1
|
Jyothy A, Hussain J, S SC, Chandraprabha VR, Nair MG, Vasudevan S, Sreedharan H, Abraham B, Maliekal TT, Natarajan K, Sengupta S. α-Fodrin-CENP-E interaction is critical for pancreatic cancer progression and drug response. Cell Cycle 2025:1-25. [PMID: 40211684 DOI: 10.1080/15384101.2025.2485837] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 04/15/2025] Open
Abstract
α-Fodrin, a known scaffolding protein for cytoskeleton stabilization, performs various functions including cell adhesion, cell motility, DNA repair and apoptosis. Based on our previous results revealing its role in mitosis in glioblastoma, we have examined its effect in pancreatic cancer, which is often linked to mitotic aberrations including aneuploidy and chromosome instability. Here, we show that the expression of α-Fodrin increases in pancreatic adenocarcinoma tissues compared to its normal counterpart, suggesting its tumor promoting role. shRNA-mediated knock-down of α-Fodrin significantly reduces the xenograft growth in immunocompromised mice underscoring the importance of α-Fodrin in tumor progression. CENP-E (centromere-associated protein E) is a motor protein essential for chromosomal alignment and segregation during mitosis. We have found that α-Fodrin interacts with CENP-E to recruit it to the kinetochore and depletion of α-Fodrin has a crucial role in controlling aneuploidy. As these mitotic defects can lead to apoptosis, we have further evaluated the activation of possible upstream pathways. Paclitaxel, a chemotherapeutic agent that stabilizes microtubules, disrupts mitosis and induces apoptosis. We found that Paclitaxel triggered stronger activation of JNK, ERK, and P38 MAPKs, altered BCL2/BAX ratios, cytochrome C release causing increased apoptosis in α-Fodrin knockdown cells compared to cells with wild-type α-Fodrin. This enhanced sensitivity to paclitaxel is consistent with improved survival in pancreatic cancer patients with low α-Fodrin (SPTAN1) and low CENP-E expression compared to poor prognosis with high expressions of both the genes. Taken together, this study provides the molecular mechanism by which α-Fodrin - CENP-E axis regulates pancreatic cancer progression and drug response.
Collapse
Affiliation(s)
- Athira Jyothy
- Department of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
- Department of Biotechnology, University of Kerala, Thiruvananthapuram, India
| | - Julfequar Hussain
- Department of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Sharanya C S
- Transdisciplinary Biology Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | | | - Madhumathy G Nair
- Division of Molecular Medicine, St. John's Research Institute, St John's Medical College, Bangalore, India
| | - Smreti Vasudevan
- Research Department, Rajiv Gandhi Cancer Institute and Research, Delhi, India
| | - Hariharan Sreedharan
- Laboratory of Cytogenetics and Molecular Diagnostics, Regional Cancer Centre, Thiruvananthapuram, India
| | - Betty Abraham
- Department of Pathology, DDRC SRL Diagnostics private limited, Thiruvananthapuram, India
| | - Tessy Thomas Maliekal
- Department of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Kathiresan Natarajan
- Transdisciplinary Biology Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| | - Suparna Sengupta
- Department of Cancer Research, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
2
|
Scarberry L, Thesing G, Brennan K, Williams M, Summers MK. p31 Comet Splice Variants Induce Distinct Spindle Assembly Checkpoint Dynamics due to Their Unique N-Termini. Int J Mol Sci 2025; 26:3089. [PMID: 40243841 PMCID: PMC11989133 DOI: 10.3390/ijms26073089] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/20/2025] [Accepted: 03/21/2025] [Indexed: 04/18/2025] Open
Abstract
The role of p31Comet in deactivating the spindle assembly checkpoint is well described in the literature; however, the data are all completed using Variant 2 of p31Comet. p31Comet is known to be expressed as two different splice variants: Variant 1 and Variant 2. Variant 1 contains an additional 32 N-terminal residues compared to Variant 2. We report that Variant 1 exhibits a reduced ability to bind to MAD2 and thus a reduced ability to induce mitotic progression. Additionally, we show that Variant 1 exhibits reduced stability compared to Variant 2. We further show that Variant 1 is uniquely expressed in the Testes, indicating a potentially unique role of Variant 1 in that organ. Overall, we demonstrate the N-terminus of p31Comet is capable of modulating p31Comet activity in mitosis.
Collapse
Affiliation(s)
- Luke Scarberry
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; (L.S.); (G.T.); (K.B.); (M.W.)
- Biomedical Sciences Graduate Program, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | - Garrett Thesing
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; (L.S.); (G.T.); (K.B.); (M.W.)
| | - Kevin Brennan
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; (L.S.); (G.T.); (K.B.); (M.W.)
| | - Madison Williams
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; (L.S.); (G.T.); (K.B.); (M.W.)
| | - Matthew K. Summers
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, OH 43210, USA; (L.S.); (G.T.); (K.B.); (M.W.)
| |
Collapse
|
3
|
Lu T, Yang J, Cai Y, Ding M, Yu Z, Fang X, Zhou X, Wang X. NCAPD3 promotes diffuse large B-cell lymphoma progression through modulating SIRT1 expression in an H3K9 monomethylation-dependent manner. J Adv Res 2025; 68:163-178. [PMID: 38432395 PMCID: PMC11785590 DOI: 10.1016/j.jare.2024.02.024] [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: 10/10/2023] [Revised: 01/31/2024] [Accepted: 02/29/2024] [Indexed: 03/05/2024] Open
Abstract
INTRODUCTION Condensin, a family of structural maintenance of chromosome complexes, has been shown to regulate chromosome compaction and segregation during mitosis. NCAPD3, a HEAT-repeat subunit of condensin II, plays a dominant role in condensin-mediated chromosome dynamics but remains unexplored in lymphoma. OBJECTIVES The study aims to unravel the molecular function and mechanism of NCAPD3 in diffuse large B-cell lymphoma (DLBCL). METHODS The expression and clinical significance of NCAPD3 were assessed in public database and clinical specimens. Chromosome spreads, co-immunoprecipitation (co-IP), mass spectrometry (MS), and chromatin immunoprecipitation (ChIP) assays were conducted to untangle the role and mechanism of NCAPD3 in DLBCL. RESULTS NCAPD3 was highly expressed in DLBCL, correlated with poor prognosis. NCAPD3 deficiency impeded cell proliferation, induced apoptosis and increased the chemosensitivity. Instead, NCAPD3 overexpression facilitated cell proliferation. In vivo experiments further indicated targeting NCAPD3 suppressed tumor growth. Noteworthily, NCAPD3 deficiency disturbed the mitosis, triggering the formation of aneuploids. To reveal the function of NCAPD3 in DLBCL, chromosome spreads were conducted, presenting that chromosomes became compact upon NCAPD3 overexpression, instead, loose, twisted and lacking axial rigidity upon NCAPD3 absence. Meanwhile, the classical transcription-activated marker, H3K4 trimethylation, was found globally upregulated after NCAPD3 knockout, suggesting that NCAPD3 might participate in chromatin remodeling and transcription regulation. MS revealed NCAPD3 could interact with transcription factor, TFII I. Further co-IP and ChIP assays verified NCAPD3 could be anchored at the promoter of SIRT1 by TFII I and then supported the transcription of SIRT1 via recognizing H3K9 monomethylation (H3K9me1) on SIRT1 promoter. Function reversion assay verified the oncogenic role of NCAPD3 in DLBCL was partially mediated by SIRT1. CONCLUSION This study demonstrated that dysregulation of NCAPD3 could disturb chromosome compaction and segregation and regulate the transcription activity of SIRT1 in an H3K9me1-dependent manner, which provided novel insights into targeted strategy for DLBCL.
Collapse
Affiliation(s)
- Tiange Lu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Juan Yang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China
| | - Yiqing Cai
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Mengfei Ding
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Zhuoya Yu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Xiaosheng Fang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| | - Xiangxiang Zhou
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China; Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China; Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China; Branch of National Clinical Research Center for Hematologic Diseases, Jinan, Shandong 250021, China; National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou 251006, China.
| |
Collapse
|
4
|
Chen J, Wu S, He JJ, Liu YP, Deng ZY, Fang HK, Chen JF, Wei YL, She ZY. Kinesin-7 CENP-E mediates centrosome organization and spindle assembly to regulate chromosome alignment and genome stability. Cell Prolif 2025; 58:e13745. [PMID: 39266203 DOI: 10.1111/cpr.13745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/14/2024] Open
Abstract
Chromosome congression and alignment are essential for cell cycle progression and genomic stability. Kinesin-7 CENP-E, a plus-end-directed kinesin motor, is required for chromosome biorientation, congression and alignment in cell division. However, it remains unclear how chromosomes are aligned and segregated in the absence of CENP-E in mitosis. In this study, we utilize the CRISPR-Cas9 gene editing method and high-throughput screening to establish CENP-E knockout cell lines and reveal that CENP-E deletion results in defects in chromosome congression, alignment and segregation, which further promotes aneuploidy and genomic instability in mitosis. Both CENP-E inhibition and deletion lead to the dispersion of spindle poles, the formation of the multipolar spindle and spindle disorganization, which indicates that CENP-E is necessary for the organization and maintenance of spindle poles. In addition, CENP-E heterozygous deletion in spleen tissues also leads to the accumulation of dividing lymphocytes and cell cycle arrest in vivo. Furthermore, CENP-E deletion also disrupts the localization of key kinetochore proteins and triggers the activation of the spindle assembly checkpoint. In summary, our findings demonstrate that CENP-E promotes kinetochore-microtubule attachment and spindle pole organization to regulate chromosome alignment and spindle assembly checkpoint during cell division.
Collapse
Affiliation(s)
- Jie Chen
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Shan Wu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Jie-Jie He
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Yu-Peng Liu
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Zhao-Yang Deng
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Han-Kai Fang
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Jian-Fan Chen
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| | - Ya-Lan Wei
- Medical Research Center, Fujian Maternity and Child Health Hospital, Fuzhou, Fujian, China
- College of Clinical Medicine for Obstetrics & Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Zhen-Yu She
- Department of Cell Biology and Genetics, The School of Basic Medical Sciences, Fujian Medical University, Fuzhou, Fujian, China
- Key Laboratory of Stem Cell Engineering and Regenerative Medicine, Fujian Province University, Fuzhou, Fujian, China
| |
Collapse
|
5
|
Malumbres M, Villarroya-Beltri C. Mosaic variegated aneuploidy in development, ageing and cancer. Nat Rev Genet 2024; 25:864-878. [PMID: 39169218 DOI: 10.1038/s41576-024-00762-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/27/2024] [Indexed: 08/23/2024]
Abstract
Mosaic variegated aneuploidy (MVA) is a rare condition in which abnormal chromosome counts (that is, aneuploidies), affecting different chromosomes in each cell (making it variegated) are found only in a certain number of cells (making it mosaic). MVA is characterized by various developmental defects and, despite its rarity, presents a unique clinical scenario to understand the consequences of chromosomal instability and copy number variation in humans. Research from patients with MVA, genetically engineered mouse models and functional cellular studies have found the genetic causes to be mutations in components of the spindle-assembly checkpoint as well as in related proteins involved in centrosome dynamics during mitosis. MVA is accompanied by tumour susceptibility (depending on the genetic basis) as well as cellular and systemic stress, including chronic immune response and the associated clinical implications.
Collapse
Affiliation(s)
- Marcos Malumbres
- Cancer Cell Cycle Group, Systems Oncology Program, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.
- Cell Division and Cancer Group, Spanish National Cancer Research Centre (CNIO) Madrid, Madrid, Spain.
- Catalan Institution for Research and Advanced Studies (ICREA) Barcelona, Barcelona, Spain.
| | | |
Collapse
|
6
|
Campos Gudiño R, Neudorf NM, Andromidas D, Lichtensztejn Z, McManus KJ. Loss of EMI1 compromises chromosome stability and is associated with cellular transformation in colonic epithelial cell contexts. Br J Cancer 2024; 131:1516-1528. [PMID: 39358461 PMCID: PMC11519589 DOI: 10.1038/s41416-024-02855-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is still a leading cause of cancer deaths worldwide. Thus, identifying the aberrant genes and proteins underlying disease pathogenesis is critical to improve early detection methods and develop novel therapeutic strategies. Chromosome instability (CIN), or ongoing changes in chromosome complements, is a predominant form of genome instability. It is a driver of genetic heterogeneity found in ~85% of CRCs. Although CIN contributes to CRC pathogenesis, the molecular determinants underlying CIN remain poorly understood. Recently, EMI1, an F-box protein, was identified as a candidate CIN gene. In this study, we sought to determine the impact reduced EMI1 expression has on CIN and cellular transformation. METHODS Coupling siRNA-based silencing and CRISPR/Cas9 knockout clones with quantitative imaging microscopy we evaluated the impact reduced EMI1 expression has on CIN and cellular transformation in four colonic epithelial cell contexts. RESULTS Quantitative imaging microscopy data revealed that reduced EMI1 expression induces increases in CIN phenotypes in both transient (siRNA) and constitutive (CRISPR/Cas9) cell models that are associated with increases in DNA damage and cellular transformation phenotypes in long-term studies. CONCLUSIONS This study determined that reduced EMI1 expression induces CIN and promotes cellular transformation, which is consistent with a role in early CRC development.
Collapse
Affiliation(s)
- Rubi Campos Gudiño
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Nicole M Neudorf
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
| | - Demi Andromidas
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Zelda Lichtensztejn
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Kirk J McManus
- Paul Albrechtsen Research Institute, CancerCare Manitoba, Winnipeg, MB, Canada.
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
| |
Collapse
|
7
|
Cosper PF, Paracha M, Jones KM, Hrycyniak L, Henderson L, Bryan A, Eyzaguirre D, McCunn E, Boulanger E, Wan J, Nickel KP, Horner V, Hu R, Harari PM, Kimple RJ, Weaver BA. Chromosomal instability increases radiation sensitivity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.13.612942. [PMID: 39345631 PMCID: PMC11429890 DOI: 10.1101/2024.09.13.612942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Continuous chromosome missegregation over successive mitotic divisions, known as chromosomal instability (CIN), is common in cancer. Increasing CIN above a maximally tolerated threshold leads to cell death due to loss of essential chromosomes. Here, we show in two tissue contexts that otherwise isogenic cancer cells with higher levels of CIN are more sensitive to ionizing radiation, which itself induces CIN. CIN also sensitizes HPV-positive and HPV-negative head and neck cancer patient derived xenograft (PDX) tumors to radiation. Moreover, laryngeal cancers with higher CIN prior to treatment show improved response to radiation therapy. In addition, we reveal a novel mechanism of radiosensitization by docetaxel, a microtubule stabilizing drug commonly used in combination with radiation. Docetaxel causes cell death by inducing CIN due to abnormal multipolar spindles rather than causing mitotic arrest, as previously assumed. Docetaxel-induced CIN, rather than mitotic arrest, is responsible for the enhanced radiation sensitivity observed in vitro and in vivo, challenging the mechanistic dogma of the last 40 years. These results implicate CIN as a potential biomarker and inducer of radiation response, which could provide valuable cancer therapeutic opportunities. Statement of Significance Cancer cells and laryngeal tumors with higher chromosome missegregation rates are more sensitive to radiation therapy, supporting chromosomal instability as a promising biomarker of radiation response.
Collapse
Affiliation(s)
- Pippa F. Cosper
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Maha Paracha
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kathryn M. Jones
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Laura Hrycyniak
- Molecular and Cellular Pharmacology Graduate Training Program, University of Wisconsin, Madison, WI 53705, USA
| | - Les Henderson
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, WI
| | - Ava Bryan
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Diego Eyzaguirre
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Emily McCunn
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Elizabeth Boulanger
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jun Wan
- Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Kwangok P. Nickel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Vanessa Horner
- Wisconsin State Laboratory of Hygiene, University of Wisconsin, Madison, WI
| | - Rong Hu
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI
| | - Paul M. Harari
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Randall J. Kimple
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI 53705, USA
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Beth A. Weaver
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI 53705, USA
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI 53705, USA
| |
Collapse
|
8
|
LaBella KA, Hsu WH, Li J, Qi Y, Liu Y, Liu J, Wu CC, Liu Y, Song Z, Lin Y, Blecher JM, Jiang S, Shang X, Han J, Spring DJ, Zhang J, Xia Y, DePinho RA. Telomere dysfunction alters intestinal stem cell dynamics to promote cancer. Dev Cell 2024; 59:1475-1486.e5. [PMID: 38574731 PMCID: PMC11379129 DOI: 10.1016/j.devcel.2024.03.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2023] [Revised: 01/22/2024] [Accepted: 03/11/2024] [Indexed: 04/06/2024]
Abstract
Telomere dynamics are linked to aging hallmarks, and age-associated telomere loss fuels the development of epithelial cancers. In Apc-mutant mice, the onset of DNA damage associated with telomere dysfunction has been shown to accelerate adenoma initiation via unknown mechanisms. Here, we observed that Apc-mutant mice engineered to experience telomere dysfunction show accelerated adenoma formation resulting from augmented cell competition and clonal expansion. Mechanistically, telomere dysfunction induces the repression of EZH2, resulting in the derepression of Wnt antagonists, which causes the differentiation of adjacent stem cells and a relative growth advantage to Apc-deficient telomere dysfunctional cells. Correspondingly, in this mouse model, GSK3β inhibition countered the actions of Wnt antagonists on intestinal stem cells, resulting in impaired adenoma formation of telomere dysfunctional Apc-mutant cells. Thus, telomere dysfunction contributes to cancer initiation through altered stem cell dynamics, identifying an interception strategy for human APC-mutant cancers with shortened telomeres.
Collapse
Affiliation(s)
- Kyle A LaBella
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Wen-Hao Hsu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jiexi Li
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yutao Qi
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yonghong Liu
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jingjing Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Chia-Chin Wu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yang Liu
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Zingzhi Song
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yiyun Lin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jonathan M Blecher
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Shan Jiang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Xiaoying Shang
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jincheng Han
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Denise J Spring
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Jianhua Zhang
- Department of Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Yan Xia
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Ronald A DePinho
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
9
|
Wang W, Zhou X, Kong L, Pan Z, Chen G. BUB1 Promotes Gemcitabine Resistance in Pancreatic Cancer Cells by Inhibiting Ferroptosis. Cancers (Basel) 2024; 16:1540. [PMID: 38672622 PMCID: PMC11048608 DOI: 10.3390/cancers16081540] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 03/31/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
The development of chemotherapy resistance severely limits the therapeutic efficacy of gemcitabine (GEM) in pancreatic cancer (PC), and the dysregulation of ferroptosis is a crucial factor in the development of chemotherapy resistance. BUB1 Mitotic Checkpoint Serine/Threonine Kinase (BUB1) is highly overexpressed in PC patients and is closely associated with patient prognosis. However, none of the literature reports the connection between BUB1 and ferroptosis. The molecular mechanisms underlying GEM resistance are also not well understood. Therefore, this study first established the high expression levels of BUB1 in PC patients, then explored the role of BUB1 in the process of ferroptosis, and finally investigated the mechanisms by which BUB1 regulates ferroptosis and contributes to GEM resistance in PC cells. In this study, downregulation of BUB1 enhanced the sensitivity of PC cells to Erastin, and inhibited cell proliferation and migration. Mechanistically, BUB1 could inhibit the expression levels of Neurofibromin 2 (NF2) and MOB kinase activator 1 (MOB1), and promote Yes-associated protein (YAP) expression, thereby inhibiting ferroptosis and promoting GEM resistance in PC cells. Furthermore, the combination of BUB1 inhibition with GEM exhibited a synergistic therapeutic effect. These findings reveal the mechanisms underlying the development of GEM chemotherapy resistance based on ferroptosis and suggest that the combined use of BUB1 inhibitors may be an effective approach to enhance GEM efficacy.
Collapse
Affiliation(s)
- Weiming Wang
- Department of Hepato-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (W.W.); (L.K.); (Z.P.)
| | - Xiang Zhou
- Department of Breast Cancer, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou 325035, China;
| | - Lingming Kong
- Department of Hepato-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (W.W.); (L.K.); (Z.P.)
| | - Zhenyan Pan
- Department of Hepato-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (W.W.); (L.K.); (Z.P.)
| | - Gang Chen
- Department of Hepato-Pancreatic Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China; (W.W.); (L.K.); (Z.P.)
| |
Collapse
|
10
|
Hosea R, Hillary S, Naqvi S, Wu S, Kasim V. The two sides of chromosomal instability: drivers and brakes in cancer. Signal Transduct Target Ther 2024; 9:75. [PMID: 38553459 PMCID: PMC10980778 DOI: 10.1038/s41392-024-01767-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/18/2024] [Accepted: 02/06/2024] [Indexed: 04/02/2024] Open
Abstract
Chromosomal instability (CIN) is a hallmark of cancer and is associated with tumor cell malignancy. CIN triggers a chain reaction in cells leading to chromosomal abnormalities, including deviations from the normal chromosome number or structural changes in chromosomes. CIN arises from errors in DNA replication and chromosome segregation during cell division, leading to the formation of cells with abnormal number and/or structure of chromosomes. Errors in DNA replication result from abnormal replication licensing as well as replication stress, such as double-strand breaks and stalled replication forks; meanwhile, errors in chromosome segregation stem from defects in chromosome segregation machinery, including centrosome amplification, erroneous microtubule-kinetochore attachments, spindle assembly checkpoint, or defective sister chromatids cohesion. In normal cells, CIN is deleterious and is associated with DNA damage, proteotoxic stress, metabolic alteration, cell cycle arrest, and senescence. Paradoxically, despite these negative consequences, CIN is one of the hallmarks of cancer found in over 90% of solid tumors and in blood cancers. Furthermore, CIN could endow tumors with enhanced adaptation capabilities due to increased intratumor heterogeneity, thereby facilitating adaptive resistance to therapies; however, excessive CIN could induce tumor cells death, leading to the "just-right" model for CIN in tumors. Elucidating the complex nature of CIN is crucial for understanding the dynamics of tumorigenesis and for developing effective anti-tumor treatments. This review provides an overview of causes and consequences of CIN, as well as the paradox of CIN, a phenomenon that continues to perplex researchers. Finally, this review explores the potential of CIN-based anti-tumor therapy.
Collapse
Affiliation(s)
- Rendy Hosea
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sharon Hillary
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Sumera Naqvi
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Shourong Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| | - Vivi Kasim
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400045, China.
- The 111 Project Laboratory of Biomechanics and Tissue Repair, College of Bioengineering, Chongqing University, Chongqing, 400044, China.
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
11
|
Marques JF, Kops GJPL. Permission to pass: on the role of p53 as a gatekeeper for aneuploidy. Chromosome Res 2023; 31:31. [PMID: 37864038 PMCID: PMC10589155 DOI: 10.1007/s10577-023-09741-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 10/22/2023]
Abstract
Aneuploidy-the karyotype state in which the number of chromosomes deviates from a multiple of the haploid chromosome set-is common in cancer, where it is thought to facilitate tumor initiation and progression. However, it is poorly tolerated in healthy cells: during development and tissue homeostasis, aneuploid cells are efficiently cleared from the population. It is still largely unknown how cancer cells become, and adapt to being, aneuploid. P53, the gatekeeper of the genome, has been proposed to guard against aneuploidy. Aneuploidy in cancer genomes strongly correlates with mutations in TP53, and p53 is thought to prevent the propagation of aneuploid cells. Whether p53 also participates in preventing the mistakes in cell division that lead to aneuploidy is still under debate. In this review, we summarize the current understanding of the role of p53 in protecting cells from aneuploidy, and we explore the consequences of functional p53 loss for the propagation of aneuploidy in cancer.
Collapse
Affiliation(s)
- Joana F Marques
- Royal Netherlands Academy of Arts and Sciences (KNAW), Hubrecht Institute, Uppsalalaan 8, 3584CT, Utrecht, the Netherlands
- University Medical Center Utrecht, Heidelberglaan 100, 3584CX, Utrecht, the Netherlands
- Oncode Institute, Jaarbeursplein 6, 3521AL, Utrecht, the Netherlands
| | - Geert J P L Kops
- Royal Netherlands Academy of Arts and Sciences (KNAW), Hubrecht Institute, Uppsalalaan 8, 3584CT, Utrecht, the Netherlands.
- University Medical Center Utrecht, Heidelberglaan 100, 3584CX, Utrecht, the Netherlands.
- Oncode Institute, Jaarbeursplein 6, 3521AL, Utrecht, the Netherlands.
| |
Collapse
|
12
|
Zhou AS, Tucker JB, Scribano CM, Lynch AR, Carlsen CL, Pop-Vicas ST, Pattaswamy SM, Burkard ME, Weaver BA. Diverse microtubule-targeted anticancer agents kill cells by inducing chromosome missegregation on multipolar spindles. PLoS Biol 2023; 21:e3002339. [PMID: 37883329 PMCID: PMC10602348 DOI: 10.1371/journal.pbio.3002339] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/18/2023] [Indexed: 10/28/2023] Open
Abstract
Microtubule-targeted agents are commonly used for cancer treatment, though many patients do not benefit. Microtubule-targeted drugs were assumed to elicit anticancer activity via mitotic arrest because they cause cell death following mitotic arrest in cell culture. However, we recently demonstrated that intratumoral paclitaxel concentrations are insufficient to induce mitotic arrest and rather induce chromosomal instability (CIN) via multipolar mitotic spindles. Here, we show in metastatic breast cancer and relevant human cellular models that this mechanism is conserved among clinically useful microtubule poisons. While multipolar divisions typically produce inviable progeny, multipolar spindles can be focused into near-normal bipolar spindles at any stage of mitosis. Using a novel method to quantify the rate of CIN, we demonstrate that cell death positively correlates with net loss of DNA. Spindle focusing decreases CIN and causes resistance to diverse microtubule poisons, which can be counteracted by addition of a drug that increases CIN without affecting spindle polarity. These results demonstrate conserved mechanisms of action and resistance for diverse microtubule-targeted agents. Trial registration: clinicaltrials.gov, NCT03393741.
Collapse
Affiliation(s)
- Amber S. Zhou
- Molecular and Cellular Pharmacology Graduate Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - John B. Tucker
- Cancer Biology Graduate Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Christina M. Scribano
- Molecular and Cellular Pharmacology Graduate Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Andrew R. Lynch
- Cellular and Molecular Pathology Graduate Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Caleb L. Carlsen
- Cellular and Molecular Biology Graduate Training Program, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Sophia T. Pop-Vicas
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Srishrika M. Pattaswamy
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Mark E. Burkard
- Department of Medicine, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, United States of America
| | - Beth A. Weaver
- Department of Cell and Regenerative Biology, University of Wisconsin, Madison, Wisconsin, United States of America
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin, Madison, Wisconsin, United States of America
- Carbone Cancer Center, University of Wisconsin, Madison, Wisconsin, United States of America
| |
Collapse
|
13
|
Truong MA, Cané-Gasull P, Lens SMA. Modeling specific aneuploidies: from karyotype manipulations to biological insights. Chromosome Res 2023; 31:25. [PMID: 37640903 PMCID: PMC10462580 DOI: 10.1007/s10577-023-09735-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/11/2023] [Accepted: 08/09/2023] [Indexed: 08/31/2023]
Abstract
An abnormal chromosome number, or aneuploidy, underlies developmental disorders and is a common feature of cancer, with different cancer types exhibiting distinct patterns of chromosomal gains and losses. To understand how specific aneuploidies emerge in certain tissues and how they contribute to disease development, various methods have been developed to alter the karyotype of mammalian cells and mice. In this review, we provide an overview of both classic and novel strategies for inducing or selecting specific chromosomal gains and losses in human and murine cell systems. We highlight how these customized aneuploidy models helped expanding our knowledge of the consequences of specific aneuploidies to (cancer) cell physiology.
Collapse
Affiliation(s)
- My Anh Truong
- Oncode Institute and Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584, CG, Utrecht, The Netherlands
| | - Paula Cané-Gasull
- Oncode Institute and Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584, CG, Utrecht, The Netherlands
| | - Susanne M A Lens
- Oncode Institute and Center for Molecular Medicine, University Medical Center Utrecht, Universiteitsweg 100, 3584, CG, Utrecht, The Netherlands.
| |
Collapse
|
14
|
Cosper PF, Hrycyniak LCF, Paracha M, Lee DL, Wan J, Jones K, Bice SA, Nickel K, Mallick S, Taylor AM, Kimple RJ, Lambert PF, Weaver BA. HPV16 E6 induces chromosomal instability due to polar chromosomes caused by E6AP-dependent degradation of the mitotic kinesin CENP-E. Proc Natl Acad Sci U S A 2023; 120:e2216700120. [PMID: 36989302 PMCID: PMC10083562 DOI: 10.1073/pnas.2216700120] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 02/20/2023] [Indexed: 03/30/2023] Open
Abstract
Chromosome segregation during mitosis is highly regulated to ensure production of genetically identical progeny. Recurrent mitotic errors cause chromosomal instability (CIN), a hallmark of tumors. The E6 and E7 oncoproteins of high-risk human papillomavirus (HPV), which causes cervical, anal, and head and neck cancers (HNC), cause mitotic defects consistent with CIN in models of anogenital cancers, but this has not been studied in the context of HNC. Here, we show that HPV16 induces a specific type of CIN in patient HNC tumors, patient-derived xenografts, and cell lines, which is due to defects in chromosome congression. These defects are specifically induced by the HPV16 oncogene E6 rather than E7. We show that HPV16 E6 expression causes degradation of the mitotic kinesin CENP-E, whose depletion produces chromosomes that are chronically misaligned near spindle poles (polar chromosomes) and fail to congress. Though the canonical oncogenic role of E6 is the degradation of the tumor suppressor p53, CENP-E degradation and polar chromosomes occur independently of p53. Instead, E6 directs CENP-E degradation in a proteasome-dependent manner via the E6-associated ubiquitin protein ligase E6AP/UBE3A. This study reveals a mechanism by which HPV induces CIN, which may impact HPV-mediated tumor initiation, progression, and therapeutic response.
Collapse
Affiliation(s)
- Pippa F. Cosper
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI53705
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI53705
| | - Laura C. F. Hrycyniak
- Molecular and Cellular Pharmacology Graduate Training Program, University of Wisconsin-Madison, Madison, WI53705
| | - Maha Paracha
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI53705
| | - Denis L. Lee
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI53705
| | - Jun Wan
- Physiology Graduate Training Program, University of Wisconsin-Madison, Madison, WI53705
| | - Kathryn Jones
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI53705
| | - Sophie A. Bice
- University of Wisconsin School of Medicine and Public Health, Madison, WI53705
| | - Kwangok Nickel
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI53705
| | - Samyukta Mallick
- Department of Pathology and Cell Biology at the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
- Integrated Program in Cellular, Molecular, and Biomedical Studies, Columbia University, New York, NY10032
| | - Alison M. Taylor
- Department of Pathology and Cell Biology at the Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY10032
| | - Randall J. Kimple
- Department of Human Oncology, University of Wisconsin-Madison, Madison, WI53705
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI53705
| | - Paul F. Lambert
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI53705
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI53705
| | - Beth A. Weaver
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI53705
- Department of Oncology, McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, WI53705
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, WI53705
| |
Collapse
|
15
|
Aberrant HMGA2 Expression Sustains Genome Instability That Promotes Metastasis and Therapeutic Resistance in Colorectal Cancer. Cancers (Basel) 2023; 15:cancers15061735. [PMID: 36980621 PMCID: PMC10046046 DOI: 10.3390/cancers15061735] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/06/2023] [Accepted: 03/11/2023] [Indexed: 03/16/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most lethal cancers worldwide, accounting for nearly ~10% of all cancer diagnoses and deaths. Current therapeutic approaches have considerably increased survival for patients diagnosed at early stages; however, ~20% of CRC patients are diagnosed with late-stage, metastatic CRC, where 5-year survival rates drop to 6–13% and treatment options are limited. Genome instability is an enabling hallmark of cancer that confers increased acquisition of genetic alterations, mutations, copy number variations and chromosomal rearrangements. In that regard, research has shown a clear association between genome instability and CRC, as the accumulation of aberrations in cancer-related genes provides subpopulations of cells with several advantages, such as increased proliferation rates, metastatic potential and therapeutic resistance. Although numerous genes have been associated with CRC, few have been validated as predictive biomarkers of metastasis or therapeutic resistance. A growing body of evidence suggests a member of the High-Mobility Group A (HMGA) gene family, HMGA2, is a potential biomarker of metastatic spread and therapeutic resistance. HMGA2 is expressed in embryonic tissues and is frequently upregulated in aggressively growing cancers, including CRC. As an architectural, non-histone chromatin binding factor, it initiates chromatin decompaction to facilitate transcriptional regulation. HMGA2 maintains the capacity for stem cell renewal in embryonic and cancer tissues and is a known promoter of epithelial-to-mesenchymal transition in tumor cells. This review will focus on the known molecular mechanisms by which HMGA2 exerts genome protective functions that contribute to cancer cell survival and chemoresistance in CRC.
Collapse
|
16
|
Girish V, Lakhani AA, Scaduto CM, Thompson SL, Brown LM, Hagenson RA, Sausville EL, Mendelson BE, Lukow DA, Yuan ML, Kandikuppa PK, Stevens EC, Lee SN, Salovska B, Li W, Smith JC, Taylor AM, Martienssen RA, Liu Y, Sun R, Sheltzer JM. Oncogene-like addiction to aneuploidy in human cancers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.09.523344. [PMID: 36711674 PMCID: PMC9882055 DOI: 10.1101/2023.01.09.523344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Most cancers exhibit aneuploidy, but its functional significance in tumor development is controversial. Here, we describe ReDACT (Restoring Disomy in Aneuploid cells using CRISPR Targeting), a set of chromosome engineering tools that allow us to eliminate specific aneuploidies from cancer genomes. Using ReDACT, we created a panel of isogenic cells that have or lack common aneuploidies, and we demonstrate that trisomy of chromosome 1q is required for malignant growth in cancers harboring this alteration. Mechanistically, gaining chromosome 1q increases the expression of MDM4 and suppresses TP53 signaling, and we show that TP53 mutations are mutually-exclusive with 1q aneuploidy in human cancers. Thus, specific aneuploidies play essential roles in tumorigenesis, raising the possibility that targeting these "aneuploidy addictions" could represent a novel approach for cancer treatment.
Collapse
Affiliation(s)
- Vishruth Girish
- Yale University School of Medicine, New Haven, CT 06511
- Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | | | | | | | | | | | | | | | - Monet Lou Yuan
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | | | | | - Sophia N. Lee
- Yale University School of Medicine, New Haven, CT 06511
| | | | - Wenxue Li
- Yale University School of Medicine, New Haven, CT 06511
| | - Joan C. Smith
- Yale University School of Medicine, New Haven, CT 06511
| | | | - Robert A. Martienssen
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- Howard Hughes Medical Institute, Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, USA
| | - Yansheng Liu
- Yale University School of Medicine, New Haven, CT 06511
| | - Ruping Sun
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55455
| | | |
Collapse
|
17
|
Baker NE, Montagna C. Reducing the aneuploid cell burden - cell competition and the ribosome connection. Dis Model Mech 2022; 15:dmm049673. [PMID: 36444717 PMCID: PMC10621665 DOI: 10.1242/dmm.049673] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
Aneuploidy, the gain or loss of chromosomes, is the cause of birth defects and miscarriage and is almost ubiquitous in cancer cells. Mosaic aneuploidy causes cancer predisposition, as well as age-related disorders. Despite the cell-intrinsic mechanisms that prevent aneuploidy, sporadic aneuploid cells do arise in otherwise normal tissues. These aneuploid cells can differ from normal cells in the copy number of specific dose-sensitive genes, and may also experience proteotoxic stress associated with mismatched expression levels of many proteins. These differences may mark aneuploid cells for recognition and elimination. The ribosomal protein gene dose in aneuploid cells could be important because, in Drosophila, haploinsufficiency for these genes leads to elimination by the process of cell competition. Constitutive haploinsufficiency for human ribosomal protein genes causes Diamond Blackfan anemia, but it is not yet known whether ribosomal protein gene dose contributes to aneuploid cell elimination in mammals. In this Review, we discuss whether cell competition on the basis of ribosomal protein gene dose is a tumor suppressor mechanism, reducing the accumulation of aneuploid cells. We also discuss how this might relate to the tumor suppressor function of p53 and the p53-mediated elimination of aneuploid cells from murine embryos, and how cell competition defects could contribute to the cancer predisposition of Diamond Blackfan anemia.
Collapse
Affiliation(s)
- Nicholas E. Baker
- Departments of Genetics, Developmental and Molecular Biology, and Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Cristina Montagna
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey, 195 Little Albany Street, New Brunswick, NJ 08901, USA
| |
Collapse
|
18
|
Wang J, Thomas HR, Chen Y, Percival SM, Waldrep SC, Ramaker RC, Thompson RG, Cooper SJ, Chong Z, Parant JM. Reduced sister chromatid cohesion acts as a tumor penetrance modifier. PLoS Genet 2022; 18:e1010341. [PMID: 35994499 PMCID: PMC9436123 DOI: 10.1371/journal.pgen.1010341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 09/01/2022] [Accepted: 07/14/2022] [Indexed: 11/23/2022] Open
Abstract
Sister chromatid cohesion (SCC) is an important process in chromosome segregation. ESCO2 is essential for establishment of SCC and is often deleted/altered in human cancers. We demonstrate that esco2 haploinsufficiency results in reduced SCC and accelerates the timing of tumor onset in both zebrafish and mouse p53 heterozygous null models, but not in p53 homozygous mutant or wild-type animals. These data indicate that esco2 haploinsufficiency accelerates tumor onset in a loss of heterozygosity (LOH) sensitive background. Analysis of The Cancer Genome Atlas (TCGA) confirmed ESCO2 deficient tumors have elevated number of LOH events throughout the genome. Further, we demonstrated heterozygous loss of sgo1, important in maintaining SCC, also results in reduced SCC and accelerated tumor formation in a p53 heterozygous background. Surprisingly, while we did observe elevated levels of chromosome missegregation and micronuclei formation in esco2 heterozygous mutant animals, this chromosomal instability did not contribute to the accelerated tumor onset in a p53 heterozygous background. Interestingly, SCC also plays a role in homologous recombination, and we did observe elevated levels of mitotic recombination derived p53 LOH in tumors from esco2 haploinsufficient animals; as well as elevated levels of mitotic recombination throughout the genome of human ESCO2 deficient tumors. Together these data suggest that reduced SCC contributes to accelerated tumor penetrance through elevated mitotic recombination. Tumorigenesis often involves the inactivation of tumor suppressor genes. This often encompasses an inactivation mutation in one allele and loss of the other wild-type allele, referred to as loss of heterozygosity (LOH). The rate at which the cells lose the wild-type allele can influence the timing of tumor onset, and therefore an indicator of a patient’s risk of cancer. Factors that influence this process could be used as a predictive indicator of cancer risk, however these factors are still unclear. We demonstrate that partial impairment of sister chromatid cohesion (SCC), a fundamental component of the chromosome segregation in mitosis and homologous recombination repair, enhanced tumorigenesis. Our data suggest this is through elevated levels of mitotic recombination derived p53 LOH. This study emphasizes the importance of understanding how impaired SCC, mitotic recombination rates, and LOH rates influence cancer risk.
Collapse
Affiliation(s)
- Jun Wang
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Holly R. Thomas
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Yu Chen
- Department of Genetics, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
- Informatics Institute, University of Alabama at Birmingham Heersink School of Medicine, Alabama, United States of America
| | - Stefanie M. Percival
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Stephanie C. Waldrep
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Ryne C. Ramaker
- Hudson Alpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Robert G. Thompson
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
| | - Sara J. Cooper
- Hudson Alpha Institute for Biotechnology, Huntsville, Alabama, United States of America
| | - Zechen Chong
- Department of Genetics, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
- Informatics Institute, University of Alabama at Birmingham Heersink School of Medicine, Alabama, United States of America
| | - John M. Parant
- Department of Pharmacology and Toxicology, University of Alabama at Birmingham Heersink School of Medicine, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
19
|
Lauriola A, Davalli P, Marverti G, Caporali A, Mai S, D’Arca D. Telomere Dysfunction Is Associated with Altered DNA Organization in Trichoplein/Tchp/Mitostatin (TpMs) Depleted Cells. Biomedicines 2022; 10:biomedicines10071602. [PMID: 35884905 PMCID: PMC9312488 DOI: 10.3390/biomedicines10071602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 12/02/2022] Open
Abstract
Recently, we highlighted a novel role for the protein Trichoplein/TCHP/Mitostatin (TpMs), both as mitotic checkpoint regulator and guardian of chromosomal stability. TpMs-depleted cells show numerical and structural chromosome alterations that lead to genomic instability. This condition is a major driving force in malignant transformation as it allows for the cells acquiring new functional capabilities to proliferate and disseminate. Here, the effect of TpMs depletion was investigated in different TpMs-depleted cell lines by means of 3D imaging and 3D Structured illumination Microscopy. We show that TpMs depletion causes alterations in the 3D architecture of telomeres in colon cancer HCT116 cells. These findings are consistent with chromosome alterations that lead to genomic instability. Furthermore, TpMs depletion changes the spatial arrangement of chromosomes and other nuclear components. Modified nuclear architecture and organization potentially induce variations that precede the onset of genomic instability and are considered as markers of malignant transformation. Our present observations connect the tumor suppression ability of TpMs with its novel functions in maintaining the proper chromosomal segregation as well as the proper telomere and nuclear architecture. Further investigations will investigate the connection between alterations in telomeres and nuclear architecture with the progression of human tumors with the aim of developing personalized therapeutic interventions.
Collapse
Affiliation(s)
- Angela Lauriola
- Department of Biotechnology, University of Verona, 37134 Verona, Italy;
| | - Pierpaola Davalli
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy; (P.D.); (G.M.)
| | - Gaetano Marverti
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy; (P.D.); (G.M.)
| | - Andrea Caporali
- The Queen’s Medical Research Institute, BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh EH10 4AH, UK;
| | - Sabine Mai
- CancerCare Manitoba Research Institute, University of Manitoba, CancerCare Manitoba, Winnipeg, MB R3E 0V9, Canada
- Correspondence: (S.M.); (D.D.); Tel.: +1-204-272-3174 (S.M.); +39-059-205-5610 (D.D.)
| | - Domenico D’Arca
- Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Via G. Campi 287, 41125 Modena, Italy; (P.D.); (G.M.)
- Correspondence: (S.M.); (D.D.); Tel.: +1-204-272-3174 (S.M.); +39-059-205-5610 (D.D.)
| |
Collapse
|
20
|
Li M, Duan X, Xiao Y, Yuan M, Zhao Z, Cui X, Wu D, Shi J. BUB1 Is Identified as a Potential Therapeutic Target for Pancreatic Cancer Treatment. Front Public Health 2022; 10:900853. [PMID: 35769782 PMCID: PMC9235519 DOI: 10.3389/fpubh.2022.900853] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 05/03/2022] [Indexed: 11/17/2022] Open
Abstract
Pancreatic cancer is one of the most challenging cancer types in clinical treatment worldwide. This study aimed to understand the tumorigenesis mechanism and explore potential therapeutic targets for patients with pancreatic cancer. Single-cell data and expression profiles of pancreatic cancer samples and normal tissues from multiple databases were included. Comprehensive bioinformatics analyses were applied to clarify tumor microenvironment and identify key genes involved in cancer development. Immense difference of cell types was shown between tumor and normal samples. Four cell types (B cell_1, B cell_2, cancer cell_3, and CD1C+_B dendritic cell_3) were screened to be significantly associated with prognosis. Three ligand-receptor pairs, including CD74-MIF, CD74-COPA, and CD74-APP, greatly contributed to tumorigenesis. High expression of BUB1 (BUB1 Mitotic Checkpoint Serine/Threonine Kinase) was closely correlated with worse prognosis. CD1C+_B dendritic cell_3 played a key role in tumorigenesis and cancer progression possibly through CD74-MIF. BUB1 can serve as a prognostic biomarker and a therapeutic target for patients with pancreatic cancer. The study provided a novel insight into studying the molecular mechanism of pancreatic cancer development and proposed a potential strategy for exploiting new drugs.
Collapse
Affiliation(s)
- Ming Li
- Department of General Surgery, Shijiazhuang People's Hospital, Shijiazhuang, China
| | - Xiaoyang Duan
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Hebei Tumor Hospital, Shijiazhuang, China
| | - Yajie Xiao
- Translational Medicine, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Meng Yuan
- Internal Medical, University of Occupational and Environmental Health, Fukuoka, Japan
| | - Zhikun Zhao
- Translational Medicine, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Xiaoli Cui
- Translational Medicine, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Dongfang Wu
- Translational Medicine, YuceBio Technology Co., Ltd., Shenzhen, China
| | - Jian Shi
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, Hebei Tumor Hospital, Shijiazhuang, China
| |
Collapse
|
21
|
Guo S, Li T, Xu D, Xu J, Wang H, Li J, Bi X, Cao M, Xu Z, Xia Q, Cui Y, Li K. Prognostic Implications and Immune Infiltration Characteristics of Chromosomal Instability-Related Dysregulated CeRNA in Lung Adenocarcinoma. Front Mol Biosci 2022; 9:843640. [PMID: 35419410 PMCID: PMC8995899 DOI: 10.3389/fmolb.2022.843640] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Accepted: 02/22/2022] [Indexed: 12/14/2022] Open
Abstract
An accumulating body of research indicates that long-noncoding RNAs (lncRNAs) regulate the target genes and act as competitive endogenous RNAs (ceRNAs) playing an indispensable role in lung adenocarcinoma (LUAD). LUAD is frequently accompanied by the feature of chromosomal instability (CIN); however, CIN-related ceRNAs have not been investigated yet. We systematically analyzed and integrated CIN-related dysregulated ceRNAs characteristics in LUAD samples for the first time. In TCGA LUAD cohort, CIN in tumor samples was significantly higher than that in those of adjacent, and patients with high CIN risk tended to have worse clinical outcomes. We constructed a double-weighted CIN-related dysregulated ceRNA network, in which edge weight and node weight represented the disorder extent of ceRNA and the correlation of RNA expression level and prognosis, respectively. After module mining and analysis, a potential prognostic biomarker composed of 12 RNAs (8 mRNAs and 4 lncRNAs) named CIN-related dysregulated ceRNAs (CRDC) was obtained. The CRDC risk score had a positive relation with clinical stage and CIN, and patients with high CRDC risk scores exhibited poor prognosis. Moreover, CRDC tended to be an independent risk factor with high robustness to overcome the effect of multicollinearity among other explanatory variables for disease-specific survival (DSS) in TCGA and two GEO cohorts. The result of functional analysis indicated that CRDC was involved in multiple cancer progresses, especially immune-related pathways. The patients with lower CRDC risk had higher B cell, T cell CD4+, T cell CD8+, neutrophil, macrophage, and myeloid dendritic cell infiltration than the patients with higher CRDC risk. Meanwhile, patients with lower CRDC risk could get more benefits from immunological therapy. The results suggested that the CRDC could be a potential prognostic biomarker and an immunotherapy predictor for lung adenocarcinoma.
Collapse
Affiliation(s)
- Shengnan Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Institute of Nephrology Second Affiliated Hospital and Hainan General Hospital, Hainan Medical University, Haikou, China
| | - Tianhao Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Institute of Nephrology Second Affiliated Hospital and Hainan General Hospital, Hainan Medical University, Haikou, China
| | - Dahua Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Institute of Nephrology Second Affiliated Hospital and Hainan General Hospital, Hainan Medical University, Haikou, China
| | - Jiankai Xu
- College of Bioinformatics Science and Technology, Cancer Hospital, Harbin Medical University, Harbin, China
| | - Hong Wang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Institute of Nephrology Second Affiliated Hospital and Hainan General Hospital, Hainan Medical University, Haikou, China
| | - Jian Li
- College of Bioinformatics Science and Technology, Cancer Hospital, Harbin Medical University, Harbin, China
| | - Xiaoman Bi
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Institute of Nephrology Second Affiliated Hospital and Hainan General Hospital, Hainan Medical University, Haikou, China
| | - Meng Cao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Institute of Nephrology Second Affiliated Hospital and Hainan General Hospital, Hainan Medical University, Haikou, China
| | - Zhizhou Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Institute of Nephrology Second Affiliated Hospital and Hainan General Hospital, Hainan Medical University, Haikou, China
| | - Qianfeng Xia
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, NHC Key Laboratory of Control of Tropical Diseases, School of Tropical Medicine, The Second Affiliated Hospital, Hainan Medical University, Haikou, China
- *Correspondence: Qianfeng Xia, ; Ying Cui, ; Kongning Li,
| | - Ying Cui
- College of Bioinformatics Science and Technology, Cancer Hospital, Harbin Medical University, Harbin, China
- *Correspondence: Qianfeng Xia, ; Ying Cui, ; Kongning Li,
| | - Kongning Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Institute of Nephrology Second Affiliated Hospital and Hainan General Hospital, Hainan Medical University, Haikou, China
- *Correspondence: Qianfeng Xia, ; Ying Cui, ; Kongning Li,
| |
Collapse
|
22
|
Yan X, Liu SM, Liu C. Clinical Applications of Aneuploidies in Evolution of NSCLC Patients: Current Status and Application Prospect. Onco Targets Ther 2022; 15:1355-1368. [PMID: 36388157 PMCID: PMC9662021 DOI: 10.2147/ott.s380016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 10/22/2022] [Indexed: 11/11/2022] Open
Abstract
As one of the first characteristics of cancer cells, chromosomal aberrations during cell division have been well documented. Aneuploidy is a feature of most cancer cells accompanied by an elevated rate of mis-segregation of chromosomes, called chromosome instability (CIN). Aneuploidy causes ongoing karyotypic changes that contribute to tumor heterogeneity, drug resistance, and treatment failure, which are considered predictors of poor prognosis. Lung cancer (LC) is the leading cause of cancer-related deaths worldwide, and its genome map shows extensive aneuploid changes. Elucidating the role of aneuploidy in the pathogenesis of LC will reveal information about the key factors of tumor occurrence and development, help to predict the prognosis of cancer, clarify tumor evolution, metastasis, and drug response, and may promote the development of precision oncology. In this review, we describe many possible causes of aneuploidy and provide evidence of the role of aneuploidy in the evolution of LC, providing a basis for future biological and clinical research.
Collapse
Affiliation(s)
- Xing Yan
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, People's Republic of China
| | - Shan Mei Liu
- Inner Mongolia Medical University, Hohhot, 150110, People's Republic of China
| | - Changhong Liu
- The Second Affiliated Hospital of Dalian Medical University, Dalian, 116000, People's Republic of China
| |
Collapse
|
23
|
Hill W, Caswell DR, Swanton C. Capturing cancer evolution using genetically engineered mouse models (GEMMs). Trends Cell Biol 2021; 31:1007-1018. [PMID: 34400045 DOI: 10.1016/j.tcb.2021.07.003] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Revised: 07/11/2021] [Accepted: 07/15/2021] [Indexed: 12/17/2022]
Abstract
Initiating from a single cell, cancer undergoes clonal evolution, leading to a high degree of intratumor heterogeneity (ITH). The arising genetic heterogeneity between cancer cells is influenced by exogenous and endogenous forces that shape the composition of clones within tumors. Preclinical mouse models have provided a valuable tool for understanding cancer, helping to build a fundamental understanding of tumor initiation, progression, and metastasis. Until recently, genetically engineered mouse models (GEMMS) of cancer had lacked the genetic diversity found in human tumors, in which evolution may be driven by long-term carcinogen exposure and DNA damage. However, advances in sequencing technology and in our understanding of the drivers of genetic instability have given us the knowledge to generate new mouse models, offering an approach to functionally explore mechanisms of tumor evolution.
Collapse
Affiliation(s)
- William Hill
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Deborah R Caswell
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK
| | - Charles Swanton
- Cancer Evolution and Genome Instability Laboratory, The Francis Crick Institute, London, UK; Cancer Research UK Lung Cancer Centre of Excellence, University College London Cancer Institute, University College London, London, UK; University College London Hospitals NHS Trust, London, UK.
| |
Collapse
|
24
|
Herlihy CP, Hahn S, Hermance NM, Crowley EA, Manning AL. Suv420 enrichment at the centromere limits Aurora B localization and function. J Cell Sci 2021; 134:jcs249763. [PMID: 34342353 PMCID: PMC8353524 DOI: 10.1242/jcs.249763] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 07/05/2021] [Indexed: 12/16/2022] Open
Abstract
Centromere structure and function are defined by the epigenetic modification of histones at centromeric and pericentromeric chromatin. The constitutive heterochromatin found at pericentromeric regions is highly enriched for H3K9me3 and H4K20me3. Although mis-expression of the methyltransferase enzymes that regulate these marks, Suv39 and Suv420, is common in disease, the consequences of such changes are not well understood. Our data show that increased centromere localization of Suv39 and Suv420 suppresses centromere transcription and compromises localization of the mitotic kinase Aurora B, decreasing microtubule dynamics and compromising chromosome alignment and segregation. We find that inhibition of Suv420 methyltransferase activity partially restores Aurora B localization to centromeres and that restoration of the Aurora B-containing chromosomal passenger complex to the centromere is sufficient to suppress mitotic errors that result when Suv420 and H4K20me3 is enriched at centromeres. Consistent with a role for Suv39 and Suv420 in negatively regulating Aurora B, high expression of these enzymes corresponds with increased sensitivity to Aurora kinase inhibition in human cancer cells, suggesting that increased H3K9 and H4K20 methylation may be an underappreciated source of chromosome mis-segregation in cancer. This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
| | | | | | | | - Amity L. Manning
- Department of Biology and Biotechnology, Worcester Polytechnic Institute, Worcester, MA, 01609USA
| |
Collapse
|
25
|
Shoshani O, Bakker B, de Haan L, Tijhuis AE, Wang Y, Kim DH, Maldonado M, Demarest MA, Artates J, Zhengyu O, Mark A, Wardenaar R, Sasik R, Spierings DCJ, Vitre B, Fisch K, Foijer F, Cleveland DW. Transient genomic instability drives tumorigenesis through accelerated clonal evolution. Genes Dev 2021; 35:1093-1108. [PMID: 34266887 PMCID: PMC8336898 DOI: 10.1101/gad.348319.121] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/10/2021] [Indexed: 12/13/2022]
Abstract
In this study, Shoshani et al. tested the role of aneuploidy in tumor initiation and progression, and generated mice with random aneuploidies by transient induction of polo-like kinase 4 (Plk4), a master regulator of centrosome number. Their findings show how transient CIN generates cells with random aneuploidies from which ones that acquire a karyotype with specific chromosome gains are sufficient to drive cancer formation, and that distinct CIN mechanisms can lead to similar karyotypic cancer-causing outcomes. Abnormal numerical and structural chromosome content is frequently found in human cancer. To test the role of aneuploidy in tumor initiation and progression, we generated mice with random aneuploidies by transient induction of polo-like kinase 4 (Plk4), a master regulator of centrosome number. Short-term chromosome instability (CIN) from transient Plk4 induction resulted in formation of aggressive T-cell lymphomas in mice with heterozygous inactivation of one p53 allele and accelerated tumor development in the absence of p53. Transient CIN increased the frequency of lymphoma-initiating cells with a specific karyotype profile, including trisomy of chromosomes 4, 5, 14, and 15 occurring early in tumorigenesis. Tumor development in mice with chronic CIN induced by an independent mechanism (through inactivation of the spindle assembly checkpoint) gradually trended toward a similar karyotypic profile, as determined by single-cell whole-genome DNA sequencing. Overall, we show how transient CIN generates cells with random aneuploidies from which ones that acquire a karyotype with specific chromosome gains are sufficient to drive cancer formation, and that distinct CIN mechanisms can lead to similar karyotypic cancer-causing outcomes.
Collapse
Affiliation(s)
- Ofer Shoshani
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Bjorn Bakker
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Lauren de Haan
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA.,European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Andréa E Tijhuis
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Yin Wang
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Dong Hyun Kim
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Marcus Maldonado
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Matthew A Demarest
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Jon Artates
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Ouyang Zhengyu
- Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Adam Mark
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - René Wardenaar
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Roman Sasik
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Diana C J Spierings
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Benjamin Vitre
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Kathleen Fisch
- Center for Computational Biology and Bioinformatics, Department of Medicine, University of California at San Diego, La Jolla, California 92093, USA
| | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, 9713 AV Groningen, The Netherlands
| | - Don W Cleveland
- Ludwig Cancer Research, Department of Cellular and Molecular Medicine, University of California at San Diego, La Jolla, California 92093, USA
| |
Collapse
|
26
|
Trakala M, Aggarwal M, Sniffen C, Zasadil L, Carroll A, Ma D, Su XA, Wangsa D, Meyer A, Sieben CJ, Zhong J, Hsu PH, Paradis G, Ried T, Holland A, Van Deursen J, Amon A. Clonal selection of stable aneuploidies in progenitor cells drives high-prevalence tumorigenesis. Genes Dev 2021; 35:1079-1092. [PMID: 34266888 PMCID: PMC8336892 DOI: 10.1101/gad.348341.121] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 06/04/2021] [Indexed: 12/16/2022]
Abstract
In this study Trakala et al. investigated how chromosome gains and losses, which are a frequent feature of human cancers, can overcome the detrimental effects of aneuploidy. They developed a novel mouse model that enables unprecedented levels of chromosome missegregation in the adult animal and their results show that the initial detrimental effects of random missegregation are outweighed by clonal selection, which is dependent on chromosomal location and the nature of specific genes and is sufficient to drive cancer. Chromosome gains and losses are a frequent feature of human cancers. However, how these aberrations can outweigh the detrimental effects of aneuploidy remains unclear. An initial comparison of existing chromosomal instability (CIN) mouse models suggests that aneuploidy accumulates to low levels in these animals. We therefore developed a novel mouse model that enables unprecedented levels of chromosome missegregation in the adult animal. At the earliest stages of T-cell development, cells with random chromosome gains and/or losses are selected against, but CIN eventually results in the expansion of progenitors with clonal chromosomal imbalances. Clonal selection leads to the development of T-cell lymphomas with stereotypic karyotypes in which chromosome 15, containing the Myc oncogene, is gained with high prevalence. Expressing human MYC from chromosome 6 (MYCChr6) is sufficient to change the karyotype of these lymphomas to include universal chromosome 6 gains. Interestingly, while chromosome 15 is still gained in MYCChr6 tumors after genetic ablation of the endogenous Myc locus, this chromosome is not efficiently gained after deletion of one copy of Rad21, suggesting a synergistic effect of both MYC and RAD21 in driving chromosome 15 gains. Our results show that the initial detrimental effects of random missegregation are outbalanced by clonal selection, which is dictated by the chromosomal location and nature of certain genes and is sufficient to drive cancer with high prevalence.
Collapse
Affiliation(s)
- Marianna Trakala
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Muskaan Aggarwal
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Courtney Sniffen
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Lauren Zasadil
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Allison Carroll
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Duanduan Ma
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Xiaofeng A Su
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Darawalee Wangsa
- Genetics Branch National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Ashleigh Meyer
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Cynthia J Sieben
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Jian Zhong
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Pei-Hsin Hsu
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Glenn Paradis
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| | - Thomas Ried
- Genetics Branch National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Andrew Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Jan Van Deursen
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Angelika Amon
- David H. Koch Institute for Integrative Cancer Research, Howard Hughes Medical Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts 02142, USA
| |
Collapse
|
27
|
Cao P, Li G, Xiang J. Genome instability and lymphoma. ZHONG NAN DA XUE XUE BAO. YI XUE BAN = JOURNAL OF CENTRAL SOUTH UNIVERSITY. MEDICAL SCIENCES 2021; 46:552-557. [PMID: 34148893 PMCID: PMC10930211 DOI: 10.11817/j.issn.1672-7347.2021.190427] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Indexed: 11/03/2022]
Abstract
Lymphoma is one of the most common malignant tumor of the hematologic system. The genome instability is not only an important molecular basis for the development of lymphoma, but also has important value in the diagnosis and prognosis of lymphoma. There are 2 types of genome instability: Microsatellite instability (MSI/MIN) at gene level and chromosomal instability at chromosome level. Through the study on genes associated with lymphoma, the unstable genes associated with lymphoma could be found, meanwhile the mechanism of its occurrence and development of lymphoma could be explored, and the important basis of molecular biology could also be provided in the field of current hot lymphoma precision medical research.
Collapse
Affiliation(s)
- Pengfei Cao
- Department of Hematology, Xiangya Hospital, Central South University, Changsha 410008.
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China.
| | - Guiyuan Li
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China.
| | - Juanjuan Xiang
- Cancer Research Institute, School of Basic Medical Science, Central South University, Changsha 410078, China
| |
Collapse
|
28
|
Zhang Y, Mandemaker IK, Matsumoto S, Foreman O, Holland CP, Lloyd WR, Sugasawa K, Vermeulen W, Marteijn JA, Galardy PJ. USP44 Stabilizes DDB2 to Facilitate Nucleotide Excision Repair and Prevent Tumors. Front Cell Dev Biol 2021; 9:663411. [PMID: 33937266 PMCID: PMC8085418 DOI: 10.3389/fcell.2021.663411] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 03/29/2021] [Indexed: 02/02/2023] Open
Abstract
Nucleotide excision repair (NER) is a pathway involved in the repair of a variety of potentially mutagenic lesions that distort the DNA double helix. The ubiquitin E3-ligase complex UV-DDB is required for the recognition and repair of UV-induced cyclobutane pyrimidine dimers (CPDs) lesions through NER. DDB2 directly binds CPDs and subsequently undergoes ubiquitination and proteasomal degradation. DDB2 must remain on damaged chromatin, however, for sufficient time to recruit and hand-off lesions to XPC, a factor essential in the assembly of downstream repair components. Here we show that the tumor suppressor USP44 directly deubiquitinates DDB2 to prevent its premature degradation and is selectively required for CPD repair. Cells lacking USP44 have impaired DDB2 accumulation on DNA lesions with subsequent defects in XPC retention. The physiological importance of this mechanism is evident in that mice lacking Usp44 are prone to tumors induced by NER lesions introduced by DMBA or UV light. These data reveal the requirement for highly regulated ubiquitin addition and removal in the recognition and repair of helix-distorting DNA damage and identify another mechanism by which USP44 protects genomic integrity and prevents tumors.
Collapse
Affiliation(s)
- Ying Zhang
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States
| | - Imke K Mandemaker
- Department of Molecular Genetics, Oncode Institute, Erasmus MC, Rotterdam, Netherlands
| | | | - Oded Foreman
- Department of Pathology, Genentech, South San Francisco, CA, United States
| | - Christopher P Holland
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States
| | - Whitney R Lloyd
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States
| | - Kaoru Sugasawa
- Biosignal Research Center, Kobe University, Hyogo, Japan
| | - Wim Vermeulen
- Department of Molecular Genetics, Oncode Institute, Erasmus MC, Rotterdam, Netherlands
| | - Jurgen A Marteijn
- Department of Molecular Genetics, Oncode Institute, Erasmus MC, Rotterdam, Netherlands
| | - Paul J Galardy
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, MN, United States.,Division of Pediatric Hematology-Oncology, Mayo Clinic, Rochester, MN, United States.,Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
29
|
Ji Z, Chuen J, Kiparaki M, Baker N. Cell competition removes segmental aneuploid cells from Drosophila imaginal disc-derived tissues based on ribosomal protein gene dose. eLife 2021; 10:61172. [PMID: 33847264 PMCID: PMC8043752 DOI: 10.7554/elife.61172] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 03/08/2021] [Indexed: 12/22/2022] Open
Abstract
Aneuploidy causes birth defects and miscarriages, occurs in nearly all cancers and is a hallmark of aging. Individual aneuploid cells can be eliminated from developing tissues by unknown mechanisms. Cells with ribosomal protein (Rp) gene mutations are also eliminated, by cell competition with normal cells. Because Rp genes are spread across the genome, their copy number is a potential marker for aneuploidy. We found that elimination of imaginal disc cells with irradiation-induced genome damage often required cell competition genes. Segmentally aneuploid cells derived from targeted chromosome excisions were eliminated by the RpS12-Xrp1 cell competition pathway if they differed from neighboring cells in Rp gene dose, whereas cells with normal doses of the Rp and eIF2γ genes survived and differentiated adult tissues. Thus, cell competition, triggered by differences in Rp gene dose between cells, is a significant mechanism for the elimination of aneuploid somatic cells, likely to contribute to preventing cancer. Aneuploid cells emerge when cellular division goes awry and a cell ends up with the wrong number of chromosomes, the tiny genetic structures carrying the instructions that control life’s processes. Aneuploidy can lead to fatal conditions during development, and to cancer in an adult organism. A safety mechanism may exist that helps the body to detect and remove these cells. Yet, exactly this happens is still poorly understood: in particular, it is unclear how cells manage to ‘count’ their chromosomes. One way they could do so is through the ribosomes, the molecular ‘factories’ that create the building blocks required for life. In a cell, every chromosome carries genes that code for the proteins (known as Rps) forming ribosomes. Aneuploidy will alter the number of Rp genes, and in turn the amount and type of Rps the cell produces, so that ribosomes and the genes for Rps could act as a ‘readout’ of aneuploidy. Ji et al set out to test this theory in fruit flies. The first experiment used a genetic manipulation technique called site-specific recombination to remove parts of chromosomes from cells in the developing eye and wing. Cells which retained all their Rp genes survived, while those that were missing some usually died – but only when the surrounding cells were normal. In this situation, healthy cells eliminated their damaged neighbours through a process known as cell competition. A second experiment, using radiation as an alternative method of damaging chromosomes, also gave similar results. The work by Ji et al. reveals how the body can detect and eliminate aneuploid cells, potentially before they can cause harm. If the same mechanism applies in humans, boosting cell competition may, one day, helps to combat diseases like cancer.
Collapse
Affiliation(s)
- Zhejun Ji
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Jacky Chuen
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Marianthi Kiparaki
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| | - Nicholas Baker
- Department of Genetics, Albert Einstein College of Medicine, Bronx, United States
| |
Collapse
|
30
|
Baudoin NC, Bloomfield M. Karyotype Aberrations in Action: The Evolution of Cancer Genomes and the Tumor Microenvironment. Genes (Basel) 2021; 12:558. [PMID: 33921421 PMCID: PMC8068843 DOI: 10.3390/genes12040558] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 03/27/2021] [Accepted: 04/06/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer is a disease of cellular evolution. For this cellular evolution to take place, a population of cells must contain functional heterogeneity and an assessment of this heterogeneity in the form of natural selection. Cancer cells from advanced malignancies are genomically and functionally very different compared to the healthy cells from which they evolved. Genomic alterations include aneuploidy (numerical and structural changes in chromosome content) and polyploidy (e.g., whole genome doubling), which can have considerable effects on cell physiology and phenotype. Likewise, conditions in the tumor microenvironment are spatially heterogeneous and vastly different than in healthy tissues, resulting in a number of environmental niches that play important roles in driving the evolution of tumor cells. While a number of studies have documented abnormal conditions of the tumor microenvironment and the cellular consequences of aneuploidy and polyploidy, a thorough overview of the interplay between karyotypically abnormal cells and the tissue and tumor microenvironments is not available. Here, we examine the evidence for how this interaction may unfold during tumor evolution. We describe a bidirectional interplay in which aneuploid and polyploid cells alter and shape the microenvironment in which they and their progeny reside; in turn, this microenvironment modulates the rate of genesis for new karyotype aberrations and selects for cells that are most fit under a given condition. We conclude by discussing the importance of this interaction for tumor evolution and the possibility of leveraging our understanding of this interplay for cancer therapy.
Collapse
Affiliation(s)
- Nicolaas C. Baudoin
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | - Mathew Bloomfield
- Department of Biological Sciences and Fralin Life Sciences Institute, Virginia Tech, Blacksburg, VA 24061, USA
| |
Collapse
|
31
|
Gheghiani L, Wang L, Zhang Y, Moore XTR, Zhang J, Smith SC, Tian Y, Wang L, Turner K, Jackson-Cook CK, Mukhopadhyay ND, Fu Z. PLK1 Induces Chromosomal Instability and Overrides Cell-Cycle Checkpoints to Drive Tumorigenesis. Cancer Res 2021; 81:1293-1307. [PMID: 33376114 PMCID: PMC8026515 DOI: 10.1158/0008-5472.can-20-1377] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 10/19/2020] [Accepted: 12/21/2020] [Indexed: 11/16/2022]
Abstract
Polo-like kinase 1 (PLK1) is an essential cell-cycle regulator that is frequently overexpressed in various human cancers. To determine whether Plk1 overexpression drives tumorigenesis, we established transgenic mouse lines that ubiquitously express increased levels of Plk1. High Plk1 levels were a driving force for different types of spontaneous tumors. Increased Plk1 levels resulted in multiple defects in mitosis and cytokinesis, supernumerary centrosomes, and compromised cell-cycle checkpoints, allowing accumulation of chromosomal instability (CIN), which resulted in aneuploidy and tumor formation. Clinically, higher expression of PLK1 positively associated with an increase in genome-wide copy-number alterations in multiple human cancers. This study provides in vivo evidence that aberrant expression of PLK1 triggers CIN and tumorigenesis and highlights potential therapeutic opportunities for CIN-positive cancers. SIGNIFICANCE: These findings establish roles for PLK1 as a potent proto-oncogene and a CIN gene and provide insights for the development of effective treatment regimens across PLK1-overexpressing and CIN-positive cancers.
Collapse
Affiliation(s)
- Lilia Gheghiani
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Lei Wang
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Youwei Zhang
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Xavier T R Moore
- Department of Biology, Virginia Commonwealth University, Richmond, Virginia
| | - Jinglei Zhang
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Steven C Smith
- Department of Pathology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Yijun Tian
- Department of Tumor Biology, Moffitt Cancer Center, University of South Florida, Tampa, Florida
| | - Liang Wang
- Department of Tumor Biology, Moffitt Cancer Center, University of South Florida, Tampa, Florida
| | - Kristi Turner
- Department of Pathology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Colleen K Jackson-Cook
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
- Department of Pathology, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Nitai D Mukhopadhyay
- Department of Biostatistics, Virginia Commonwealth University, School of Medicine, Richmond, Virginia
| | - Zheng Fu
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, VCU Massey Cancer Center, Virginia Commonwealth University, School of Medicine, Richmond, Virginia.
| |
Collapse
|
32
|
Bedekovics T, Hussain S, Zhang Y, Ali A, Jeon YJ, Galardy PJ. USP24 Is a Cancer-Associated Ubiquitin Hydrolase, Novel Tumor Suppressor, and Chromosome Instability Gene Deleted in Neuroblastoma. Cancer Res 2021; 81:1321-1331. [PMID: 33355202 DOI: 10.1158/0008-5472.can-20-1777] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 11/16/2020] [Accepted: 12/18/2020] [Indexed: 11/16/2022]
Abstract
Deubiquitinating enzymes are increasingly recognized to play important roles in cancer, with many acting as oncogenes or tumor suppressors. In this study, we employed a bioinformatics approach to screen for enzymes from this family involved in cancer and found USP24 as a potent predictor of poor outcomes in neuroblastoma, an aggressive childhood cancer. USP24 resides in a region commonly deleted in neuroblastoma, yet was independently associated with poor outcomes in this disease. Deletion of Usp24 in a murine model resulted in degradation of collapsin response mediator protein 2 (CRMP2), a regulator of axon growth, guidance, and neuronal polarity. Cells lacking USP24 had significant increases in spindle defects, chromosome missegregation, and aneuploidy, phenotypes that were rescued by the restoration of CRMP2. USP24 prevented aneuploidy by maintaining spindle-associated CRMP2, which is required for mitotic accuracy. Our findings further indicate that USP24 is a tumor suppressor that may play an important role in the pathogenesis of neuroblastoma. SIGNIFICANCE: This study identifies the chromosome instability gene USP24 as frequently deleted in neuroblastoma and provides important insight into the pathogenesis of this aggressive childhood cancer.
Collapse
Affiliation(s)
- Tibor Bedekovics
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Sajjad Hussain
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Ying Zhang
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Asma Ali
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
| | - Young J Jeon
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota
- Department of Pharmacology, Chosun University College of Medicine, Gwangju, South Korea
| | - Paul J Galardy
- Department of Pediatric and Adolescent Medicine, Mayo Clinic, Rochester, Minnesota.
- Division of Pediatric Hematology-Oncology, Mayo Clinic, Rochester, Minnesota
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
33
|
Jilderda LJ, Zhou L, Foijer F. Understanding How Genetic Mutations Collaborate with Genomic Instability in Cancer. Cells 2021; 10:342. [PMID: 33562057 PMCID: PMC7914657 DOI: 10.3390/cells10020342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 01/25/2021] [Accepted: 02/03/2021] [Indexed: 01/23/2023] Open
Abstract
Chromosomal instability is the process of mis-segregation for ongoing chromosomes, which leads to cells with an abnormal number of chromosomes, also known as an aneuploid state. Induced aneuploidy is detrimental during development and in primary cells but aneuploidy is also a hallmark of cancer cells. It is therefore believed that premalignant cells need to overcome aneuploidy-imposed stresses to become tumorigenic. Over the past decade, some aneuploidy-tolerating pathways have been identified through small-scale screens, which suggest that aneuploidy tolerance pathways can potentially be therapeutically exploited. However, to better understand the processes that lead to aneuploidy tolerance in cancer cells, large-scale and unbiased genetic screens are needed, both in euploid and aneuploid cancer models. In this review, we describe some of the currently known aneuploidy-tolerating hits, how large-scale genome-wide screens can broaden our knowledge on aneuploidy specific cancer driver genes, and how we can exploit the outcomes of these screens to improve future cancer therapy.
Collapse
Affiliation(s)
| | | | - Floris Foijer
- European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Centre Groningen, 9713 AV Groningen, The Netherlands; (L.J.J.); (L.Z.)
| |
Collapse
|
34
|
Vasudevan A, Schukken KM, Sausville EL, Girish V, Adebambo OA, Sheltzer JM. Aneuploidy as a promoter and suppressor of malignant growth. Nat Rev Cancer 2021; 21:89-103. [PMID: 33432169 DOI: 10.1038/s41568-020-00321-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/12/2020] [Indexed: 12/13/2022]
Abstract
Aneuploidy has been recognized as a hallmark of tumorigenesis for more than 100 years, but the connection between chromosomal errors and malignant growth has remained obscure. New evidence emerging from both basic and clinical research has illuminated a complicated relationship: despite its frequency in human tumours, aneuploidy is not a universal driver of cancer development and instead can exert substantial tumour-suppressive effects. The specific consequences of aneuploidy are highly context dependent and are influenced by a cell's genetic and environmental milieu. In this Review, we discuss the diverse facets of cancer biology that are shaped by aneuploidy, including metastasis, drug resistance and immune recognition, and we highlight aneuploidy's distinct roles as both a tumour promoter and an anticancer vulnerability.
Collapse
|
35
|
Cohen-Sharir Y, McFarland JM, Abdusamad M, Marquis C, Bernhard SV, Kazachkova M, Tang H, Ippolito MR, Laue K, Zerbib J, Malaby HLH, Jones A, Stautmeister LM, Bockaj I, Wardenaar R, Lyons N, Nagaraja A, Bass AJ, Spierings DCJ, Foijer F, Beroukhim R, Santaguida S, Golub TR, Stumpff J, Storchová Z, Ben-David U. Aneuploidy renders cancer cells vulnerable to mitotic checkpoint inhibition. Nature 2021; 590:486-491. [PMID: 33505028 PMCID: PMC8262644 DOI: 10.1038/s41586-020-03114-6] [Citation(s) in RCA: 164] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 11/19/2020] [Indexed: 01/30/2023]
Abstract
Selective targeting of aneuploid cells is an attractive strategy for cancer treatment1. However, it is unclear whether aneuploidy generates any clinically relevant vulnerabilities in cancer cells. Here we mapped the aneuploidy landscapes of about 1,000 human cancer cell lines, and analysed genetic and chemical perturbation screens2-9 to identify cellular vulnerabilities associated with aneuploidy. We found that aneuploid cancer cells show increased sensitivity to genetic perturbation of core components of the spindle assembly checkpoint (SAC), which ensures the proper segregation of chromosomes during mitosis10. Unexpectedly, we also found that aneuploid cancer cells were less sensitive than diploid cells to short-term exposure to multiple SAC inhibitors. Indeed, aneuploid cancer cells became increasingly sensitive to inhibition of SAC over time. Aneuploid cells exhibited aberrant spindle geometry and dynamics, and kept dividing when the SAC was inhibited, resulting in the accumulation of mitotic defects, and in unstable and less-fit karyotypes. Therefore, although aneuploid cancer cells could overcome inhibition of SAC more readily than diploid cells, their long-term proliferation was jeopardized. We identified a specific mitotic kinesin, KIF18A, whose activity was perturbed in aneuploid cancer cells. Aneuploid cancer cells were particularly vulnerable to depletion of KIF18A, and KIF18A overexpression restored their response to SAC inhibition. Our results identify a therapeutically relevant, synthetic lethal interaction between aneuploidy and the SAC.
Collapse
Affiliation(s)
- Yael Cohen-Sharir
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - James M McFarland
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Mai Abdusamad
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carolyn Marquis
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Sara V Bernhard
- Department of Molecular Genetics, TU Kaiserlautern, Kaiserlautern, Germany
| | - Mariya Kazachkova
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Helen Tang
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Marica R Ippolito
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
| | - Kathrin Laue
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Johanna Zerbib
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Heidi L H Malaby
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Andrew Jones
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Irena Bockaj
- European Research Institute for the Biology of Aging (ERIBA), University of Groningen, Groningen, The Netherlands
| | - René Wardenaar
- European Research Institute for the Biology of Aging (ERIBA), University of Groningen, Groningen, The Netherlands
| | - Nicholas Lyons
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ankur Nagaraja
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Adam J Bass
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Diana C J Spierings
- European Research Institute for the Biology of Aging (ERIBA), University of Groningen, Groningen, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Aging (ERIBA), University of Groningen, Groningen, The Netherlands
| | - Rameen Beroukhim
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Stefano Santaguida
- Department of Experimental Oncology at IEO, European Institute of Oncology IRCCS, Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Todd R Golub
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Dana Farber Cancer Institute, Boston, MA, USA
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Zuzana Storchová
- Department of Molecular Genetics, TU Kaiserlautern, Kaiserlautern, Germany
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
36
|
Funk LC, Wan J, Ryan SD, Kaur C, Sullivan R, Roopra A, Weaver BA. p53 Is Not Required for High CIN to Induce Tumor Suppression. Mol Cancer Res 2021; 19:112-123. [PMID: 32948674 PMCID: PMC7810023 DOI: 10.1158/1541-7786.mcr-20-0488] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/14/2020] [Accepted: 09/13/2020] [Indexed: 11/16/2022]
Abstract
Chromosomal instability (CIN) is a hallmark of cancer. While low levels of CIN can be tumor promoting, high levels of CIN cause cell death and tumor suppression. The widely used chemotherapeutic, paclitaxel (Taxol), exerts its anticancer effects by increasing CIN above a maximally tolerated threshold. One significant outstanding question is whether the p53 tumor suppressor is required for the cell death and tumor suppression caused by high CIN. Both p53 loss and reduction of the mitotic kinesin, centromere-associated protein-E, cause low CIN. Combining both genetic insults in the same cell leads to high CIN. Here, we test whether high CIN causes cell death and tumor suppression even in the absence p53. Despite a surprising sex-specific difference in tumor spectrum and latency in p53 heterozygous animals, these studies demonstrate that p53 is not required for high CIN to induce tumor suppression. Pharmacologic induction of high CIN results in equivalent levels of cell death due to loss of essential chromosomes in p53+/+ and p53-/- cells, further demonstrating that high CIN elicits cell death independently of p53 function. IMPLICATIONS: These results provide support for the efficacy of anticancer therapies that induce high CIN, even in tumors that lack functional p53.
Collapse
Affiliation(s)
- Laura C Funk
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Jun Wan
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Sean D Ryan
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Charanjeet Kaur
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin
| | - Ruth Sullivan
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
- Department of Comparative Biosciences, School of Veterinary Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Avtar Roopra
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
- Department of Neuroscience, University of Wisconsin-Madison, Madison, Wisconsin
| | - Beth A Weaver
- Department of Cell and Regenerative Biology, University of Wisconsin-Madison, Madison, Wisconsin.
- Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
- Department of Oncology/McArdle Laboratory for Cancer Research, University of Wisconsin-Madison, Madison, Wisconsin
| |
Collapse
|
37
|
Li X, He J, Yu M, Zhang W, Sun J. [BUB1 gene is highly expressed in gastric cancer:analysis based on Oncomine database and bioinformatics]. NAN FANG YI KE DA XUE XUE BAO = JOURNAL OF SOUTHERN MEDICAL UNIVERSITY 2020; 40:683-692. [PMID: 32897212 DOI: 10.12122/j.issn.1673-4254.2020.05.11] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
OBJECTIVE To investigate the expression of BUB1 gene in gastric cancer. METHODS Oncomine, GEPIA, BioGPS and Kaplan-Meier Plotter databases were used to analyze the difference of BUB1 gene expression between gastric cancer tissue and normal gastric tissue. The association of BUB1 expression level with the prognosis of gastric cancer patients was also analyzed. The Cancer Cell Line Encyclopedia (CCLE) was explored to analyze the expression of BUB1 in T cells and B cells in gastric cancer patients, and the String database was used to generate the network map of BUB1-related proteins and functional annotation of gene ontology (GO). The related pathways of KEGG were analyzed. Tumor immune assessment resource (TIMER) database was used to analyze the expression of BUB1 in immune infiltration and its effect on prognosis of gastric cancer patients. To further verify the results of gene chip analysis in Oncomine database, we collected 30 pairs of surgical specimens of gastric adenocarcinoma and adjacent tissues from patients admitted to the First Affiliated Hospital of Chengdu Medical College from March, 2018 to July, 2019. The results of BUB1 gene expression in Oncomine database were verified by PCR and immunohistochemistry. RESULTS Oncomine, GEPIA and BioGPS analyses showed that BUB1 was highly expressed in gastric cancer compared with normal gastric tissue. Kaplan-Meier survival analysis showed that the progression-free survival time (HR=0.52, 95% CI:0.41-0.67, P < 0.05) and the overall survival time (HR=0.67, 95% CI:0.55-0.82, P < 0.05) were prolonged in gastric cancer patients with a high expression of BUB1. Through String data collection, BUB1-related proteins were mainly enriched in 13 cellular components, 4 molecular functions and 12 biological processes, involving 4 signal pathways. TIMER database analysis showed that CD4+ T cells and macrophages with high expressions of BUB1 mRNA in the immune microenvironment were associated with a favorable 5-year survival outcome of patients with gastric cancer. In the surgical specimens, real-time quantitative PCR showed that the expression level of BUB1 mRNA was significantly higher in gastric cancer tissues than in the adjacent gastric mucosa tissues, and immunohistochemical results demonstrated positive BUB1 staining in the gastric cancer tissues. CONCLUSIONS BUB1 gene is highly expressed in gastric cancer. BUB1 may reduce tumor immunosuppression and helps to evaluate the prognosis of patients with gastric cancer.
Collapse
Affiliation(s)
- Xiaoyan Li
- Department of endocrinology, First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Jie He
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Mi Yu
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| | - Jian Sun
- Department of Respiratory and Critical Care Medicine, First Affiliated Hospital of Chengdu Medical College, Chengdu 610500, China
| |
Collapse
|
38
|
Zhou L, Jilderda LJ, Foijer F. Exploiting aneuploidy-imposed stresses and coping mechanisms to battle cancer. Open Biol 2020; 10:200148. [PMID: 32873156 PMCID: PMC7536071 DOI: 10.1098/rsob.200148] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Aneuploidy, an irregular number of chromosomes in cells, is a hallmark feature of cancer. Aneuploidy results from chromosomal instability (CIN) and occurs in almost 90% of all tumours. While many cancers display an ongoing CIN phenotype, cells can also be aneuploid without displaying CIN. CIN drives tumour evolution as ongoing chromosomal missegregation will yield a progeny of cells with variable aneuploid karyotypes. The resulting aneuploidy is initially toxic to cells because it leads to proteotoxic and metabolic stress, cell cycle arrest, cell death, immune cell activation and further genomic instability. In order to overcome these aneuploidy-imposed stresses and adopt a malignant fate, aneuploid cancer cells must develop aneuploidy-tolerating mechanisms to cope with CIN. Aneuploidy-coping mechanisms can thus be considered as promising therapeutic targets. However, before such therapies can make it into the clinic, we first need to better understand the molecular mechanisms that are activated upon aneuploidization and the coping mechanisms that are selected for in aneuploid cancer cells. In this review, we discuss the key biological responses to aneuploidization, some of the recently uncovered aneuploidy-coping mechanisms and some strategies to exploit these in cancer therapy.
Collapse
Affiliation(s)
| | | | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, 9713 AV, Groningen, The Netherlands
| |
Collapse
|
39
|
Khot M, Sreekumar D, Jahagirdar S, Kulkarni A, Hari K, Faseela EE, Sabarinathan R, Jolly MK, Sengupta K. Twist1 induces chromosomal instability (CIN) in colorectal cancer cells. Hum Mol Genet 2020; 29:1673-1688. [PMID: 32337580 PMCID: PMC7322571 DOI: 10.1093/hmg/ddaa076] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2020] [Revised: 04/22/2020] [Accepted: 04/22/2020] [Indexed: 12/22/2022] Open
Abstract
Twist1 is a basic helix-loop-helix transcription factor, essential during early development in mammals. While Twist1 induces epithelial-to-mesenchymal transition (EMT), here we show that Twist1 overexpression enhances nuclear and mitotic aberrations. This is accompanied by an increase in whole chromosomal copy number gains and losses, underscoring the role of Twist1 in inducing chromosomal instability (CIN) in colorectal cancer cells. Array comparative genomic hybridization (array CGH) analysis further shows sub-chromosomal deletions, consistent with an increased frequency of DNA double strand breaks (DSBs). Remarkably, Twist1 overexpression downmodulates key cell cycle checkpoint factors-Bub1, BubR1, Mad1 and Mad2-that regulate CIN. Mathematical simulations using the RACIPE tool show a negative correlation of Twist1 with E-cadherin and BubR1. Data analyses of gene expression profiles of patient samples from The Cancer Genome Atlas (TCGA) reveal a positive correlation between Twist1 and mesenchymal genes across cancers, whereas the correlation of TWIST1 with CIN and DSB genes is cancer subtype-specific. Taken together, these studies highlight the mechanistic involvement of Twist1 in the deregulation of factors that maintain genome stability during EMT in colorectal cancer cells. Twist1 overexpression enhances genome instability in the context of EMT that further contributes to cellular heterogeneity. In addition, these studies imply that Twist1 downmodulates nuclear lamins that further alter spatiotemporal organization of the cancer genome and epigenome. Notwithstanding their genetic background, colorectal cancer cells nevertheless maintain their overall ploidy, while the downstream effects of Twist1 enhance CIN and DNA damage enriching for sub-populations of aggressive cancer cells.
Collapse
Affiliation(s)
- Maithilee Khot
- B-216, Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune 411008, India
| | - Dyuthi Sreekumar
- B-216, Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune 411008, India
| | - Sanika Jahagirdar
- B-216, Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune 411008, India
| | - Apoorva Kulkarni
- B-216, Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune 411008, India
| | - Kishore Hari
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | | | - Radhakrishnan Sabarinathan
- National Centre for Biological Sciences, Tata Institute of Fundamental Research, Bengaluru 560065, India
| | - Mohit Kumar Jolly
- Center for BioSystems Science and Engineering, Indian Institute of Science, Bengaluru 560012, India
| | - Kundan Sengupta
- B-216, Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune 411008, India
- To whom correspondence should be addressed at: B-216, Chromosome Biology Lab (CBL), Indian Institute of Science Education and Research (IISER), Dr Homi Bhabha Road, Pashan, Pune 411008, India. Tel: +91 20 25908071; Fax: +91-20-20251566;
| |
Collapse
|
40
|
Hoevenaar WHM, Janssen A, Quirindongo AI, Ma H, Klaasen SJ, Teixeira A, van Gerwen B, Lansu N, Morsink FHM, Offerhaus GJA, Medema RH, Kops GJPL, Jelluma N. Degree and site of chromosomal instability define its oncogenic potential. Nat Commun 2020; 11:1501. [PMID: 32198375 PMCID: PMC7083897 DOI: 10.1038/s41467-020-15279-9] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 02/27/2020] [Indexed: 12/17/2022] Open
Abstract
Most human cancers are aneuploid, due to a chromosomal instability (CIN) phenotype. Despite being hallmarks of cancer, however, the roles of CIN and aneuploidy in tumor formation have not unequivocally emerged from animal studies and are thus still unclear. Using a conditional mouse model for diverse degrees of CIN, we find that a particular range is sufficient to drive very early onset spontaneous adenoma formation in the intestine. In mice predisposed to intestinal cancer (ApcMin/+), moderate CIN causes a remarkable increase in adenoma burden in the entire intestinal tract and especially in the distal colon, which resembles human disease. Strikingly, a higher level of CIN promotes adenoma formation in the distal colon even more than moderate CIN does, but has no effect in the small intestine. Our results thus show that CIN can be potently oncogenic, but that certain levels of CIN can have contrasting effects in distinct tissues.
Collapse
Affiliation(s)
- Wilma H M Hoevenaar
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Aniek Janssen
- Center for Molecular Medicine, Section Molecular Cancer Research, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ajit I Quirindongo
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Huiying Ma
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Sjoerd J Klaasen
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Antoinette Teixeira
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bastiaan van Gerwen
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Nico Lansu
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands
| | - Folkert H M Morsink
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - G Johan A Offerhaus
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - René H Medema
- Division of Cell Biology, Netherlands Cancer Institute, Oncode Institute, Amsterdam, The Netherlands
| | - Geert J P L Kops
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.
| | - Nannette Jelluma
- Oncode Institute, Hubrecht Institute-KNAW and University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
41
|
Genetic Changes in Experimental Populations of a Hybrid in the Cryptococcus neoformans Species Complex. Pathogens 2019; 9:pathogens9010003. [PMID: 31861437 PMCID: PMC7168662 DOI: 10.3390/pathogens9010003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/14/2019] [Accepted: 12/16/2019] [Indexed: 01/19/2023] Open
Abstract
Hybrids between Cryptococcus neoformans and Cryptococcus deneoformans are commonly found in patients and the environment. However, the genetic stability of these hybrids remains largely unknown. Here, we established mutation accumulation lines of a diploid C. neoformans × C. deneoformans laboratory hybrid and analyzed the genotypes at 33 markers distributed across all 14 chromosomes. Our analyses found that under standard culture conditions, heterozygosity at most loci was maintained over 800 mitotic generations, with an estimated 6.44 × 10−5 loss-of-heterozygosity (LoH) event per mitotic division. However, under fluconazole stress, the observed LoH frequency increased by > 50 folds for the two markers on Chromosome 1, all due to the loss of the fluconazole susceptible allele on this chromosome. Flow cytometry analyses showed that after the 40th transfer (120 days), 19 of the 20 lines maintained the original ploidy level (2N), while one line was between 2N and 3N. The combined flow cytometry, genotyping at 33 markers, and quantitative PCR analyses showed the allelic loss was compensated for by amplification of the resistant ERG11 allele in eight of the ten fluconazole-stress lines. Our results suggest that hybrids in C. neoformans species complex are generally stable but that they can undergo rapid adaptation to environmental stresses through LoH and gene duplication.
Collapse
|
42
|
Lorca V, Garre P. Current status of the genetic susceptibility in attenuated adenomatous polyposis. World J Gastrointest Oncol 2019; 11:1101-1114. [PMID: 31908716 PMCID: PMC6937445 DOI: 10.4251/wjgo.v11.i12.1101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 08/18/2019] [Accepted: 10/14/2019] [Indexed: 02/05/2023] Open
Abstract
Adenomatous polyposis (AP) is classified according to cumulative adenoma number in classical AP (CAP) and attenuated AP (AAP). Genetic susceptibility is the major risk factor in CAP due to mutations in the known high predisposition genes APC and MUTYH. However, the contribution of genetic susceptibility to AAP is lower and less understood. New predisposition genes have been recently proposed, and some of them have been validated, but their scarcity hinders accurate risk estimations and prevalence calculations. AAP is a heterogeneous condition in terms of severity, clinical features and heritability. Therefore, clinicians do not have strong discriminating criteria for the recommendation of the genetic study of known predisposition genes, and the detection rate is low. Elucidation and knowledge of new AAP high predisposition genes are of great importance to offer accurate genetic counseling to the patient and family members. This review aims to update the genetic knowledge of AAP, and to expound the difficulties involved in the genetic analysis of a highly heterogeneous condition such as AAP.
Collapse
Affiliation(s)
- Víctor Lorca
- Laboratorio de Oncología Molecular, Grupo de Investigación Clínica y Traslacional en Oncología, Hospital Clínico San Carlos, Madrid 28040, Spain
| | - Pilar Garre
- Laboratorio de Oncología Molecular, Servicio de Oncología, Hospital Clínico San Carlos, Madrid 28040, Spain
| |
Collapse
|
43
|
Njeru SN, Kraus J, Meena JK, Lechel A, Katz SF, Kumar M, Knippschild U, Azoitei A, Wezel F, Bolenz C, Leithäuser F, Gollowitzer A, Omrani O, Hoischen C, Koeberle A, Kestler HA, Günes C, Rudolph KL. Aneuploidy-inducing gene knockdowns overlap with cancer mutations and identify Orp3 as a B-cell lymphoma suppressor. Oncogene 2019; 39:1445-1465. [PMID: 31659255 DOI: 10.1038/s41388-019-1073-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 09/25/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022]
Abstract
Aneuploidy can instigate tumorigenesis. However, mutations in genes that control chromosome segregation are rare in human tumors as these mutations reduce cell fitness. Screening experiments indicate that the knockdown of multiple classes of genes that are not directly involved in chromosome segregation can lead to aneuploidy induction. The possible contribution of these genes to cancer formation remains yet to be defined. Here we identified gene knockdowns that lead to an increase in aneuploidy in checkpoint-deficient human cancer cells. Computational analysis revealed that the identified genes overlap with recurrent mutations in human cancers. The knockdown of the three strongest selected candidate genes (ORP3, GJB3, and RXFP1) enhances the malignant transformation of human fibroblasts in culture. Furthermore, the knockout of Orp3 results in an aberrant expansion of lymphoid progenitor cells and a high penetrance formation of chromosomal instable, pauci-clonal B-cell lymphoma in aging mice. At pre-tumorous stages, lymphoid cells from the animals exhibit deregulated phospholipid metabolism and an aberrant induction of proliferation regulating pathways associating with increased aneuploidy in hematopoietic progenitor cells. Together, these results support the concept that aneuploidy-inducing gene deficiencies contribute to cellular transformation and carcinogenesis involving the deregulation of various molecular processes such as lipid metabolism, proliferation, and cell survival.
Collapse
Affiliation(s)
- Sospeter N Njeru
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany.,Paul-Ehrlich-Institute, Division Immunology, 63225, Langen, Germany
| | - Johann Kraus
- Institute of Medical Systems Biology, Ulm University, 89081, Ulm, Germany
| | - Jitendra K Meena
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany.,Baylor College of Medicine, Houston, TX, USA
| | - André Lechel
- Department of Internal Medicine I, Ulm University Hospital, 89081, Ulm, Germany
| | - Sarah-Fee Katz
- Department of Internal Medicine I, Ulm University Hospital, 89081, Ulm, Germany
| | - Mukesh Kumar
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, 89081, Ulm, Germany
| | - Anca Azoitei
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany
| | - Felix Wezel
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany
| | - Christian Bolenz
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany
| | | | - André Gollowitzer
- Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Omid Omrani
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany
| | - Christian Hoischen
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany
| | - Andreas Koeberle
- Institute of Pharmacy, Friedrich-Schiller-University Jena, 07743, Jena, Germany.,Michael Popp Research Institute, University of Innsbruck, Innsbruck, Austria
| | - Hans A Kestler
- Institute of Medical Systems Biology, Ulm University, 89081, Ulm, Germany.
| | - Cagatay Günes
- Department of Urology, Ulm University Hospital, 89081, Ulm, Germany.
| | - K Lenhard Rudolph
- Leibniz Institute on Aging, Fritz Lipmann Institute e.V., 07745, Jena, Germany.
| |
Collapse
|
44
|
Ben-David U, Amon A. Context is everything: aneuploidy in cancer. Nat Rev Genet 2019; 21:44-62. [DOI: 10.1038/s41576-019-0171-x] [Citation(s) in RCA: 234] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/14/2019] [Indexed: 02/07/2023]
|
45
|
Shastri S, Chatterjee B, Thakur SS. Achievements in Cancer Research and its Therapeutics in Hundred Years. Curr Top Med Chem 2019; 19:1545-1562. [PMID: 31362690 DOI: 10.2174/1568026619666190730093034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Revised: 07/16/2019] [Accepted: 07/18/2019] [Indexed: 12/19/2022]
Abstract
Cancer research has progressed leaps and bounds over the years. This review is a brief overview of the cancer research, milestone achievements and therapeutic studies on it over the one hundred ten years which would give us an insight into how far we have come to understand and combat this fatal disease leading to millions of deaths worldwide. Modern biology has proved that cancer is a very complex disease as still we do not know precisely how it triggers. It involves several factors such as protooncogene, oncogene, kinase, tumor suppressor gene, growth factor, signalling cascade, micro RNA, immunity, environmental factors and carcinogens. However, modern technology now helps the cancer patient on the basis of acquired and established knowledge in the last hundred years to save human lives.
Collapse
Affiliation(s)
- Sravanthi Shastri
- Proteomics and Cell Signaling, Centre for Cellular and Molecular Biology, Hyderabad, India
| | - Bhaswati Chatterjee
- National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Suman S Thakur
- Proteomics and Cell Signaling, Centre for Cellular and Molecular Biology, Hyderabad, India
| |
Collapse
|
46
|
Laucius CD, Orr B, Compton DA. Chromosomal instability suppresses the growth of K-Ras-induced lung adenomas. Cell Cycle 2019; 18:1702-1713. [PMID: 31179849 DOI: 10.1080/15384101.2019.1629790] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022] Open
Abstract
Chromosomal instability (CIN) is defined as a high rate of whole chromosome loss or gain and is a hallmark of many aneuploid solid tumors. CIN positively correlates with poor patient prognosis and chemotherapeutic resistance. Despite this clinical importance, the role of CIN in tumor initiation, growth and/or progression remains poorly understood. To date, the only strategies developed to determine how CIN contributes to tumorigenesis have relied on transgenic mouse models that deliberately increase the rate of chromosomal mis-segregation. Here we develop a strain of transgenic mice that is designed to strategically decrease the rate of chromosome mis-segregation and suppress CIN. These animals modestly overexpress the kinesin-13 microtubule depolymerase Kif2b, a strategy proven successful in restoring faithful chromosome segregation to human cancer cells in culture. Using the LA2 K-Ras G12D-induced model for lung cancer, we show that Kif2b expression reduces the number of chromosome segregation defects but does not change the incidence of lung tumor lesions. However, pulmonary tumors were significantly larger in animals expressing Kif2b and those tumors exhibited elevated rates of Ki-67 positive cells relative to controls. Thus, in lung cancers driven by mutations in K-Ras, CIN has little impact on tumor initiation but suppresses tumor growth. These data support a model in which CIN imposes a burden on tumor cells, and that enhancement of mitotic fidelity results in accelerated tumor growth.
Collapse
Affiliation(s)
- Christopher D Laucius
- a Department of Biochemistry , Geisel School of Medicine at Dartmouth , Hanover , NH , USA.,b Norris Cotton Cancer Center , Lebanon , NH , USA
| | - Bernardo Orr
- a Department of Biochemistry , Geisel School of Medicine at Dartmouth , Hanover , NH , USA.,b Norris Cotton Cancer Center , Lebanon , NH , USA
| | - Duane A Compton
- a Department of Biochemistry , Geisel School of Medicine at Dartmouth , Hanover , NH , USA.,b Norris Cotton Cancer Center , Lebanon , NH , USA
| |
Collapse
|
47
|
Beh TT, Kalitsis P. The Role of Centromere Defects in Cancer. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 56:541-554. [PMID: 28840252 DOI: 10.1007/978-3-319-58592-5_22] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/11/2023]
Abstract
The accurate segregation of chromosomes to daughter cells is essential for healthy development to occur. Imbalances in chromosome number have long been associated with cancers amongst other medical disorders. Little is known whether abnormal chromosome numbers are an early contributor to the cancer progression pathway. Centromere DNA and protein defects are known to impact on the fidelity of chromosome segregation in cell and model systems. In this chapter we discuss recent developments in understanding the contribution of centromere abnormalities at the protein and DNA level and their role in cancer in human and mouse systems.
Collapse
Affiliation(s)
- Thian Thian Beh
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Melbourne, 3052, Australia.,Department of Paediatrics, University of Melbourne, Parkville, Melbourne, 3052, Australia
| | - Paul Kalitsis
- Murdoch Childrens Research Institute, Royal Children's Hospital, Parkville, Melbourne, 3052, Australia. .,Department of Paediatrics, University of Melbourne, Parkville, Melbourne, 3052, Australia.
| |
Collapse
|
48
|
Pagotto S, Veronese A, Soranno A, Balatti V, Ramassone A, Guanciali-Franchi PE, Palka G, Innocenti I, Autore F, Rassenti LZ, Kipps TJ, Mariani-Costantini R, Laurenti L, Croce CM, Visone R. HNRNPL Restrains miR-155 Targeting of BUB1 to Stabilize Aberrant Karyotypes of Transformed Cells in Chronic Lymphocytic Leukemia. Cancers (Basel) 2019; 11:575. [PMID: 31018621 PMCID: PMC6520824 DOI: 10.3390/cancers11040575] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/15/2019] [Accepted: 04/19/2019] [Indexed: 11/18/2022] Open
Abstract
Aneuploidy and overexpression of hsa-miR-155-5p (miR-155) characterize most solid and hematological malignancies. We recently demonstrated that miR-155 sustains aneuploidy at early stages of in vitro cellular transformation. During in vitro transformation of normal human fibroblast, upregulation of miR-155 downregulates spindle checkpoint proteins as the mitotic checkpoint serine/threonine kinase budding uninhibited by benzimidazoles 1 (BUB1), the centromere protein F (CENPF) and the zw10 kinetochore protein (ZW10), compromising the chromosome alignment at the metaphase plate and leading to aneuploidy in daughter cells. Here we show that the heterogeneous nuclear ribonucleoprotein L (HNRNPL) binds to the polymorphic marker D2S1888 at the 3'UTR of BUB1 gene, impairs the miR-155 targeting, and restores BUB1 expression in chronic lymphocytic leukemia. This mechanism occurs at advanced passages of cell transformation and allows the expansion of more favorable clones. Our findings have revealed, at least in part, the molecular mechanisms behind the chromosomal stabilization of cell lines and the concept that, to survive, tumor cells cannot continuously change their genetic heritage but need to stabilize the most suitable karyotype.
Collapse
Affiliation(s)
- Sara Pagotto
- Ageing Research Center and Translational medicine-CeSI-MeT, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University Chieti-Pescara, 66100 Chieti, Italy.
| | - Angelo Veronese
- Ageing Research Center and Translational medicine-CeSI-MeT, 66100 Chieti, Italy.
- Department of Medicine and Aging Sciences, "G. d'Annunzio" University Chieti-Pescara, 66100 Chieti, Italy.
| | - Alessandra Soranno
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University Chieti-Pescara, 66100 Chieti, Italy.
| | - Veronica Balatti
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
| | - Alice Ramassone
- Ageing Research Center and Translational medicine-CeSI-MeT, 66100 Chieti, Italy.
- Department of Medicine and Aging Sciences, "G. d'Annunzio" University Chieti-Pescara, 66100 Chieti, Italy.
| | - Paolo E Guanciali-Franchi
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University Chieti-Pescara, 66100 Chieti, Italy.
| | - Giandomenico Palka
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University Chieti-Pescara, 66100 Chieti, Italy.
| | - Idanna Innocenti
- Institute of Hematology, Catholic University of the Sacred Heart, 00168 Rome, Italy.
| | - Francesco Autore
- Institute of Hematology, Catholic University of the Sacred Heart, 00168 Rome, Italy.
| | - Laura Z Rassenti
- Department of Medicine, Moores Cancer Center, University of California at San Diego, La Jolla, CA 92093, USA.
- Chronic Lymphocytic Leukemia Research Consortium, San Diego, CA 92093, USA.
| | - Thomas J Kipps
- Department of Medicine, Moores Cancer Center, University of California at San Diego, La Jolla, CA 92093, USA.
- Chronic Lymphocytic Leukemia Research Consortium, San Diego, CA 92093, USA.
| | - Renato Mariani-Costantini
- Ageing Research Center and Translational medicine-CeSI-MeT, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University Chieti-Pescara, 66100 Chieti, Italy.
| | - Luca Laurenti
- Institute of Hematology, Catholic University of the Sacred Heart, 00168 Rome, Italy.
| | - Carlo M Croce
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA.
- Chronic Lymphocytic Leukemia Research Consortium, San Diego, CA 92093, USA.
| | - Rosa Visone
- Ageing Research Center and Translational medicine-CeSI-MeT, 66100 Chieti, Italy.
- Department of Medical, Oral and Biotechnological Sciences, "G. d'Annunzio" University Chieti-Pescara, 66100 Chieti, Italy.
| |
Collapse
|
49
|
Lynch AM, Eastmond D, Elhajouji A, Froetschl R, Kirsch-Volders M, Marchetti F, Masumura K, Pacchierotti F, Schuler M, Tweats D. Targets and mechanisms of chemically induced aneuploidy. Part 1 of the report of the 2017 IWGT workgroup on assessing the risk of aneugens for carcinogenesis and hereditary diseases. MUTATION RESEARCH-GENETIC TOXICOLOGY AND ENVIRONMENTAL MUTAGENESIS 2019; 847:403025. [PMID: 31699346 DOI: 10.1016/j.mrgentox.2019.02.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 01/22/2019] [Accepted: 02/20/2019] [Indexed: 02/06/2023]
Abstract
An aneuploidy workgroup was established as part of the 7th International Workshops on Genotoxicity Testing. The workgroup conducted a review of the scientific literature on the biological mechanisms of aneuploidy in mammalian cells and methods used to detect chemical aneugens. In addition, the current regulatory framework was discussed, with the objective to arrive at consensus statements on the ramifications of exposure to chemical aneugens for human health risk assessment. As part of these efforts, the workgroup explored the use of adverse outcome pathways (AOPs) to document mechanisms of chemically induced aneuploidy in mammalian somatic cells. The group worked on two molecular initiating events (MIEs), tubulin binding and binding to the catalytic domain of aurora kinase B, which result in several adverse outcomes, including aneuploidy. The workgroup agreed that the AOP framework provides a useful approach to link evidence for MIEs with aneuploidy on a cellular level. The evidence linking chemically induced aneuploidy with carcinogenicity and hereditary disease was also reviewed and is presented in two companion papers. In addition, the group came to the consensus that the current regulatory test batteries, while not ideal, are sufficient for the identification of aneugens and human risk assessment. While it is obvious that there are many different MIEs that could lead to the induction of aneuploidy, the most commonly observed mechanisms involving chemical aneugens are related to tubulin binding and, to a lesser extent, inhibition of mitotic kinases. The comprehensive review presented here should help with the identification and risk management of aneugenic agents.
Collapse
Affiliation(s)
| | | | - Azeddine Elhajouji
- Novartis Institutes for Biomedical Research, Preclinical Safety, Basel, Switzerland
| | | | | | - Francesco Marchetti
- Environmental Health Science and Research Bureau, Health Canada, Ottawa, ON, Canada
| | - Kenichi Masumura
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Kanagawa, Japan
| | - Francesca Pacchierotti
- Health Protection Technology Division, Laboratory of Biosafety and Risk Assessment, ENEA, CR Casaccia, Rome, Italy
| | | | | |
Collapse
|
50
|
Choi E, Yu H. Spindle Checkpoint Regulators in Insulin Signaling. Front Cell Dev Biol 2018; 6:161. [PMID: 30555826 PMCID: PMC6281718 DOI: 10.3389/fcell.2018.00161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 11/08/2018] [Indexed: 12/17/2022] Open
Abstract
The spindle checkpoint ensures accurate chromosome segregation during mitosis and guards against aneuploidy. Insulin signaling governs metabolic homeostasis and cell growth, and its dysregulation leads to metabolic disorders, such as diabetes. These critical pathways have been extensively investigated, but a link between the two has not been established until recently. Our recent study reveals a critical role of spindle checkpoint regulators in insulin signaling and metabolic homeostasis through regulating endocytosis of the insulin receptor (IR). These findings have linked spindle checkpoint proteins to metabolic regulation, expanding the connection between cell division and metabolism. Here, we briefly review the unexpected roles of spindle checkpoint regulators in vesicle trafficking and insulin signaling.
Collapse
Affiliation(s)
- Eunhee Choi
- Department of Pharmacology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| | - Hongtao Yu
- Department of Pharmacology, Howard Hughes Medical Institute, University of Texas Southwestern Medical Center, Dallas, TX, United States
| |
Collapse
|