1
|
Peruffo A, Bassan I, Gonella A, Maccatrozzo L, Otero-Sabio C, Iannuzzi L, Perucatti A, Pistucci R, Giacomello M, Centelleghe C. Establishment and characterization of the Cuvier's beaked whale (Ziphius cavirostris) myogenic cell line. Res Vet Sci 2025; 182:105471. [PMID: 39608061 DOI: 10.1016/j.rvsc.2024.105471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 11/30/2024]
Abstract
In this study we present the first in vitro model based on a muscle-derived cell line from a male Cuvier's Beaked whale stranded along the Italian coastline. In the myogenic cell line at T0 stage we performed the growth rate assay, the karyotyping, the CBA chromosome banding, G banding, nucleolar organizer regions and telomere analysis and immunocytochemical analysis. The cell characterization at the T2 and T4 stage included: assessment of fusion index, the ultrastructural analysis and immunocytochemical analysis. The population doubling time was determined to be ∼54 h. The cell fusion index at the T2 stage was 8.2 +/- 5 %, at the T4 stage increased at index 28 +/- 10 %. The karyotype analysis revealed a 2n = 42, XY, two pairs of nucleolus organizer regions (NORs), characteristic CBA-banding and PNA-telomeric regions by FISH-mapping. The immunocytochemical results revealed that we generated a heterogeneous population of myogenic cells. A cell population express β-actin, myosin and vimentin and a sub-population of cells was desmin-positive. In details, at the T0 stage, both cell populations were undifferentiated mononucleated myoblast. At the T2 and T4 stage, the cells were capable of fusion in elongated multinucleated myotubes, probably resulting from the fusion of the myoblasts. This Cuvier's Beaked whale cell line, represents a new opportunity to better understand the physiological features of the cetacean Ziphius cavirostris myogenic cells. The possibility of setting up culture conditions that mimic the in vivo microenvironment, strengthen the importance of in vitro models for toxicological studies investigating water pollutants effects on cells.
Collapse
Affiliation(s)
- Antonella Peruffo
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, PD, Italy.
| | - Irene Bassan
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, PD, Italy
| | - Alice Gonella
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, PD, Italy
| | - Lisa Maccatrozzo
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, PD, Italy
| | - Cristina Otero-Sabio
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, PD, Italy
| | - Leopoldo Iannuzzi
- Institute of Animal Production System in Mediterranean Environment (ISPAAM), National Research Council (CNR), Portici, Naples, Italy
| | - Angela Perucatti
- Institute of Animal Production System in Mediterranean Environment (ISPAAM), National Research Council (CNR), Portici, Naples, Italy
| | - Ramona Pistucci
- Institute of Animal Production System in Mediterranean Environment (ISPAAM), National Research Council (CNR), Portici, Naples, Italy
| | | | - Cinzia Centelleghe
- Department of Comparative Biomedicine and Food Science, University of Padova, Legnaro, PD, Italy
| |
Collapse
|
2
|
Shen Y, Gao F, Wang Y, Wang Y, Zheng J, Gong J, Zhang J, Luo Z, Schindler D, Deng Y, Ding W, Lin T, Swidah R, Zhao H, Jiang S, Zeng C, Chen S, Chen T, Wang Y, Luo Y, Mitchell L, Bader JS, Zhang G, Shen X, Wang J, Fu X, Dai J, Boeke JD, Yang H, Xu X, Cai Y. Dissecting aneuploidy phenotypes by constructing Sc2.0 chromosome VII and SCRaMbLEing synthetic disomic yeast. CELL GENOMICS 2023; 3:100364. [PMID: 38020968 PMCID: PMC10667312 DOI: 10.1016/j.xgen.2023.100364] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 04/03/2023] [Accepted: 07/06/2023] [Indexed: 12/01/2023]
Abstract
Aneuploidy compromises genomic stability, often leading to embryo inviability, and is frequently associated with tumorigenesis and aging. Different aneuploid chromosome stoichiometries lead to distinct transcriptomic and phenotypic changes, making it helpful to study aneuploidy in tightly controlled genetic backgrounds. By deploying the engineered SCRaMbLE (synthetic chromosome rearrangement and modification by loxP-mediated evolution) system to the newly synthesized megabase Sc2.0 chromosome VII (synVII), we constructed a synthetic disomic yeast and screened hundreds of SCRaMbLEd derivatives with diverse chromosomal rearrangements. Phenotypic characterization and multi-omics analysis revealed that fitness defects associated with aneuploidy could be restored by (1) removing most of the chromosome content or (2) modifying specific regions in the duplicated chromosome. These findings indicate that both chromosome copy number and specific chromosomal regions contribute to the aneuploidy-related phenotypes, and the synthetic chromosome resource opens new paradigms in studying aneuploidy.
Collapse
Affiliation(s)
- Yue Shen
- BGI Research, Shenzhen 518083, China
- BGI Research, Changzhou 213299, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Feng Gao
- BGI Research, Shenzhen 518083, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| | - Yun Wang
- BGI Research, Shenzhen 518083, China
- BGI Research, Changzhou 213299, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
- University of Copenhagen, Universitetsparken 15, 2100 Copenhagen, Denmark
| | - Yuerong Wang
- BGI Research, Shenzhen 518083, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ju Zheng
- BGI Research, Shenzhen 518083, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| | | | | | - Zhouqing Luo
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Key Laboratory of Synthetic Genomics, Center for Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, China
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen 361102, China
| | - Daniel Schindler
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
- Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch-Strasse 10, 35043 Marburg, Germany
| | - Yang Deng
- BGI Research, Shenzhen 518083, China
| | - Weichao Ding
- BGI Research, Shenzhen 518083, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Lin
- BGI Research, Shenzhen 518083, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| | - Reem Swidah
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Hongcui Zhao
- BGI Research, Shenzhen 518083, China
- BGI Research, Changzhou 213299, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| | - Shuangying Jiang
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Key Laboratory of Synthetic Genomics, Center for Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, China
| | - Cheng Zeng
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Key Laboratory of Synthetic Genomics, Center for Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, China
| | | | - Tai Chen
- BGI Research, Shenzhen 518083, China
- BGI Research, Changzhou 213299, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| | - Yong Wang
- BGI Research, Shenzhen 518083, China
| | - Yisha Luo
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| | - Leslie Mitchell
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY 10016, USA
| | - Joel S. Bader
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Guojie Zhang
- University of Copenhagen, Universitetsparken 15, 2100 Copenhagen, Denmark
| | - Xia Shen
- Greater Bay Area Institute of Precision Medicine (Guangzhou), Fudan University, Guangzhou, China
- Center for Global Health Research, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Jian Wang
- BGI Research, Shenzhen 518083, China
| | - Xian Fu
- BGI Research, Shenzhen 518083, China
- BGI Research, Changzhou 213299, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| | - Junbiao Dai
- Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Key Laboratory of Synthetic Genomics, Center for Synthetic Genomics, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- College of Life Sciences and Oceanography, Shenzhen University, Shenzhen 518055, China
| | - Jef D. Boeke
- Institute for Systems Genetics and Department of Biochemistry and Molecular Pharmacology, NYU Langone Health, New York, NY 10016, USA
- Department of Biomedical Engineering, NYU Tandon School of Engineering, Brooklyn, NY 11201, USA
| | | | - Xun Xu
- BGI Research, Shenzhen 518083, China
- BGI Research, Changzhou 213299, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI-Shenzhen, Shenzhen 518120, China
| | - Yizhi Cai
- Manchester Institute of Biotechnology, University of Manchester, 131 Princess Street, Manchester M1 7DN, UK
| |
Collapse
|
3
|
Feng Q, Yu J, Yu J, Hu M, Gu L, Wang H, Du X, Zhu B, Cai M. Identification and Genome-Wide Gene Expression Perturbation of a Trisomy in Chinese Kale ( Brassica oleracea var. alboglabra). PLANTS (BASEL, SWITZERLAND) 2023; 12:3199. [PMID: 37765363 PMCID: PMC10536521 DOI: 10.3390/plants12183199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 08/29/2023] [Accepted: 09/06/2023] [Indexed: 09/29/2023]
Abstract
Trisomy harbouring an extra copy of the chromosome generally causes a variety of physical and intellectual disabilities in mammals but is an extremely rare and important genetic stock in plants. In this study, a spontaneous trisomy plant in a Chinese kale accession (Brassica oleracea var. alboglabra, CC, 2n = 18) that showed significantly smaller plant architecture when compared to other normal plants was found and subsequently confirmed by cytological analysis in which the chromosome set of 2n = 19 and abnormal chromosome behaviour were observed. Then, based on the gene expression deviation determined by RNA-seq, the extra chromosome copy in this trisomy was identified as chromosome C2 (TC2). Compared to normal plants, TC2 not only showed generally upregulated differentially expressed genes (DEGs) on chromosome C2 (97.21% of 573 DEGs in chromosome C2) but also exhibited a whole-genome expression perturbation, in which 1329 DEGs (69.87% of total DEGs) were observed along two-copy chromosomes (trans-effect). The genes in the high (gene expression value > 100) and medium (100 > gene expression value > 10) groups were more prone to decreased gene expression, but the genes in the low group (10 > gene expression value > 0.1) showed upregulated expression deviation. In addition, GO (Gene ontology) annotation analysis revealed that the upregulated DEGs in the trans-effect group were overrepresented by the genes involved in the response to stress category, while the downregulated DEGs in the trans-effect group were mostly enriched in pathways related to DNA synthesis. In conclusion, we think our results can provide important resources for genetic analysis in B. oleracea and show some novel insights for understanding trisomy plant biology.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Bin Zhu
- School of Life Sciences, Guizhou Normal University, Guiyang 550025, China; (Q.F.); (J.Y.); (J.Y.); (M.H.); (L.G.); (H.W.); (X.D.)
| | - Mengxian Cai
- School of Life Sciences, Guizhou Normal University, Guiyang 550025, China; (Q.F.); (J.Y.); (J.Y.); (M.H.); (L.G.); (H.W.); (X.D.)
| |
Collapse
|
4
|
Sun S, Liu K, Xue C, Hu Y, Yu H, Qi G, Chen J, Li X, Zhao X, Gong Z. Genome-Wide Effects on Gene Expression Between Parental and Filial Generations of Trisomy 11 and 12 of Rice. RICE (NEW YORK, N.Y.) 2023; 16:17. [PMID: 36964817 PMCID: PMC10039966 DOI: 10.1186/s12284-023-00632-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
Aneuploid refers to the gene dosage imbalance due to copy number alterations. Aneuploidy is generally harmful to the growth, development and reproduction of organisms according to the numerous research. However, it has rarely been reported on whether aneuploid have a relevant pattern of genome expression between the parental and its offspring generations. In this study, mRNA sequencing analysis was performed on rice (Oryza sativa L.) primary trisomes 11 and 12, same primary trisomes and normal individuals in their filial generation. We systematically summarized the changes in gene expression patterns that occur on cis genes and on trans genes between parental and filial generations. In T11 and T12, the ratio of cis-gene expression showed intermediate type in parents and dosage compensation in filial generations, which maybe due to more genes being downregulated. The trans genes were also affected by aneuploidy and manifested as cis-related. The strains with normal chromosomes in filial generations, there are still aneuploid-sensitive genes differentially expressed in their genomes, indicating that the effect of aneuploidy is far-reaching and could not be easily eliminated. Meanwhile, among these differentially expressed genes, genes with low-expression level were more likely to be upregulated, while genes with medium- and high-expression level were easy to be downregulated. For the different types of rice aneuploid, upregulated genes were mainly associated with genomic imbalance while downregulated genes were mainly influenced by the specific added chromosome. In conclusion, our results provide new insights into the genetic characterization and evolution of biological aneuploidy genomes.
Collapse
Affiliation(s)
- Shang Sun
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Key Laboratory of Plant Functional Genomics of the Ministry of Education/Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University, Yangzhou, 225009, China
| | - Kai Liu
- Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, 225009, China
| | - Chao Xue
- Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, 225009, China
| | - Yingying Hu
- Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, 225009, China
| | - Hengxiu Yu
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Key Laboratory of Plant Functional Genomics of the Ministry of Education/Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University, Yangzhou, 225009, China
| | - Guoxiao Qi
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Key Laboratory of Plant Functional Genomics of the Ministry of Education/Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University, Yangzhou, 225009, China
| | - Jijin Chen
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Key Laboratory of Plant Functional Genomics of the Ministry of Education/Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University, Yangzhou, 225009, China
| | - Xiya Li
- Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, 225009, China
| | - Xinru Zhao
- Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, 225009, China
| | - Zhiyun Gong
- Jiangsu Key Laboratory of Crop Genomics and Molecular Breeding/Key Laboratory of Plant Functional Genomics of the Ministry of Education/Jiangsu Key Laboratory of Crop Genetics and Physiology, Agricultural College of Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Co-Innovation Center for Modern Production Technology of Grain Crops, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
5
|
Andrews PW, Barbaric I, Benvenisty N, Draper JS, Ludwig T, Merkle FT, Sato Y, Spits C, Stacey GN, Wang H, Pera MF. The consequences of recurrent genetic and epigenetic variants in human pluripotent stem cells. Cell Stem Cell 2022; 29:1624-1636. [PMID: 36459966 DOI: 10.1016/j.stem.2022.11.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/08/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022]
Abstract
It is well established that human pluripotent stem cells (hPSCs) can acquire genetic and epigenetic changes during culture in vitro. Given the increasing use of hPSCs in research and therapy and the vast expansion in the number of hPSC lines available for researchers, the International Society for Stem Cell Research has recognized the need to reassess quality control standards for ensuring the genetic integrity of hPSCs. Here, we summarize current knowledge of the nature of recurrent genetic and epigenetic variants in hPSC culture, the methods for their detection, and what is known concerning their effects on cell behavior in vitro or in vivo. We argue that the potential consequences of low-level contamination of cell therapy products with cells bearing oncogenic variants are essentially unknown at present. We highlight the key challenges facing the field with particular reference to safety assessment of hPSC-derived cellular therapeutics.
Collapse
Affiliation(s)
- Peter W Andrews
- Centre for Stem Cell Biology, School of Biological Sciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK; Steering Committee, International Stem Cell Initiative
| | - Ivana Barbaric
- Centre for Stem Cell Biology, School of Biological Sciences, University of Sheffield, Western Bank, Sheffield, S10 2TN, UK; Steering Committee, International Stem Cell Initiative
| | - Nissim Benvenisty
- The Azrieli Center for Stem Cells and Genetic Research, Department of Genetics, Silberman Institute of Life Sciences, The Hebrew University of Jerusalem, Edmond J. Safra Campus, Givat Ram, Jerusalem 91904, Israel; Steering Committee, International Stem Cell Initiative
| | - Jonathan S Draper
- Stem Cell Network, 501 Smyth Road, Ottawa, ON, K1H 8L6, Canada; Steering Committee, International Stem Cell Initiative
| | - Tenneille Ludwig
- WiCell Research Institute, Madison, WI, USA; University of Wisconsin-Madison, Madison, WI 53719, USA; Steering Committee, International Stem Cell Initiative
| | - Florian T Merkle
- Wellcome Trust-Medical Research Council Institute of Metabolic Science, Wellcome Trust-Medical Research Council Cambridge Stem Cell Institute, University of Cambridge, Cambridge CB2 0QQ, UK; Steering Committee, International Stem Cell Initiative
| | - Yoji Sato
- Division of Cell-Based Therapeutic Products, National Institute of Health Sciences, 3-25-26 Tonomachi, Kawasaki Ward, Kawasaki City, Kanagawa 210-9501, Japan; Steering Committee, International Stem Cell Initiative
| | - Claudia Spits
- Research Group Reproduction and Genetics, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussels, Belgium; Steering Committee, International Stem Cell Initiative
| | - Glyn N Stacey
- International Stem Cell Banking Initiative, 2 High Street, Barley, UK; National Stem Cell Resource Centre, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100190, China; Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China; Steering Committee, International Stem Cell Initiative
| | - Haoyi Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, 100101, Beijing, China; Beijing Institute for Stem Cell and Regenerative Medicine, 100101, Beijing, China; Steering Committee, International Stem Cell Initiative
| | - Martin F Pera
- The Jackson Laboratory, 600 Main Street, Bar Harbor, ME 04609, USA; Steering Committee, International Stem Cell Initiative.
| |
Collapse
|
6
|
Satoh H, Arai Y, Furukawa E, Moriguchi T, Hama N, Urushidate T, Totoki Y, Kato M, Ohe Y, Yamamoto M, Shibata T. Genomic landscape of chemical-induced lung tumors under Nrf2 different expression levels. Carcinogenesis 2022; 43:613-623. [PMID: 35561328 DOI: 10.1093/carcin/bgac041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 04/19/2022] [Accepted: 05/12/2022] [Indexed: 11/14/2022] Open
Abstract
The transcription factor Nrf2 plays a crucial role in the anti-oxidative stress response, protection of DNA from injury, and DNA repair mechanisms. Nrf2 activity reduces cancer initiation, but how Nrf2 affects whole-genome alterations upon carcinogenic stimulus remains unexplored. Although recent genome-wide analysis using next-generation sequencing revealed landscapes of nucleotide mutations and copy number alterations in various human cancers, genomic changes in murine cancer models have not been thoroughly examined. We elucidated the relationship between Nrf2 expression levels and whole exon mutation patterns using an ethyl-carbamate (urethane)-induced lung carcinogenesis model employing Nrf2-deficient and Keap1-kd mice, the latter of which express high levels of Nrf2. Exome analysis demonstrated that single nucleotide and trinucleotide mutation patterns and the Kras mutational signature differed significantly and were dependent on the expression level of Nrf2. The Nrf2-deficient tumors exhibited fewer copy number alterations relative to the Nrf2-wt and Keap1-kd tumors. The observed trend in genomic alterations likely prevented the Nrf2-deficient tumors from progressing into malignancy. For the first time, we present whole-exome sequencing results for chemically-induced lung tumors in the Nrf2 gain or loss of function mouse models. Our results demonstrate that different Nrf2 expression levels lead to distinct gene mutation patterns that underly different oncogenic mechanisms in each tumor genotype.
Collapse
Affiliation(s)
- Hironori Satoh
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan.,Department of Respiratory Medicine, Pulmonary Center, National Cancer Center Hospital, Tokyo, Japan.,Division of Cancer Pathophysiology, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasuhito Arai
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Eisaku Furukawa
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan.,Division of Bioinformatics, National Cancer Center Research Institute, Tokyo, Japan
| | - Takashi Moriguchi
- Division of Medical Biochemistry, Tohoku Medical Pharmaceutical University, Sendai, Japan
| | - Natuko Hama
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Tomoko Urushidate
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yasushi Totoki
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Mamoru Kato
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan.,Division of Bioinformatics, National Cancer Center Research Institute, Tokyo, Japan
| | - Yuichiro Ohe
- Department of Respiratory Medicine, Pulmonary Center, National Cancer Center Hospital, Tokyo, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Graduate School of Medicine, Tohoku University, Sendai, Japan.,Department of Integrative Genomics, Tohoku Medical Megabank, Tohoku University, Sendai, Japan
| | - Tatsuhiro Shibata
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| |
Collapse
|
7
|
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
|
8
|
Impaired condensin complex and Aurora B kinase underlie mitotic and chromosomal defects in hyperdiploid B-cell ALL. Blood 2021; 136:313-327. [PMID: 32321174 DOI: 10.1182/blood.2019002538] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 03/27/2020] [Indexed: 12/14/2022] Open
Abstract
B-cell acute lymphoblastic leukemia (ALL; B-ALL) is the most common pediatric cancer, and high hyperdiploidy (HyperD) identifies the most common subtype of pediatric B-ALL. Despite HyperD being an initiating oncogenic event affiliated with childhood B-ALL, the mitotic and chromosomal defects associated with HyperD B-ALL (HyperD-ALL) remain poorly characterized. Here, we have used 54 primary pediatric B-ALL samples to characterize the cellular-molecular mechanisms underlying the mitotic/chromosome defects predicated to be early pathogenic contributors in HyperD-ALL. We report that HyperD-ALL blasts are low proliferative and show a delay in early mitosis at prometaphase, associated with chromosome-alignment defects at the metaphase plate leading to robust chromosome-segregation defects and nonmodal karyotypes. Mechanistically, biochemical, functional, and mass-spectrometry assays revealed that condensin complex is impaired in HyperD-ALL cells, leading to chromosome hypocondensation, loss of centromere stiffness, and mislocalization of the chromosome passenger complex proteins Aurora B kinase (AURKB) and Survivin in early mitosis. HyperD-ALL cells show chromatid cohesion defects and an impaired spindle assembly checkpoint (SAC), thus undergoing mitotic slippage due to defective AURKB and impaired SAC activity, downstream of condensin complex defects. Chromosome structure/condensation defects and hyperdiploidy were reproduced in healthy CD34+ stem/progenitor cells upon inhibition of AURKB and/or SAC. Collectively, hyperdiploid B-ALL is associated with a defective condensin complex, AURKB, and SAC.
Collapse
|
9
|
Hochwagen A, Berchowitz LE. Remembering Angelika Amon (1967–2020). J Cell Sci 2020. [DOI: 10.1242/jcs.257444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Affiliation(s)
| | - Luke E. Berchowitz
- Department of Genetics and Development, Columbia University Irving Medical Center, New York, NY 10032, USA
| |
Collapse
|
10
|
Andreychuk YV, Zadorsky SP, Zhuk AS, Stepchenkova EI, Inge-Vechtomov SG. Relationship between Type I and Type II Template Processes: Amyloids and Genome Stability. Mol Biol 2020. [DOI: 10.1134/s0026893320050027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
11
|
Gorter de Vries AR, Knibbe E, van Roosmalen R, van den Broek M, de la Torre Cortés P, O'Herne SF, Vijverberg PA, El Masoudi A, Brouwers N, Pronk JT, Daran JMG. Improving Industrially Relevant Phenotypic Traits by Engineering Chromosome Copy Number in Saccharomyces pastorianus. Front Genet 2020; 11:518. [PMID: 32582279 PMCID: PMC7283523 DOI: 10.3389/fgene.2020.00518] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 04/28/2020] [Indexed: 11/13/2022] Open
Abstract
The lager-brewing yeast Saccharomyces pastorianus is a hybrid between S. cerevisiae and S. eubayanus with an exceptional degree of aneuploidy. While chromosome copy number variation (CCNV) is present in many industrial Saccharomyces strains and has been linked to various industrially-relevant traits, its impact on the brewing performance of S. pastorianus remains elusive. Here we attempt to delete single copies of chromosomes which are relevant for the production of off-flavor compound diacetyl by centromere silencing. However, the engineered strains display CNV of multiple non-targeted chromosomes. We attribute this unintended CCNV to inherent instability and to a mutagenic effect of electroporation and of centromere-silencing. Regardless, the resulting strains displayed large phenotypic diversity. By growing centromere-silenced cells in repeated sequential batches in medium containing 10% ethanol, mutants with increased ethanol tolerance were obtained. By using CCNV mutagenesis by exposure to the mitotic inhibitor MBC, selection in the same set-up yielded even more tolerant mutants that would not classify as genetically modified organisms. These results show that CCNV of alloaneuploid S. pastorianus genomes is highly unstable, and that CCNV mutagenesis can generate broad diversity. Coupled to effective selection or screening, CCNV mutagenesis presents a potent tool for strain improvement.
Collapse
Affiliation(s)
| | - Ewout Knibbe
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | | | | | | | - Stephanie F O'Herne
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Pascal A Vijverberg
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Anissa El Masoudi
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Nick Brouwers
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Jack T Pronk
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| | - Jean-Marc G Daran
- Department of Biotechnology, Delft University of Technology, Delft, Netherlands
| |
Collapse
|
12
|
Schukken KM, Lin YC, Bakker PL, Schubert M, Preuss SF, Simon JE, van den Bos H, Storchova Z, Colomé-Tatché M, Bastians H, Spierings DC, Foijer F. Altering microtubule dynamics is synergistically toxic with spindle assembly checkpoint inhibition. Life Sci Alliance 2020; 3:3/2/e201900499. [PMID: 31980556 PMCID: PMC6985455 DOI: 10.26508/lsa.201900499] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 01/07/2020] [Accepted: 01/08/2020] [Indexed: 02/06/2023] Open
Abstract
Chromosomal instability (CIN) is a hallmark feature of cancer cells. In this study, Schukken and colleagues screen for compounds that selectively target CIN cells and identify an inhibitor of Src kinase to be selectively toxic for CIN cells. Chromosomal instability (CIN) and aneuploidy are hallmarks of cancer. As most cancers are aneuploid, targeting aneuploidy or CIN may be an effective way to target a broad spectrum of cancers. Here, we perform two small molecule compound screens to identify drugs that selectively target cells that are aneuploid or exhibit a CIN phenotype. We find that aneuploid cells are much more sensitive to the energy metabolism regulating drug ZLN005 than their euploid counterparts. Furthermore, cells with an ongoing CIN phenotype, induced by spindle assembly checkpoint (SAC) alleviation, are significantly more sensitive to the Src kinase inhibitor SKI606. We show that inhibiting Src kinase increases microtubule polymerization rates and, more generally, that deregulating microtubule polymerization rates is particularly toxic to cells with a defective SAC. Our findings, therefore, suggest that tumors with a dysfunctional SAC are particularly sensitive to microtubule poisons and, vice versa, that compounds alleviating the SAC provide a powerful means to treat tumors with deregulated microtubule dynamics.
Collapse
Affiliation(s)
- Klaske M Schukken
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Yu-Chih Lin
- Goettingen Center for Molecular Biosciences and University Medical Center, Goettingen, Germany
| | - Petra L Bakker
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Michael Schubert
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Stephanie F Preuss
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Judith E Simon
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Hilda van den Bos
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Zuzana Storchova
- Department of Molecular Genetics, University of Kaiserslautern, Germany
| | - Maria Colomé-Tatché
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands.,Institute of Computational Biology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany.,Technical University of Munich, School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Holger Bastians
- Goettingen Center for Molecular Biosciences and University Medical Center, Goettingen, Germany
| | - Diana Cj Spierings
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Centre Groningen, Groningen, The Netherlands
| |
Collapse
|
13
|
Chen Y, Chen S, Li K, Zhang Y, Huang X, Li T, Wu S, Wang Y, Carey LB, Qian W. Overdosage of Balanced Protein Complexes Reduces Proliferation Rate in Aneuploid Cells. Cell Syst 2019; 9:129-142.e5. [PMID: 31351919 DOI: 10.1016/j.cels.2019.06.007] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Revised: 02/27/2019] [Accepted: 06/17/2019] [Indexed: 11/26/2022]
Abstract
Cells with complex aneuploidies display a wide range of phenotypic abnormalities. However, the molecular basis for this has been mainly studied in trisomic (2n + 1) and disomic (n + 1) cells. To determine how karyotype affects proliferation in cells with complex aneuploidies, we generated 92 2n + x yeast strains in which each diploid cell has between 3 and 12 extra chromosomes. Genome-wide and, for individual protein complexes, proliferation defects are caused by the presence of protein complexes in which all subunits are balanced at the 3-copy level. Proteomics revealed that over 50% of 3-copy members of imbalanced complexes were expressed at only 2n protein levels, whereas members of complexes in which all subunits are stoichiometrically balanced at 3 copies per cell had 3n protein levels. We validated this finding using orthogonal datasets from yeast and from human cancers. Taken together, our study provides an explanation of how aneuploidy affects phenotype.
Collapse
Affiliation(s)
- Ying Chen
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Siyu Chen
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ke Li
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Yuliang Zhang
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiahe Huang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Ting Li
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Shaohuan Wu
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yingchun Wang
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China
| | - Lucas B Carey
- Department of Experimental and Health Sciences, Universitat Pompeu Fabra, Barcelona 08003, Spain; Center for Quantitative Biology and Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing 100871, China.
| | - Wenfeng Qian
- State Key Laboratory of Plant Genomics, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; Key Laboratory of Genetic Network Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100101, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| |
Collapse
|
14
|
Schukken KM, Foijer F. CIN and Aneuploidy: Different Concepts, Different Consequences. Bioessays 2017; 40. [PMID: 29160563 DOI: 10.1002/bies.201700147] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 10/10/2017] [Indexed: 12/19/2022]
Abstract
Chromosomal instability (CIN) and aneuploidy are similar concepts but not synonymous. CIN is the process that leads to chromosome copy number alterations, and aneuploidy is the result. While CIN and resulting aneuploidy often cause growth defects, they are also selected for in cancer cells. Although such contradicting fates may seem paradoxical at first, they can be better understood when CIN and aneuploidy are assessed separately, taking into account the in vitro or in vivo context, the rate of CIN, and severity of the aneuploid karyotype. As CIN can only be measured in living cells, which proves to be technically challenging in vivo, aneuploidy is more frequently quantified. However, CIN rates might be more predictive for tumor outcome than assessing aneuploidy rates alone. In reviewing the literature, we therefore conclude that there is an urgent need for new models in which we can monitor chromosome mis-segregation and its consequences in vivo. Also see the video abstract here: https://youtu.be/fL3LxZduchg.
Collapse
Affiliation(s)
- Klaske M Schukken
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
15
|
Davies AE, Kortright K, Kaplan KB. Adenomatous polyposis coli mutants dominantly activate Hsf1-dependent cell stress pathways through inhibition of microtubule dynamics. Oncotarget 2016; 6:25202-16. [PMID: 26320184 PMCID: PMC4694825 DOI: 10.18632/oncotarget.4513] [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: 02/23/2015] [Accepted: 07/10/2015] [Indexed: 01/13/2023] Open
Abstract
Cancer cells up-regulate cell stress pathways, including the protein chaperone Hsp90. Increases in Hsp90 are believed “buffer” mutant protein activities necessary for cancer phenotypes. Activation of the cell stress pathway also alters the transcriptional landscape of cells in ways that are critical for cancer progression. However, it is unclear when and how the cell stress pathway is de-regulated during cancer progression. Here we report that mutations in adenomatous polyposis coli (APC) found in colorectal cancer activate cell stress pathways in mouse intestinal crypt cells, prior to loss of heterozygosity at APC or to the appearance of canonical intestinal cancer markers. Hsp90 levels are elevated in normal APC heterozygote crypt cells and further elevated in non-cancer cells adjacent to dysplasias, suggesting that the Hsp90 stress pathway marks the “cancer-field” effect. Expression of mutant APC in normal human epithelial cells is sufficient to activate a cell stress pathway via perturbations in microtubule dynamics. Inhibition of microtubule dynamics is sufficient to activate an Hsf1-dependent increase in gene transcription and protein levels. We suggest that the early activation of this Hsf1 dependent cell stress pathway by mono-allelic mutations in APC can affect cell programming in a way that contributes to cancer onset.
Collapse
Affiliation(s)
- Alexander E Davies
- Department of Cell and Molecular Biology, University of California, Davis, CA, USA
| | - Kaitlyn Kortright
- Department of Cell and Molecular Biology, University of California, Davis, CA, USA
| | - Kenneth B Kaplan
- Department of Cell and Molecular Biology, University of California, Davis, CA, USA
| |
Collapse
|
16
|
Zhang CZ, Pellman D. From Mutational Mechanisms in Single Cells to Mutational Patterns in Cancer Genomes. COLD SPRING HARBOR SYMPOSIA ON QUANTITATIVE BIOLOGY 2016; 80:117-37. [PMID: 26968629 DOI: 10.1101/sqb.2015.80.027623] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Analysis of mutations in thousands of cancer genomes has revealed many characteristic patterns of mutagenesis. The search for the molecular mechanisms underlying these mutational patterns has not only generated novel biological insight but also led to the development of new experimental strategies to study cell-to-cell variation and genome evolution. In this essay, we discuss recent progress in the study of mutational mechanisms with a particular emphasis on the analysis of mutagenesis at the single-cell level.
Collapse
Affiliation(s)
- Cheng-Zhong Zhang
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215 Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115 Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142
| | - David Pellman
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts 02215 Department of Cell Biology, Harvard Medical School, Boston, Massachusetts 02115 Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142 Howard Hughes Medical Institute, Boston, Massachusetts 02115
| |
Collapse
|
17
|
Chromosomal Copy Number Variation in Saccharomyces pastorianus Is Evidence for Extensive Genome Dynamics in Industrial Lager Brewing Strains. Appl Environ Microbiol 2015; 81:6253-67. [PMID: 26150454 PMCID: PMC4542246 DOI: 10.1128/aem.01263-15] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/27/2015] [Indexed: 11/20/2022] Open
Abstract
Lager brewing strains of Saccharomyces pastorianus are natural interspecific hybrids originating from the spontaneous hybridization of Saccharomyces cerevisiae and Saccharomyces eubayanus. Over the past 500 years, S. pastorianus has been domesticated to become one of the most important industrial microorganisms. Production of lager-type beers requires a set of essential phenotypes, including the ability to ferment maltose and maltotriose at low temperature, the production of flavors and aromas, and the ability to flocculate. Understanding of the molecular basis of complex brewing-related phenotypic traits is a prerequisite for rational strain improvement. While genome sequences have been reported, the variability and dynamics of S. pastorianus genomes have not been investigated in detail. Here, using deep sequencing and chromosome copy number analysis, we showed that S. pastorianus strain CBS1483 exhibited extensive aneuploidy. This was confirmed by quantitative PCR and by flow cytometry. As a direct consequence of this aneuploidy, a massive number of sequence variants was identified, leading to at least 1,800 additional protein variants in S. pastorianus CBS1483. Analysis of eight additional S. pastorianus strains revealed that the previously defined group I strains showed comparable karyotypes, while group II strains showed large interstrain karyotypic variability. Comparison of three strains with nearly identical genome sequences revealed substantial chromosome copy number variation, which may contribute to strain-specific phenotypic traits. The observed variability of lager yeast genomes demonstrates that systematic linking of genotype to phenotype requires a three-dimensional genome analysis encompassing physical chromosomal structures, the copy number of individual chromosomes or chromosomal regions, and the allelic variation of copies of individual genes.
Collapse
|
18
|
Transcriptional Dynamics of Immortalized Human Mesenchymal Stem Cells during Transformation. PLoS One 2015; 10:e0126562. [PMID: 25978455 PMCID: PMC4433180 DOI: 10.1371/journal.pone.0126562] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Accepted: 04/03/2015] [Indexed: 12/23/2022] Open
Abstract
Comprehensive analysis of alterations in gene expression along with neoplastic transformation in human cells provides valuable information about the molecular mechanisms underlying transformation. To further address these questions, we performed whole transcriptome analysis to the human mesenchymal stem cell line, UE6E7T-3, which was immortalized with hTERT and human papillomavirus type 16 E6/E7 genes, in association with progress of transformation in these cells. At early stages of culture, UE6E7T-3 cells preferentially lost one copy of chromosome 13, as previously described; in addition, tumor suppressor genes, DNA repair genes, and apoptosis-activating genes were overexpressed. After the loss of chromosome 13, additional aneuploidy and genetic alterations that drove progressive transformation, were observed. At this stage, the cell line expressed oncogenes as well as genes related to anti-apoptotic functions, cell-cycle progression, and chromosome instability (CIN); these pro-tumorigenic changes were concomitant with a decrease in tumor suppressor gene expression. At later stages after prolong culture, the cells exhibited chromosome translocations, acquired anchorage-independent growth and tumorigenicity in nude mice, (sarcoma) and exhibited increased expression of genes encoding growth factor and DNA repair genes, and decreased expression of adhesion genes. In particular, glypican-5 (GPC5), which encodes a cell-surface proteoglycan that might be a biomarker for sarcoma, was expressed at high levels in association with transformation. Patched (Ptc1), the cell surface receptor for hedgehog (Hh) signaling, was also significantly overexpressed and co-localized with GPC5. Knockdown of GPC5 expression decreased cell proliferation, suggesting that it plays a key role in growth in U3-DT cells (transformants derived from UE6E7T-3 cells) through the Hh signaling pathway. Thus, the UE6E7T-3 cell culture model is a useful tool for assessing the functional contribution of genes showed by expression profiling to the neoplastic transformation of human fibroblasts and human mesenchymal stem cells (hMSC).
Collapse
|
19
|
Zadorsky SP, Sopova YV, Andreichuk DY, Startsev VA, Medvedeva VP, Inge-Vechtomov SG. Chromosome VIII disomy influences the nonsense suppression efficiency and transition metal tolerance of the yeast Saccharomyces cerevisiae. Yeast 2015; 32:479-97. [PMID: 25874850 DOI: 10.1002/yea.3074] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2014] [Revised: 02/24/2015] [Accepted: 03/27/2015] [Indexed: 01/26/2023] Open
Abstract
The SUP35 gene of the yeast Saccharomyces cerevisiae encodes the translation termination factor eRF3. Mutations in this gene lead to the suppression of nonsense mutations and a number of other pleiotropic phenotypes, one of which is impaired chromosome segregation during cell division. Similar effects result from replacing the S. cerevisiae SUP35 gene with its orthologues. A number of genetic and epigenetic changes that occur in the sup35 background result in partial compensation for this suppressor effect. In this study we showed that in S. cerevisiae strains in which the SUP35 orthologue from the yeast Pichia methanolica replaces the S. cerevisiae SUP35 gene, chromosome VIII disomy results in decreased efficiency of nonsense suppression. This antisuppressor effect is not associated with decreased stop codon read-through. We identified SBP1, a gene that localizes to chromosome VIII, as a dosage-dependent antisuppressor that strongly contributes to the overall antisuppressor effect of chromosome VIII disomy. Disomy of chromosome VIII also leads to a change in the yeast strains' tolerance of a number of transition metal salts.
Collapse
Affiliation(s)
- S P Zadorsky
- Department of Genetics and Biotechnology, St Petersburg State University, St Petersburg, Russian Federation.,St. Petersburg Branch Vavilov Institute of General Genetics, Russian Academy of Science, St Petersburg, Russian Federation
| | - Y V Sopova
- Department of Genetics and Biotechnology, St Petersburg State University, St Petersburg, Russian Federation.,St. Petersburg Branch Vavilov Institute of General Genetics, Russian Academy of Science, St Petersburg, Russian Federation
| | - D Y Andreichuk
- Department of Genetics and Biotechnology, St Petersburg State University, St Petersburg, Russian Federation
| | - V A Startsev
- Department of Genetics and Biotechnology, St Petersburg State University, St Petersburg, Russian Federation
| | - V P Medvedeva
- Department of Genetics and Biotechnology, St Petersburg State University, St Petersburg, Russian Federation
| | - S G Inge-Vechtomov
- Department of Genetics and Biotechnology, St Petersburg State University, St Petersburg, Russian Federation.,St. Petersburg Branch Vavilov Institute of General Genetics, Russian Academy of Science, St Petersburg, Russian Federation
| |
Collapse
|
20
|
Abnormal mitosis triggers p53-dependent cell cycle arrest in human tetraploid cells. Chromosoma 2013; 122:305-18. [DOI: 10.1007/s00412-013-0414-0] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 04/09/2013] [Accepted: 04/09/2013] [Indexed: 10/26/2022]
|
21
|
Abstract
Metastasis involves the spread of cancer cells from the primary tumor to surrounding tissues and to distant organs and is the primary cause of cancer morbidity and mortality. In order to complete the metastatic cascade, cancer cells must detach from the primary tumor, intravasate into the circulatory and lymphatic systems, evade immune attack, extravasate at distant capillary beds, and invade and proliferate in distant organs. Currently, several hypotheses have been advanced to explain the origin of cancer metastasis. These involve an epithelial mesenchymal transition, an accumulation of mutations in stem cells, a macrophage facilitation process, and a macrophage origin involving either transformation or fusion hybridization with neoplastic cells. Many of the properties of metastatic cancer cells are also seen in normal macrophages. A macrophage origin of metastasis can also explain the long-standing "seed and soil" hypothesis and the absence of metastasis in plant cancers. The view of metastasis as a macrophage metabolic disease can provide novel insight for therapeutic management.
Collapse
Affiliation(s)
- Thomas N Seyfried
- Department of Biology, Boston College, Chestnut Hill, Massachusetts 02467, USA.
| | | |
Collapse
|
22
|
Schuyler SC, Wu YF, Kuan VJW. The Mad1-Mad2 balancing act--a damaged spindle checkpoint in chromosome instability and cancer. J Cell Sci 2012; 125:4197-206. [PMID: 23093575 DOI: 10.1242/jcs.107037] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cancer cells are commonly aneuploid. The spindle checkpoint ensures accurate chromosome segregation by controlling cell cycle progression in response to aberrant microtubule-kinetochore attachment. Damage to the checkpoint, which is a partial loss or gain of checkpoint function, leads to aneuploidy during tumorigenesis. One form of damage is a change in levels of the checkpoint proteins mitotic arrest deficient 1 and 2 (Mad1 and Mad2), or in the Mad1:Mad2 ratio. Changes in Mad1 and Mad2 levels occur in human cancers, where their expression is regulated by the tumor suppressors p53 and retinoblastoma 1 (RB1). By employing a standard assay, namely the addition of a mitotic poison at mitotic entry, it has been shown that checkpoint function is normal in many cancer cells. However, in several experimental systems, it has been observed that this standard assay does not always reveal checkpoint aberrations induced by changes in Mad1 or Mad2, where excess Mad1 relative to Mad2 can lead to premature anaphase entry, and excess Mad2 can lead to a delay in entering anaphase. This Commentary highlights how changes in the levels of Mad1 and Mad2 result in a damaged spindle checkpoint, and explores how these changes cause chromosome instability that can lead to aneuploidy during tumorigenesis.
Collapse
Affiliation(s)
- Scott C Schuyler
- Department of Biomedical Sciences, College of Medicine, Chang Gung University, Kwei-Shan, Tao-Yuan, 333 Taiwan, Republic of China.
| | | | | |
Collapse
|
23
|
Akatsuka S, Yamashita Y, Ohara H, Liu YT, Izumiya M, Abe K, Ochiai M, Jiang L, Nagai H, Okazaki Y, Murakami H, Sekido Y, Arai E, Kanai Y, Hino O, Takahashi T, Nakagama H, Toyokuni S. Fenton reaction induced cancer in wild type rats recapitulates genomic alterations observed in human cancer. PLoS One 2012; 7:e43403. [PMID: 22952676 PMCID: PMC3430702 DOI: 10.1371/journal.pone.0043403] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2012] [Accepted: 07/19/2012] [Indexed: 11/19/2022] Open
Abstract
Iron overload has been associated with carcinogenesis in humans. Intraperitoneal administration of ferric nitrilotriacetate initiates a Fenton reaction in renal proximal tubules of rodents that ultimately leads to a high incidence of renal cell carcinoma (RCC) after repeated treatments. We performed high-resolution microarray comparative genomic hybridization to identify characteristics in the genomic profiles of this oxidative stress-induced rat RCCs. The results revealed extensive large-scale genomic alterations with a preference for deletions. Deletions and amplifications were numerous and sometimes fragmented, demonstrating that a Fenton reaction is a cause of such genomic alterations in vivo. Frequency plotting indicated that two of the most commonly altered loci corresponded to a Cdkn2a/2b deletion and a Met amplification. Tumor sizes were proportionally associated with Met expression and/or amplification, and clustering analysis confirmed our results. Furthermore, we developed a procedure to compare whole genomic patterns of the copy number alterations among different species based on chromosomal syntenic relationship. Patterns of the rat RCCs showed the strongest similarity to the human RCCs among five types of human cancers, followed by human malignant mesothelioma, an iron overload-associated cancer. Therefore, an iron-dependent Fenton chemical reaction causes large-scale genomic alterations during carcinogenesis, which may result in distinct genomic profiles. Based on the characteristics of extensive genome alterations in human cancer, our results suggest that this chemical reaction may play a major role during human carcinogenesis.
Collapse
Affiliation(s)
- Shinya Akatsuka
- Departments of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Yoriko Yamashita
- Departments of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hiroki Ohara
- Departments of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Yu-Ting Liu
- Department of Pathology and Biology of Diseases, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Masashi Izumiya
- Division of Cancer Development System, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Koichiro Abe
- Division of Cancer Development System, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
- Department of Internal Medicine, Teikyo University School of Medicine, Itabashi-ku, Tokyo, Japan
| | - Masako Ochiai
- Division of Cancer Development System, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Li Jiang
- Departments of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hirotaka Nagai
- Departments of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
- Department of Pathology and Biology of Diseases, Kyoto University Graduate School of Medicine, Sakyo-ku, Kyoto, Japan
| | - Yasumasa Okazaki
- Departments of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hideki Murakami
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Chikusa-Ku, Nagoya, Japan
| | - Yoshitaka Sekido
- Division of Molecular Oncology, Aichi Cancer Center Research Institute, Chikusa-Ku, Nagoya, Japan
| | - Eri Arai
- Division of Molecular Pathology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Yae Kanai
- Division of Molecular Pathology, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Okio Hino
- Department of Pathology and Oncology, Juntendo University School of Medicine, Bunkyo-ku, Tokyo, Japan
| | - Takashi Takahashi
- Molecular Carcinogenesis, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| | - Hitoshi Nakagama
- Division of Cancer Development System, National Cancer Center Research Institute, Chuo-ku, Tokyo, Japan
| | - Shinya Toyokuni
- Departments of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Showa-ku, Nagoya, Japan
| |
Collapse
|
24
|
Mannaert A, Downing T, Imamura H, Dujardin JC. Adaptive mechanisms in pathogens: universal aneuploidy in Leishmania. Trends Parasitol 2012; 28:370-6. [PMID: 22789456 DOI: 10.1016/j.pt.2012.06.003] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2012] [Revised: 06/14/2012] [Accepted: 06/14/2012] [Indexed: 02/07/2023]
Abstract
Genomic stability and maintenance of the correct chromosome number are assumed to be essential for normal development in eukaryotes. Aneuploidy is usually associated with severe abnormalities and decrease of cell fitness, but some organisms appear to rely on aneuploidy for rapid adaptation to changing environments. This phenomenon is mostly described in pathogenic fungi and cancer cells. However, recent genome studies highlight the importance of Leishmania as a new model for studies on aneuploidy. Several reports revealed extensive variation in chromosome copy number, indicating that aneuploidy is a constitutive feature of this protozoan parasite genus. Aneuploidy appears to be beneficial in organisms that are primarily asexual, unicellular, and that undergo sporadic epidemic expansions, including common pathogens as well as cancer.
Collapse
Affiliation(s)
- An Mannaert
- Unit of Molecular Parasitology, Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | | | | | | |
Collapse
|
25
|
Abstract
Deviation from a balanced genome by either gain or loss of entire chromosomes is generally tolerated poorly in all eukaryotic systems studied to date. Errors in mitotic or meiotic cell division lead to aneuploidy, which places a burden of additional or insufficient gene products from the missegregated chromosomes on the daughter cells. The burden of aneuploidy often manifests itself as impaired fitness of individual cells and whole organisms, in which abnormal development is also characteristic. However, most human cancers, noted for their rapid growth, also display various levels of aneuploidy. Here we discuss the detrimental, potentially beneficial, and sometimes puzzling effects of aneuploidy on cellular and organismal fitness and tissue function as well as its role in diseases such as cancer and neurodegeneration.
Collapse
Affiliation(s)
- Jake J Siegel
- David H. Koch Institute for Integrative Cancer Research and Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA.
| | | |
Collapse
|