepted: 01/20/2015] [Indexed: 01/31/2023] Open
Abstract
Human breast tumors harbor supernumerary centrosomes in almost 80% of tumor cells. Although amplified centrosomes compromise cell viability via multipolar spindles resulting in death-inducing aneuploidy, cancer cells tend to cluster extra centrosomes during mitosis. As a result cancer cells display bipolar spindle phenotypes to maintain a tolerable level of aneuploidy, an edge to their survival. HSET/KifC1, a kinesin-like minus-end directed microtubule motor has recently found fame as a crucial centrosome clustering molecule. Here we show that HSET promotes tumor progression via mechanisms independent of centrosome clustering. We found that HSET is overexpressed in breast carcinomas wherein nuclear HSET accumulation correlated with histological grade and predicted poor progression-free and overall survival. In addition, deregulated HSET protein expression was associated with gene amplification and/or translocation. Our data provide compelling evidence that HSET overexpression is pro-proliferative, promotes clonogenic-survival and enhances cell-cycle kinetics through G2 and M-phases. Importantly, HSET co-immunoprecipitates with survivin, and its overexpression protects survivin from proteasome-mediated degradation, resulting in its increased steady-state levels. We provide the first evidence of centrosome clustering-independent activities of HSET that fuel tumor progression and firmly establish that HSET can serve both as a potential prognostic biomarker and as a valuable cancer-selective therapeutic target.
Collapse
|
26
|
Abstract
Wilms' tumor-1 protein (WT1) is a transcription factor that can either activate or repress genes to regulate cell growth, apoptosis and differentiation. WT1 can act as either a tumor suppressor or an oncogene. The cellular functions of WT1 are predominantly regulated by its various interacting partners. Recently we have found that WT1 can regulate the fidelity of chromosome segregation through its interaction with the spindle assembly checkpoint protein, Mitotic arrest deficient-2 (MAD2). WT1 delays anaphase entry by inhibiting the ubiquitination activity of the Anaphase promoting complex/cyclosome (APC/C). Our findings have revealed an important role of WT1 in the regulation of mitotic checkpoint and genomic stability.
Collapse
Affiliation(s)
- Jayasha Shandilya
- a Department of Biological Sciences ; University at Buffalo ; Buffalo , NY USA
| | | |
Collapse
|
27
|
Tissue Regeneration in the Chronically Inflamed Tumor Environment: Implications for Cell Fusion Driven Tumor Progression and Therapy Resistant Tumor Hybrid Cells. Int J Mol Sci 2015; 16:30362-81. [PMID: 26703575 PMCID: PMC4691180 DOI: 10.3390/ijms161226240] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 12/07/2015] [Accepted: 12/09/2015] [Indexed: 12/19/2022] Open
Abstract
The biological phenomenon of cell fusion in a cancer context is still a matter of controversial debates. Even though a plethora of in vitro and in vivo data have been published in the past decades the ultimate proof that tumor hybrid cells could originate in (human) cancers and could contribute to the progression of the disease is still missing, suggesting that the cell fusion hypothesis is rather fiction than fact. However, is the lack of this ultimate proof a valid argument against this hypothesis, particularly if one has to consider that appropriate markers do not (yet) exist, thus making it virtually impossible to identify a human tumor cell clearly as a tumor hybrid cell. In the present review, we will summarize the evidence supporting the cell fusion in cancer concept. Moreover, we will refine the cell fusion hypothesis by providing evidence that cell fusion is a potent inducer of aneuploidy, genomic instability and, most likely, even chromothripsis, suggesting that cell fusion, like mutations and aneuploidy, might be an inducer of a mutator phenotype. Finally, we will show that "accidental" tissue repair processes during cancer therapy could lead to the origin of therapy resistant cancer hybrid stem cells.
Collapse
|
28
|
Su L, Shen T, Huang G, Long J, Fan J, Ling W, Jiang J. Genetic association of GWAS-supported MAD1L1 gene polymorphism rs12666575 with schizophrenia susceptibility in a Chinese population. Neurosci Lett 2015; 610:98-103. [PMID: 26528791 DOI: 10.1016/j.neulet.2015.10.061] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/03/2015] [Accepted: 10/23/2015] [Indexed: 12/20/2022]
Abstract
Schizophrenia (SCZ) is a severe neuropsychiatric disorder with high heritability. A recent European genome-wide association study has reported that mitotic arrest deficient-like 1 (MAD1L1) polymorphism rs12666575 is associated with SCZ susceptibility. This study aims to test the association of MAD1L1 variant rs12666575 with SCZ susceptibility in a Chinese population. A total of 1400 participants, which include 700 SCZ patients and 700 sex- and age-matched controls (Zhuang: 300, Han: 400, respectively), were genotyped using the Sequenom MassARRAY iPLEX platform. 591 SCZ patients underwent positive and negative syndrome scale (PANSS) assessment. Genetic association analysis was performed using the PLINK program. The results showed MAD1L1 rs12666575 polymorphism was significantly associated with SCZ susceptibility in the recessive model (p(adj)=0.013). Also, rs12666575 was significantly associated with general psychopathology sub-scale score (p(adj)=0.043) and thought disturbance factor score (p(adj)=0.045). Our data suggested that MAD1L1 rs12666575 polymorphism may play a protective role against SCZ in the Chinese population. Furthermore, rs12666575 may be associated with general psychopathology and thought disturbance in SCZ patients.
Collapse
Affiliation(s)
- Li Su
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, China.
| | - Tingting Shen
- First Affiliated Hospital, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Guifeng Huang
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianxiong Long
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China; Guangxi Colleges and Universities Key Laboratory of Prevention and Control of Highly Prevalent Diseases, Nanning, Guangxi, China
| | - Jingyuan Fan
- Pre-Clinical Faculty of Guangxi Medical University, Nanning, Guangxi, China
| | - Weijun Ling
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| | - Juan Jiang
- School of Public Health of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
29
|
Carbone L, Chavez SL. Mammalian pre-implantation chromosomal instability: species comparison, evolutionary considerations, and pathological correlations. Syst Biol Reprod Med 2015; 61:321-35. [PMID: 26366555 DOI: 10.3109/19396368.2015.1073406] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Pre-implantation embryo development in mammals begins at fertilization with the migration and fusion of the maternal and paternal pro-nuclei, followed by the degradation of inherited factors involved in germ cell specification and the activation of embryonic genes required for subsequent cell divisions, compaction, and blastulation. The majority of studies on early embryogenesis have been conducted in the mouse or non-mammalian species, often requiring extrapolation of the findings to human development. Given both conserved similarities and species-specific differences, however, even comparison between closely related mammalian species may be challenging as certain aspects, including susceptibility to chromosomal aberrations, varies considerably across mammals. Moreover, most human embryo studies are limited to patient samples obtained from in vitro fertilization (IVF) clinics and donated for research, which are generally of poorer quality and produced with germ cells that may be sub-optimal. Recent technical advances in genetic, epigenetic, chromosomal, and time-lapse imaging analyses of high quality whole human embryos have greatly improved our understanding of early human embryogenesis, particularly at the single embryo and cell level. This review summarizes the major characteristics of mammalian pre-implantation development from a chromosomal perspective, in addition to discussing the technological achievements that have recently been developed to obtain this data. We also discuss potential translation to clinical applications in reproductive medicine and conclude by examining the broader implications of these findings for the evolution of mammalian species and cancer pathology in somatic cells.
Collapse
Affiliation(s)
- Lucia Carbone
- a Division of Neuroscience , Oregon National Primate Research Center .,b Department of Behavioral Neuroscience .,c Department of Molecular & Medical Genetics .,d Bioinformatics & Computational Biology, Oregon Health & Science University
| | - Shawn L Chavez
- e Division of Reproductive & Developmental Sciences , Oregon National Primate Research Center .,f Department of Obstetrics & Gynecology , and.,g Department of Physiology & Pharmacology , Oregon Health & Science University , Portland , Oregon , USA
| |
Collapse
|
30
|
Krefman NI, Drubin DG, Barnes G. Control of the spindle checkpoint by lateral kinetochore attachment and limited Mad1 recruitment. Mol Biol Cell 2015; 26:2620-39. [PMID: 26023090 PMCID: PMC4501360 DOI: 10.1091/mbc.e15-05-0276] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 05/18/2015] [Indexed: 01/06/2023] Open
Abstract
The spindle checkpoint proteins Mad1 and Bub1 are dynamically recruited after induced de novo kinetochore assembly. Detached kinetochores compete with alternate binding sites in the nucleus to recruit Mad1 and Bub1 from very limited pools. Lateral kinetochore attachment to microtubules licenses Mad1 removal from kinetochores. We observed the dynamic recruitment of spindle checkpoint proteins Mad1 and Bub1 to detached kinetochores in budding yeast using real-time live-cell imaging and quantified recruitment in fixed cells. After induced de novo kinetochore assembly at one pair of sister centromeres, Mad1 appeared after the kinetochore protein Mtw1. Detached kinetochores were not associated with the nuclear envelope, so Mad1 does not anchor them to nuclear pore complexes (NPCs). Disrupting Mad1's NPC localization increased Mad1 recruitment to detached sister kinetochores. Conversely, increasing the number of detached kinetochores reduced the amount of Mad1 per detached kinetochore. Bub1 also relocalized completely from the spindle to detached sister centromeres after kinetochore assembly. After their capture by microtubules, Mad1 and Bub1 progressively disappeared from kinetochores. Sister chromatids that arrested with a lateral attachment to one microtubule exhibited half the Mad1 of fully detached sisters. We propose that detached kinetochores compete with alternate binding sites in the nucleus to recruit Mad1 and Bub1 from available pools that are small enough to be fully depleted by just one pair of detached kinetochores and that lateral attachment licenses Mad1 removal from kinetochores after a kinetic delay.
Collapse
Affiliation(s)
- Nathaniel I Krefman
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - David G Drubin
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| | - Georjana Barnes
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720
| |
Collapse
|
31
|
Abstract
The centromere-the primary constriction of monocentric chromosomes-is essential for correct segregation of chromosomes during mitosis and meiosis. Centromeric DNA varies between different organisms in sequence composition and extension. The main components of centromeric and pericentromeric DNA of Brassicaceae species are centromeric satellite repeats. Centromeric DNA initiates assembly of the kinetochore, the large protein complex where the spindle fibers attach during nuclear division to pull sister chromatids apart. Kinetochore assembly is initiated by incorporation of the centromeric histone H3 cenH3 into centromeric nucleosomes. The spindle assembly checkpoint acts during mitosis and meiosis at centromeres and maintains genome stability by preventing chromosome segregation before all kinetochores are correctly attached to microtubules. The function of the spindle assembly checkpoint in plants is still poorly understood. Here, we review recent advances of studies on structure and functional importance of centromeric DNA of Brassicaceae, assembly and function of cenH3 in Arabidopsis thaliana and characterization of core SAC proteins of A. thaliana in comparison with non-plant homologues.
Collapse
Affiliation(s)
- Inna Lermontova
- Leibniz Institute of Plant Genetics and Crop Plant Research, 06466, Gatersleben, Germany,
| | | | | |
Collapse
|
32
|
Restriction endonucleases from invasive Neisseria gonorrhoeae cause double-strand breaks and distort mitosis in epithelial cells during infection. PLoS One 2014; 9:e114208. [PMID: 25460012 PMCID: PMC4252111 DOI: 10.1371/journal.pone.0114208] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Accepted: 10/31/2014] [Indexed: 01/03/2023] Open
Abstract
The host epithelium is both a barrier against, and the target for microbial infections. Maintaining regulated cell growth ensures an intact protective layer towards microbial-induced cellular damage. Neisseria gonorrhoeae infections disrupt host cell cycle regulation machinery and the infection causes DNA double strand breaks that delay progression through the G2/M phase. We show that intracellular gonococci upregulate and release restriction endonucleases that enter the nucleus and damage human chromosomal DNA. Bacterial lysates containing restriction endonucleases were able to fragment genomic DNA as detected by PFGE. Lysates were also microinjected into the cytoplasm of cells in interphase and after 20 h, DNA double strand breaks were identified by 53BP1 staining. In addition, by using live-cell microscopy and NHS-ester stained live gonococci we visualized the subcellular location of the bacteria upon mitosis. Infected cells show dysregulation of the spindle assembly checkpoint proteins MAD1 and MAD2, impaired and prolonged M-phase, nuclear swelling, micronuclei formation and chromosomal instability. These data highlight basic molecular functions of how gonococcal infections affect host cell cycle regulation, cause DNA double strand breaks and predispose cellular malignancies.
Collapse
|
33
|
Structure-biological function relationship extended to mitotic arrest-deficient 2-like protein Mad2 native and mutants-new opportunity for genetic disorder control. Int J Mol Sci 2014; 15:21381-400. [PMID: 25411801 PMCID: PMC4264231 DOI: 10.3390/ijms151121381] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 11/07/2014] [Accepted: 11/07/2014] [Indexed: 02/07/2023] Open
Abstract
Overexpression of mitotic arrest-deficient proteins Mad1 and Mad2, two components of spindle assembly checkpoint, is a risk factor for chromosomal instability (CIN) and a trigger of many genetic disorders. Mad2 transition from inactive open (O-Mad2) to active closed (C-Mad2) conformations or Mad2 binding to specific partners (cell-division cycle protein 20 (Cdc20) or Mad1) were targets of previous pharmacogenomics studies. Here, Mad2 binding to Cdc20 and the interconversion rate from open to closed Mad2 were predicted and the molecular features with a critical contribution to these processes were determined by extending the quantitative structure-activity relationship (QSAR) method to large-size proteins such as Mad2. QSAR models were built based on available published data on 23 Mad2 mutants inducing CIN-related functional changes. The most relevant descriptors identified for predicting Mad2 native and mutants action mechanism and their involvement in genetic disorders are the steric (van der Waals area and solvent accessible area and their subdivided) and energetic van der Waals energy descriptors. The reliability of our QSAR models is indicated by significant values of statistical coefficients: Cross-validated correlation q2 (0.53–0.65) and fitted correlation r2 (0.82–0.90). Moreover, based on established QSAR equations, we rationally design and analyze nine de novo Mad2 mutants as possible promoters of CIN.
Collapse
|
34
|
Zheng Y, Guo J, Li X, Xie Y, Hou M, Fu X, Dai S, Diao R, Miao Y, Ren J. An integrated overview of spatiotemporal organization and regulation in mitosis in terms of the proteins in the functional supercomplexes. Front Microbiol 2014; 5:573. [PMID: 25400627 PMCID: PMC4212687 DOI: 10.3389/fmicb.2014.00573] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 10/11/2014] [Indexed: 12/22/2022] Open
Abstract
Eukaryotic cells may divide via the critical cellular process of cell division/mitosis, resulting in two daughter cells with the same genetic information. A large number of dedicated proteins are involved in this process and spatiotemporally assembled into three distinct super-complex structures/organelles, including the centrosome/spindle pole body, kinetochore/centromere and cleavage furrow/midbody/bud neck, so as to precisely modulate the cell division/mitosis events of chromosome alignment, chromosome segregation and cytokinesis in an orderly fashion. In recent years, many efforts have been made to identify the protein components and architecture of these subcellular organelles, aiming to uncover the organelle assembly pathways, determine the molecular mechanisms underlying the organelle functions, and thereby provide new therapeutic strategies for a variety of diseases. However, the organelles are highly dynamic structures, making it difficult to identify the entire components. Here, we review the current knowledge of the identified protein components governing the organization and functioning of organelles, especially in human and yeast cells, and discuss the multi-localized protein components mediating the communication between organelles during cell division.
Collapse
Affiliation(s)
- Yueyuan Zheng
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Junjie Guo
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Xu Li
- Orthopaedic Department of Anhui Medical University Affiliated Provincial Hospital Hefei, China
| | - Yubin Xie
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Mingming Hou
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Xuyang Fu
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Shengkun Dai
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Rucheng Diao
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Yanyan Miao
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| | - Jian Ren
- Cancer Center, School of Life Sciences, School of Advanced Computing, Cooperative Innovation Center for High Performance Computing, Sun Yat-sen University Guangzhou, China
| |
Collapse
|
35
|
Taipale M, Tucker G, Peng J, Krykbaeva I, Lin ZY, Larsen B, Choi H, Berger B, Gingras AC, Lindquist S. A quantitative chaperone interaction network reveals the architecture of cellular protein homeostasis pathways. Cell 2014; 158:434-448. [PMID: 25036637 DOI: 10.1016/j.cell.2014.05.039] [Citation(s) in RCA: 311] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 03/08/2014] [Accepted: 05/16/2014] [Indexed: 12/27/2022]
Abstract
Chaperones are abundant cellular proteins that promote the folding and function of their substrate proteins (clients). In vivo, chaperones also associate with a large and diverse set of cofactors (cochaperones) that regulate their specificity and function. However, how these cochaperones regulate protein folding and whether they have chaperone-independent biological functions is largely unknown. We combined mass spectrometry and quantitative high-throughput LUMIER assays to systematically characterize the chaperone-cochaperone-client interaction network in human cells. We uncover hundreds of chaperone clients, delineate their participation in specific cochaperone complexes, and establish a surprisingly distinct network of protein-protein interactions for cochaperones. As a salient example of the power of such analysis, we establish that NUDC family cochaperones specifically associate with structurally related but evolutionarily distinct β-propeller folds. We provide a framework for deciphering the proteostasis network and its regulation in development and disease and expand the use of chaperones as sensors for drug-target engagement.
Collapse
Affiliation(s)
- Mikko Taipale
- Whitehead Institute for Biomedical Research, Cambridge, MA 02114, USA
| | - George Tucker
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jian Peng
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Irina Krykbaeva
- Whitehead Institute for Biomedical Research, Cambridge, MA 02114, USA
| | - Zhen-Yuan Lin
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Brett Larsen
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada
| | - Hyungwon Choi
- National University of Singapore and National University Health System, Singapore 117597, Singapore
| | - Bonnie Berger
- Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Mathematics, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Anne-Claude Gingras
- Centre for Systems Biology, Lunenfeld-Tanenbaum Research Institute at Mount Sinai Hospital, Department of Molecular Genetics, University of Toronto, Toronto, ON M5G 1X5, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada.
| | - Susan Lindquist
- Whitehead Institute for Biomedical Research, Cambridge, MA 02114, USA; Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Howard Hughes Medical Institute, Cambridge, MA 02139, USA.
| |
Collapse
|
36
|
London N, Biggins S. Signalling dynamics in the spindle checkpoint response. Nat Rev Mol Cell Biol 2014; 15:736-47. [PMID: 25303117 DOI: 10.1038/nrm3888] [Citation(s) in RCA: 245] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The spindle checkpoint ensures proper chromosome segregation during cell division. Unravelling checkpoint signalling has been a long-standing challenge owing to the complexity of the structures and forces that regulate chromosome segregation. New reports have now substantially advanced our understanding of checkpoint signalling mechanisms at the kinetochore, the structure that connects microtubules and chromatin. In contrast to the traditional view of a binary checkpoint response - either completely on or off - new findings indicate that the checkpoint response strength is variable. This revised perspective provides insight into how checkpoint bypass can lead to aneuploidy and informs strategies to exploit these errors for cancer treatments.
Collapse
Affiliation(s)
- Nitobe London
- 1] Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N., PO Box 19024, Seattle, Washington 98109, USA. [2] Molecular and Cellular Biology Program, University of Washington/Fred Hutchinson Cancer Research Center, Seattle, Washington 98109, USA
| | - Sue Biggins
- Division of Basic Sciences, Fred Hutchinson Cancer Research Center, 1100 Fairview Ave N., PO Box 19024, Seattle, Washington 98109, USA
| |
Collapse
|
37
|
Lee S, Bolanos-Garcia VM. The dynamics of signal amplification by macromolecular assemblies for the control of chromosome segregation. Front Physiol 2014; 5:368. [PMID: 25324779 PMCID: PMC4179342 DOI: 10.3389/fphys.2014.00368] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2014] [Accepted: 09/07/2014] [Indexed: 11/13/2022] Open
Abstract
The control of chromosome segregation relies on the spindle assembly checkpoint (SAC), a complex regulatory system that ensures the high fidelity of chromosome segregation in higher organisms by delaying the onset of anaphase until each chromosome is properly bi-oriented on the mitotic spindle. Central to this process is the establishment of multiple yet specific protein-protein interactions in a narrow time-space window. Here we discuss the highly dynamic nature of multi-protein complexes that control chromosome segregation in which an intricate network of weak but cooperative interactions modulate signal amplification to ensure a proper SAC response. We also discuss the current structural understanding of the communication between the SAC and the kinetochore; how transient interactions can regulate the assembly and disassembly of the SAC as well as the challenges and opportunities for the definition and the manipulation of the flow of information in SAC signaling.
Collapse
Affiliation(s)
- Semin Lee
- Center for Biomedical Informatics, Harvard Medical School, Harvard University Boston, MA, USA
| | - Victor M Bolanos-Garcia
- Department of Biological and Medical Sciences, Faculty of Health and Life Sciences, Oxford Brookes University Oxford, UK
| |
Collapse
|
38
|
Carbone L, Harris RA, Gnerre S, Veeramah KR, Lorente-Galdos B, Huddleston J, Meyer TJ, Herrero J, Roos C, Aken B, Anaclerio F, Archidiacono N, Baker C, Barrell D, Batzer MA, Beal K, Blancher A, Bohrson CL, Brameier M, Campbell MS, Capozzi O, Casola C, Chiatante G, Cree A, Damert A, de Jong PJ, Dumas L, Fernandez-Callejo M, Flicek P, Fuchs NV, Gut I, Gut M, Hahn MW, Hernandez-Rodriguez J, Hillier LW, Hubley R, Ianc B, Izsvák Z, Jablonski NG, Johnstone LM, Karimpour-Fard A, Konkel MK, Kostka D, Lazar NH, Lee SL, Lewis LR, Liu Y, Locke DP, Mallick S, Mendez FL, Muffato M, Nazareth LV, Nevonen KA, O'Bleness M, Ochis C, Odom DT, Pollard KS, Quilez J, Reich D, Rocchi M, Schumann GG, Searle S, Sikela JM, Skollar G, Smit A, Sonmez K, ten Hallers B, Terhune E, Thomas GWC, Ullmer B, Ventura M, Walker JA, Wall JD, Walter L, Ward MC, Wheelan SJ, Whelan CW, White S, Wilhelm LJ, Woerner AE, Yandell M, Zhu B, Hammer MF, Marques-Bonet T, Eichler EE, Fulton L, Fronick C, Muzny DM, Warren WC, Worley KC, Rogers J, Wilson RK, Gibbs RA. Gibbon genome and the fast karyotype evolution of small apes. Nature 2014; 513:195-201. [PMID: 25209798 PMCID: PMC4249732 DOI: 10.1038/nature13679] [Citation(s) in RCA: 221] [Impact Index Per Article: 20.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2014] [Accepted: 07/14/2014] [Indexed: 12/22/2022]
Abstract
Gibbons are small arboreal apes that display an accelerated rate of evolutionary chromosomal rearrangement and occupy a key node in the primate phylogeny between Old World monkeys and great apes. Here we present the assembly and analysis of a northern white-cheeked gibbon (Nomascus leucogenys) genome. We describe the propensity for a gibbon-specific retrotransposon (LAVA) to insert into chromosome segregation genes and alter transcription by providing a premature termination site, suggesting a possible molecular mechanism for the genome plasticity of the gibbon lineage. We further show that the gibbon genera (Nomascus, Hylobates, Hoolock and Symphalangus) experienced a near-instantaneous radiation ∼5 million years ago, coincident with major geographical changes in southeast Asia that caused cycles of habitat compression and expansion. Finally, we identify signatures of positive selection in genes important for forelimb development (TBX5) and connective tissues (COL1A1) that may have been involved in the adaptation of gibbons to their arboreal habitat. The genome of the gibbon, a tree-dwelling ape from Asia positioned between Old World monkeys and the great apes, is presented, providing insights into the evolutionary history of gibbon species and their accelerated karyotypes, as well as evidence for selection of genes such as those for forelimb development and connective tissue that may be important for locomotion through trees. The many species of gibbons are small, tree-living apes from Southeast Asia, most of them listed as 'endangered' or 'critically endangered' on the IUCN list. In their presentation of the genome of the northern white-cheeked gibbon (Nomascus leucogenys) , Lucia Carbone and colleagues provide intriguing insights into the biology and evolutionary history of a group that straddles the divide between Old World monkeys and the great apes. The authors investigate how a novel gibbon-specific retrotransposon might be the source of gibbons' genome plasticity. Rapid karyotype evolution combined with multiple episodes of climate and environmental change might explain the almost instantaneous divergence of the four gibbon genera. Positive selection on genes involved in forelimb development and connective tissue might have been related to gibbons' unique mode of locomotion in the tropical canopy.
Collapse
Affiliation(s)
- Lucia Carbone
- 1] Oregon Health &Science University, Department of Behavioral Neuroscience, 3181 SW Sam Jackson Park Road Portland, Oregon 97239, USA. [2] Oregon National Primate Research Center, Division of Neuroscience, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA. [3] Oregon Health &Science University, Department of Molecular &Medical Genetics, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA. [4] Oregon Health &Science University, Bioinformatics and Computational Biology Division, Department of Medical Informatics &Clinical Epidemiology, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA
| | - R Alan Harris
- Baylor College of Medicine, Department of Molecular and Human Genetics, One Baylor Plaza, Houston, Texas 77030, USA
| | - Sante Gnerre
- Nabsys, 60 Clifford Street, Providence, Rhode Island 02903, USA
| | - Krishna R Veeramah
- 1] University of Arizona, ARL Division of Biotechnology, Tucson, Arizona 85721, USA. [2] Stony Brook University, Department of Ecology and Evolution, Stony Brook, New York 11790, USA
| | - Belen Lorente-Galdos
- IBE, Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, PRBB, Doctor Aiguader, 88, 08003 Barcelona, Spain
| | - John Huddleston
- 1] Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195, USA. [2] Howard Hughes Medical Institute, 1705 NE Pacific Street, Seattle, Washington 98195, USA
| | - Thomas J Meyer
- Oregon Health &Science University, Department of Behavioral Neuroscience, 3181 SW Sam Jackson Park Road Portland, Oregon 97239, USA
| | - Javier Herrero
- 1] European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK. [2] The Genome Analysis Centre, Norwich Research Park, Norwich NR4 7UH, UK. [3] Bill Lyons Informatics Center, UCL Cancer Institute, University College London, London WC1E 6DD, UK (J.He); Seven Bridges Genomics, Cambridge, Massachusetts 02138, USA (D.P.L.); Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA (F.L.M.); BioNano Genomics, San Diego, California 92121, USA (B.t.H.); University of Chicago, Department of Human Genetics, Chicago, Illinois 60637, USA (M.C.W.); Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02138, USA (C.W.W.); The CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China (B.Z.)
| | - Christian Roos
- Leibniz Institute for Primate Research, Gene Bank of Primates, German Primate Center, Göttingen 37077, Germany
| | - Bronwen Aken
- 1] European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK. [2] European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Fabio Anaclerio
- University of Bari, Department of Biology, Via Orabona 4, 70125, Bari, Italy
| | | | - Carl Baker
- Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195, USA
| | - Daniel Barrell
- 1] European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK. [2] European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Mark A Batzer
- Louisiana State University, Department of Biological Sciences, Baton Rouge, Louisiana 70803, USA
| | - Kathryn Beal
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | | | - Craig L Bohrson
- The Johns Hopkins University School of Medicine, Department of Oncology, Division of Biostatistics and Bioinformatics, Baltimore, Maryland 21205, USA
| | - Markus Brameier
- Leibniz Institute for Primate Research, Gene Bank of Primates, German Primate Center, Göttingen 37077, Germany
| | | | - Oronzo Capozzi
- University of Bari, Department of Biology, Via Orabona 4, 70125, Bari, Italy
| | - Claudio Casola
- Texas A&M University, Department of Ecosystem Science and Management, College Station, Texas 77843, USA
| | - Giorgia Chiatante
- University of Bari, Department of Biology, Via Orabona 4, 70125, Bari, Italy
| | - Andrew Cree
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Annette Damert
- Babes-Bolyai-University, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Molecular Biology Center, Cluj-Napoca 400084, Romania
| | - Pieter J de Jong
- Children's Hospital Oakland Research Institute, BACPAC Resources, Oakland, California 94609, USA
| | - Laura Dumas
- University of Colorado School of Medicine, Department of Biochemistry and Molecular Genetics, Aurora, Colorado 80045, USA
| | - Marcos Fernandez-Callejo
- IBE, Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, PRBB, Doctor Aiguader, 88, 08003 Barcelona, Spain
| | - Paul Flicek
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Nina V Fuchs
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Ivo Gut
- Centro Nacional de Análisis Genómico (CNAG), Parc Científic de Barcelona, Barcelona 08028, Spain
| | - Marta Gut
- Centro Nacional de Análisis Genómico (CNAG), Parc Científic de Barcelona, Barcelona 08028, Spain
| | - Matthew W Hahn
- Indiana University, School of Informatics and Computing, Bloomington, Indiana 47408, USA
| | - Jessica Hernandez-Rodriguez
- IBE, Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, PRBB, Doctor Aiguader, 88, 08003 Barcelona, Spain
| | - LaDeana W Hillier
- The Genome Center at Washington University, Washington University School of Medicine, 4444 Forest Park Avenue, Saint Louis, Missouri 63108, USA
| | - Robert Hubley
- Institute for Systems Biology, Seattle, Washington 98109-5234, USA
| | - Bianca Ianc
- Babes-Bolyai-University, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Molecular Biology Center, Cluj-Napoca 400084, Romania
| | - Zsuzsanna Izsvák
- Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Nina G Jablonski
- The Pennsylvania State University, Department of Anthropology, University Park, Pennsylvania 16802, USA
| | - Laurel M Johnstone
- University of Arizona, ARL Division of Biotechnology, Tucson, Arizona 85721, USA
| | - Anis Karimpour-Fard
- University of Colorado School of Medicine, Department of Biochemistry and Molecular Genetics, Aurora, Colorado 80045, USA
| | - Miriam K Konkel
- Louisiana State University, Department of Biological Sciences, Baton Rouge, Louisiana 70803, USA
| | - Dennis Kostka
- University of Pittsburgh School of Medicine, Department of Developmental Biology, Department of Computational and Systems Biology, Pittsburg, Pennsylvania 15261, USA
| | - Nathan H Lazar
- Oregon Health &Science University, Bioinformatics and Computational Biology Division, Department of Medical Informatics &Clinical Epidemiology, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA
| | - Sandra L Lee
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Lora R Lewis
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Yue Liu
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Devin P Locke
- 1] The Genome Center at Washington University, Washington University School of Medicine, 4444 Forest Park Avenue, Saint Louis, Missouri 63108, USA. [2] Bill Lyons Informatics Center, UCL Cancer Institute, University College London, London WC1E 6DD, UK (J.He); Seven Bridges Genomics, Cambridge, Massachusetts 02138, USA (D.P.L.); Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA (F.L.M.); BioNano Genomics, San Diego, California 92121, USA (B.t.H.); University of Chicago, Department of Human Genetics, Chicago, Illinois 60637, USA (M.C.W.); Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02138, USA (C.W.W.); The CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China (B.Z.)
| | - Swapan Mallick
- Harvard Medical School, Department of Genetics, Boston, Massachusetts 02115, USA
| | - Fernando L Mendez
- 1] University of Arizona, ARL Division of Biotechnology, Tucson, Arizona 85721, USA. [2] Bill Lyons Informatics Center, UCL Cancer Institute, University College London, London WC1E 6DD, UK (J.He); Seven Bridges Genomics, Cambridge, Massachusetts 02138, USA (D.P.L.); Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA (F.L.M.); BioNano Genomics, San Diego, California 92121, USA (B.t.H.); University of Chicago, Department of Human Genetics, Chicago, Illinois 60637, USA (M.C.W.); Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02138, USA (C.W.W.); The CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China (B.Z.)
| | - Matthieu Muffato
- European Molecular Biology Laboratory, European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Lynne V Nazareth
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Kimberly A Nevonen
- Oregon National Primate Research Center, Division of Neuroscience, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA
| | - Majesta O'Bleness
- University of Colorado School of Medicine, Department of Biochemistry and Molecular Genetics, Aurora, Colorado 80045, USA
| | - Cornelia Ochis
- Babes-Bolyai-University, Institute for Interdisciplinary Research in Bio-Nano-Sciences, Molecular Biology Center, Cluj-Napoca 400084, Romania
| | - Duncan T Odom
- 1] European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK. [2] University of Cambridge, Cancer Research UK-Cambridge Institute, Cambridge CB2 0RE, UK
| | - Katherine S Pollard
- 1] University of California, Gladstone Institutes, San Francisco, California 94158-226, USA. [2] Institute for Human Genetics, University of California, San Francisco, California 94143-0794, USA. [3] Division of Biostatistics, University of California, San Francisco, California 94143-0794, USA
| | - Javier Quilez
- IBE, Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, PRBB, Doctor Aiguader, 88, 08003 Barcelona, Spain
| | - David Reich
- Harvard Medical School, Department of Genetics, Boston, Massachusetts 02115, USA
| | - Mariano Rocchi
- University of Bari, Department of Biology, Via Orabona 4, 70125, Bari, Italy
| | - Gerald G Schumann
- Paul Ehrlich Institute, Division of Medical Biotechnology, 63225 Langen, Germany
| | - Stephen Searle
- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - James M Sikela
- University of Colorado School of Medicine, Department of Biochemistry and Molecular Genetics, Aurora, Colorado 80045, USA
| | - Gabriella Skollar
- Gibbon Conservation Center, 19100 Esguerra Rd, Santa Clarita, California 91350, USA
| | - Arian Smit
- The Genome Center at Washington University, Washington University School of Medicine, 4444 Forest Park Avenue, Saint Louis, Missouri 63108, USA
| | - Kemal Sonmez
- 1] Oregon Health &Science University, Bioinformatics and Computational Biology Division, Department of Medical Informatics &Clinical Epidemiology, 3181 SW Sam Jackson Park Road, Portland, Oregon 97239, USA. [2] Oregon Health &Science University, Center for Spoken Language Understanding, Institute on Development and Disability, Portland, Oregon 97239, USA
| | - Boudewijn ten Hallers
- 1] Children's Hospital Oakland Research Institute, BACPAC Resources, Oakland, California 94609, USA. [2] Bill Lyons Informatics Center, UCL Cancer Institute, University College London, London WC1E 6DD, UK (J.He); Seven Bridges Genomics, Cambridge, Massachusetts 02138, USA (D.P.L.); Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA (F.L.M.); BioNano Genomics, San Diego, California 92121, USA (B.t.H.); University of Chicago, Department of Human Genetics, Chicago, Illinois 60637, USA (M.C.W.); Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02138, USA (C.W.W.); The CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China (B.Z.)
| | - Elizabeth Terhune
- Oregon National Primate Research Center, Division of Neuroscience, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA
| | - Gregg W C Thomas
- Indiana University, School of Informatics and Computing, Bloomington, Indiana 47408, USA
| | - Brygg Ullmer
- Louisiana State University, School of Electrical Engineering and Computer Science, Baton Rouge, Louisiana 70803, USA
| | - Mario Ventura
- University of Bari, Department of Biology, Via Orabona 4, 70125, Bari, Italy
| | - Jerilyn A Walker
- Louisiana State University, Department of Biological Sciences, Baton Rouge, Louisiana 70803, USA
| | - Jeffrey D Wall
- 1] Institute for Human Genetics, University of California, San Francisco, California 94143-0794, USA. [2] Division of Biostatistics, University of California, San Francisco, California 94143-0794, USA
| | - Lutz Walter
- Leibniz Institute for Primate Research, Gene Bank of Primates, German Primate Center, Göttingen 37077, Germany
| | - Michelle C Ward
- 1] University of Cambridge, Cancer Research UK-Cambridge Institute, Cambridge CB2 0RE, UK. [2] Bill Lyons Informatics Center, UCL Cancer Institute, University College London, London WC1E 6DD, UK (J.He); Seven Bridges Genomics, Cambridge, Massachusetts 02138, USA (D.P.L.); Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA (F.L.M.); BioNano Genomics, San Diego, California 92121, USA (B.t.H.); University of Chicago, Department of Human Genetics, Chicago, Illinois 60637, USA (M.C.W.); Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02138, USA (C.W.W.); The CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China (B.Z.)
| | - Sarah J Wheelan
- The Johns Hopkins University School of Medicine, Department of Oncology, Division of Biostatistics and Bioinformatics, Baltimore, Maryland 21205, USA
| | - Christopher W Whelan
- 1] Oregon Health &Science University, Center for Spoken Language Understanding, Institute on Development and Disability, Portland, Oregon 97239, USA. [2] Bill Lyons Informatics Center, UCL Cancer Institute, University College London, London WC1E 6DD, UK (J.He); Seven Bridges Genomics, Cambridge, Massachusetts 02138, USA (D.P.L.); Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA (F.L.M.); BioNano Genomics, San Diego, California 92121, USA (B.t.H.); University of Chicago, Department of Human Genetics, Chicago, Illinois 60637, USA (M.C.W.); Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02138, USA (C.W.W.); The CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China (B.Z.)
| | - Simon White
- European Bioinformatics Institute, Wellcome Trust Genome Campus, Hinxton, Cambridge CB10 1SD, UK
| | - Larry J Wilhelm
- Oregon National Primate Research Center, Division of Neuroscience, 505 NW 185th Avenue, Beaverton, Oregon 97006, USA
| | - August E Woerner
- University of Arizona, ARL Division of Biotechnology, Tucson, Arizona 85721, USA
| | - Mark Yandell
- University of Utah, Salt Lake City, Utah 84112, USA
| | - Baoli Zhu
- 1] Children's Hospital Oakland Research Institute, BACPAC Resources, Oakland, California 94609, USA. [2] Bill Lyons Informatics Center, UCL Cancer Institute, University College London, London WC1E 6DD, UK (J.He); Seven Bridges Genomics, Cambridge, Massachusetts 02138, USA (D.P.L.); Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA (F.L.M.); BioNano Genomics, San Diego, California 92121, USA (B.t.H.); University of Chicago, Department of Human Genetics, Chicago, Illinois 60637, USA (M.C.W.); Stanley Center for Psychiatric Research, Broad Institute, Cambridge, Massachusetts 02138, USA (C.W.W.); The CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China (B.Z.)
| | - Michael F Hammer
- University of Arizona, ARL Division of Biotechnology, Tucson, Arizona 85721, USA
| | - Tomas Marques-Bonet
- 1] IBE, Institut de Biologia Evolutiva (UPF-CSIC), Universitat Pompeu Fabra, PRBB, Doctor Aiguader, 88, 08003 Barcelona, Spain. [2] Centro Nacional de Análisis Genómico (CNAG), Parc Científic de Barcelona, Barcelona 08028, Spain
| | - Evan E Eichler
- 1] Department of Genome Sciences, University of Washington School of Medicine, Seattle, Washington 98195, USA. [2] Howard Hughes Medical Institute, 1705 NE Pacific Street, Seattle, Washington 98195, USA
| | - Lucinda Fulton
- The Genome Center at Washington University, Washington University School of Medicine, 4444 Forest Park Avenue, Saint Louis, Missouri 63108, USA
| | - Catrina Fronick
- The Genome Center at Washington University, Washington University School of Medicine, 4444 Forest Park Avenue, Saint Louis, Missouri 63108, USA
| | - Donna M Muzny
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Wesley C Warren
- The Genome Center at Washington University, Washington University School of Medicine, 4444 Forest Park Avenue, Saint Louis, Missouri 63108, USA
| | - Kim C Worley
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Jeffrey Rogers
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | - Richard K Wilson
- The Genome Center at Washington University, Washington University School of Medicine, 4444 Forest Park Avenue, Saint Louis, Missouri 63108, USA
| | - Richard A Gibbs
- Human Genome Sequencing Center, Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| |
Collapse
|
39
|
Shandilya J, Toska E, Richard DJ, Medler KF, Roberts SGE. WT1 interacts with MAD2 and regulates mitotic checkpoint function. Nat Commun 2014; 5:4903. [PMID: 25232865 PMCID: PMC4170573 DOI: 10.1038/ncomms5903] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 08/02/2014] [Indexed: 01/08/2023] Open
Abstract
Tumour suppressors safeguard the fidelity of the mitotic checkpoint by transcriptional regulation of genes that encode components of the mitotic checkpoint complex (MCC). Here we report a new role for the tumour suppressor and transcription factor, WT1, in the mitotic checkpoint. We show that WT1 regulates the MCC by directly interacting with the spindle assembly checkpoint protein, MAD2. WT1 colocalizes with MAD2 during mitosis and preferentially binds to the functionally active, closed-conformer, C-MAD2. Furthermore, WT1 associates with the MCC containing MAD2, BUBR1 and CDC20, resulting in prolonged inhibition of the anaphase-promoting complex/cyclosome (APC/C) and delayed degradation of its substrates SECURIN and CYCLIN B1. Strikingly, RNA interference-mediated depletion of WT1 leads to enhanced turnover of SECURIN, decreased lag time to anaphase and defects in chromosome segregation. Our findings identify WT1 as a regulator of the mitotic checkpoint and chromosomal stability.
Collapse
Affiliation(s)
- Jayasha Shandilya
- Department of Biological Sciences, University at Buffalo, Cooke Hall, North Campus, Buffalo, New York 14260, USA
| | - Eneda Toska
- Department of Biological Sciences, University at Buffalo, Cooke Hall, North Campus, Buffalo, New York 14260, USA
| | - Derek J Richard
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation at the Translational Research Institute, Queensland University of Technology, Brisbane, Queensland 4102, Australia
| | - Kathryn F Medler
- Department of Biological Sciences, University at Buffalo, Cooke Hall, North Campus, Buffalo, New York 14260, USA
| | - Stefan G E Roberts
- 1] Department of Biological Sciences, University at Buffalo, Cooke Hall, North Campus, Buffalo, New York 14260, USA [2] School of Cellular and Molecular Medicine, University of Bristol, Bristol BS8 1TD, UK
| |
Collapse
|
40
|
Das T, Roy KS, Chakrabarti T, Mukhopadhyay S, Roychoudhury S. Withaferin A modulates the Spindle assembly checkpoint by degradation of Mad2-Cdc20 complex in colorectal cancer cell lines. Biochem Pharmacol 2014; 91:31-9. [PMID: 24995417 DOI: 10.1016/j.bcp.2014.06.022] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 06/20/2014] [Accepted: 06/23/2014] [Indexed: 02/01/2023]
Abstract
Withania somnifera L. Dunal (Ashwagandha) is used over centuries in the ayurvedic medicines in India. Withaferin A, a withanolide, is the major compound present in leaf extract of the plant which shows anticancer activity against leukemia, breast cancer and colorectal cancer. It arrests the ovarian cancer cells in the G2/M phase in dose dependent manner. In the current study we show the effect of Withaferin A on cell cycle regulation of colorectal cancer cell lines HCT116 and SW480 and its effect on cell fate. Treatment of these cells with this compound leads to apoptosis in a dose dependent manner. It causes the G2/M arrest in both the cell lines. We show that Withaferin A (WA) causes mitotic delay by blocking Spindle assembly checkpoint (SAC) function. Apoptosis induced by Withaferin A is associated with proteasomal degradation of Mad2 and Cdc20, an important constituent of the Spindle Checkpoint Complex. Further overexpression of Mad2 partially rescues the deleterious effect of WA by restoring proper anaphase initiation and keeping more number of cells viable. We hypothesize that Withaferin A kills cancer cells by delaying the mitotic exit followed by inducing chromosome instability.
Collapse
Affiliation(s)
- Tania Das
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India.
| | - Kumar Singha Roy
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Tulika Chakrabarti
- Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Sibabrata Mukhopadhyay
- Medicinal Chemistry Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India
| | - Susanta Roychoudhury
- Cancer Biology and Inflammatory Disorder Division, CSIR-Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Jadavpur, Kolkata 700032, India.
| |
Collapse
|
41
|
Iglesias Gómez JC, Mosquera Orgueira A. An integrative analysis of meningioma tumors reveals the determinant genes and pathways of malignant transformation. Front Oncol 2014; 4:147. [PMID: 25003081 PMCID: PMC4066933 DOI: 10.3389/fonc.2014.00147] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2013] [Accepted: 05/28/2014] [Indexed: 01/18/2023] Open
Abstract
Meningiomas are frequent central nervous system neoplasms, which despite their predominant benignity, show sporadically malignant behavior. Type 2 neurofibromatosis and polymorphisms in several genes have been associated with meningioma risk and are probably involved in its pathogenesis. Although GWAS studies have found loci related to meningioma risk, little is known about the factors determining malignant transformation. Thus, this study is aimed to identify the genomic and transcriptomic factors influencing evolution from benignity toward aggressive phenotypes. By applying an integrative bioinformatics pipeline combining public information on a wealth of biological layers of complexity (from genetic polymorphisms to protein interactions), this study identified a module of co-expressed genes highly correlated with tumor stage and statistically linked to several genomic regions (module Quantitative Trait Loci, mQTLs). Ontology analysis of the transcription hub genes identified microtubule-associated cell-cycle processes as key drivers of such network. mQTLs and single nucleotide polymorphisms associated with meningioma stage were replicated in an alternative meningioma cohort, and integration of these results with up-to-date scientific literature and several databases retrieved a list of genes and pathways with a potentially important role in meningioma malignancy. As a result, cytoskeleton and cell-cell adhesion pathways, calcium-channels and glutamate receptors, as well as oxidoreductase and endoplasmic reticulum-associated degradation pathways were found to be the most important and redundant findings associated to meningioma progression. This study presents an integrated view of the pathways involved in meningioma malignant conversion and paves the way for the development of new research lines that will improve our understanding of meningioma biology.
Collapse
|
42
|
Cell cycle progression in response to oxygen levels. Cell Mol Life Sci 2014; 71:3569-82. [PMID: 24858415 PMCID: PMC4143607 DOI: 10.1007/s00018-014-1645-9] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 05/01/2014] [Accepted: 05/05/2014] [Indexed: 02/06/2023]
Abstract
Hypoxia' or decreases in oxygen availability' results in the activation of a number of different responses at both the whole organism and the cellular level. These responses include drastic changes in gene expression, which allow the organism (or cell) to cope efficiently with the stresses associated with the hypoxic insult. A major breakthrough in the understanding of the cellular response to hypoxia was the discovery of a hypoxia sensitive family of transcription factors known as the hypoxia inducible factors (HIFs). The hypoxia response mounted by the HIFs promotes cell survival and energy conservation. As such, this response has to deal with important cellular process such as cell division. In this review, the integration of oxygen sensing with the cell cycle will be discussed. HIFs, as well as other components of the hypoxia pathway, can influence cell cycle progression. The role of HIF and the cell molecular oxygen sensors in the control of the cell cycle will be reviewed.
Collapse
|
43
|
Sze KMF, Chu GKY, Mak QHY, Lee JMF, Ng IOL. Proline-rich acidic protein 1 (PRAP1) is a novel interacting partner of MAD1 and has a suppressive role in mitotic checkpoint signalling in hepatocellular carcinoma. J Pathol 2014; 233:51-60. [PMID: 24374861 DOI: 10.1002/path.4319] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Revised: 12/11/2013] [Accepted: 12/17/2013] [Indexed: 01/28/2023]
Abstract
Loss of mitotic checkpoint of cells contributes to chromosomal instability and leads to carcinogenesis. Mitotic arrest deficient 1 (MAD1) is a key component in mitotic checkpoint signalling. In this study, we identified a novel MAD1 interacting partner, proline-rich acidic protein 1 (PRAP1), using yeast-two hybrid screening, and investigated its role in mitotic checkpoint signalling in hepatocellular carcinoma (HCC). We demonstrated the physical interaction of PRAP1 with MAD1 and of PRAP1 with MAD1 isoform MAD1β, using a co-immunoprecipitation assay. Moreover, stable expression of PRAP1 in mitotic checkpoint-competent HCC cells, BEL-7402 and SMMC-7721, induced impairment of the mitotic checkpoint (p < 0.01), formation of chromosome bridges (p < 0.01) and aberrant chromosome numbers (p < 0.001). Interestingly, ectopic expression PRAP1 in HCC cells led to significant under-expression of MAD1. In human HCC tumours, 40.4% (23/57) of HCCs showed under-expression of PRAP1 protein as compared with their corresponding non-tumorous livers; up-regulation of MAD1 protein was significantly associated with down-regulation of PRAP1 (p = 0.030). Our data revealed that PRAP1 is a protein interacting partner of MAD1 and that PRAP1 is able to down-regulate MAD1 and suppress mitotic checkpoint signalling in HCC.
Collapse
Affiliation(s)
- Karen Man-Fong Sze
- State Key Laboratory for Liver Research, University of Hong Kong; Department of Pathology, University of Hong Kong
| | | | | | | | | |
Collapse
|
44
|
Heinrich S, Geissen EM, Kamenz J, Trautmann S, Widmer C, Drewe P, Knop M, Radde N, Hasenauer J, Hauf S. Determinants of robustness in spindle assembly checkpoint signalling. Nat Cell Biol 2013; 15:1328-39. [DOI: 10.1038/ncb2864] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2013] [Accepted: 09/20/2013] [Indexed: 02/08/2023]
|