1
|
Wang F, Gui W, Rong M, Zhang L, Wu J, Li J, Wang R, Gouttia OG, Wang L, Yang X, Peng A. TOX High Mobility Group Box Family Member 4 Promotes DNA Double Strand Break Repair via Non-Homologous End Joining. J Biol Chem 2025:110174. [PMID: 40328361 DOI: 10.1016/j.jbc.2025.110174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 03/25/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025] Open
Abstract
Non-homologous end joining (NHEJ) is a pivotal mechanism in the repair of DNA double-strand breaks (DSBs). Central to NHEJ is the DNA-dependent protein kinase (DNA-PK) complex, comprising the KU heterodimer and the catalytic subunit, DNA-PKcs. In this study, we characterize TOX High Mobility Group Box Family Member 4 (TOX4) as a factor recruited to both laser-induced DNA damage and endonuclease-induced DNA DSBs. Depletion of TOX4 leads to accumulation of DNA damage, which is epistatic to DNA-PKcs. Consistently, TOX4 depletion substantially reduces NHEJ efficiency measured using both intrachromosomal and extrachromosomal repair assays. Our proteomic and biochemical analyses reveal TOX4 association with DNA-PK that is required for DNA-PKcs activation. Furthermore, we show that TOX4 coordinates with Phosphatase 1 Nuclear-Targeting Subunit (PNUTS) in NHEJ. PNUTS, previously shown to protect DNA-PKcs phosphorylation from protein phosphatase 1 (PP1)-mediated dephosphorylation, binds DNA-PK in a TOX4-dependent manner. In line with its role in DNA repair, TOX4 emerges as a promising target for anti-cancer drug development, and its targeting enhances tumor cell sensitivity to DNA damage in head and neck cancer and other malignancies.
Collapse
Affiliation(s)
- Feifei Wang
- Institute of Health Sciences and Technology, Institutes of Physical Sciences and Information Technology, Anhui University. Hefei, Anhui, China.
| | - Wenli Gui
- Institute of Health Sciences and Technology, Institutes of Physical Sciences and Information Technology, Anhui University. Hefei, Anhui, China
| | - Mengtao Rong
- Institute of Health Sciences and Technology, Institutes of Physical Sciences and Information Technology, Anhui University. Hefei, Anhui, China
| | - Liang Zhang
- Department of Orthopedics, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, P.R. China
| | - Jiajing Wu
- Institute of Health Sciences and Technology, Institutes of Physical Sciences and Information Technology, Anhui University. Hefei, Anhui, China
| | - Juan Li
- Institute of Health Sciences and Technology, Institutes of Physical Sciences and Information Technology, Anhui University. Hefei, Anhui, China
| | - Renqing Wang
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230022, China
| | - Odjo G Gouttia
- Department of Biomedical Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill. Chapel Hill, NC, USA
| | - Ling Wang
- Department of Biomedical Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill. Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill. Chapel Hill, NC, USA
| | - Xingyuan Yang
- Institute of Health Sciences and Technology, Institutes of Physical Sciences and Information Technology, Anhui University. Hefei, Anhui, China.
| | - Aimin Peng
- Department of Biomedical Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill. Chapel Hill, NC, USA; Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill. Chapel Hill, NC, USA.
| |
Collapse
|
2
|
Liu Y, Sun W, Liu L, Cheng J, Li J, Huang Z, Ouyang M. Elevated KIF2C Expression Drives Osteosarcoma Progression by Modulating the Wnt/β-Catenin Signaling Pathway and Contributing to an Immunosuppressive Tumor Microenvironment. Cancer Med 2025; 14:e70915. [PMID: 40292920 PMCID: PMC12035763 DOI: 10.1002/cam4.70915] [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: 01/14/2025] [Revised: 04/01/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Although kinesin family member 2C (KIF2C) is implicated in various cancers, its role in osteosarcoma (OS) and the associated inflammatory microenvironment remains unclear. METHODS Publicly available datasets were analyzed to determine KIF2C expression, diagnostic value, and prognostic relevance in OS. In vitro (proliferation, colony formation, apoptosis, migration, invasion) and in vivo assays assessed its biological functions. KEGG enrichment and GSVA explored underlying pathways. ssGSEA, ESTIMATE algorithms, and single-cell sequencing evaluated the immune context, and molecular docking and molecular dynamics identified potential inhibitory compounds. RESULTS KIF2C was significantly overexpressed in OS, effectively distinguishing OS from normal tissues. Elevated KIF2C levels correlated with poor survival outcomes. Silencing KIF2C suppressed OS cell proliferation, migration, invasion, and in vivo tumor growth, while promoting apoptosis; conversely, overexpression of KIF2C had the opposite effect. Mechanistically, co-immunoprecipitation results indicated that KIF2C can bind to β-catenin to regulate the Wnt/β-catenin pathway. Furthermore, high KIF2C expression was associated with an immunosuppressive tumor microenvironment characterized by immune exhaustion. Molecular docking and molecular dynamics suggested butein as a candidate small-molecule inhibitor targeting KIF2C-related oncogenic mechanisms. CONCLUSION KIF2C drives OS progression by enhancing Wnt/β-catenin signaling and fostering an immunosuppressive microenvironment. Targeting KIF2C may offer new therapeutic approaches in managing OS.
Collapse
Affiliation(s)
- Ya‐Yun Liu
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Wu Sun
- Discipline of Chinese and Western Integrative MedicineJiangxi University of Chinese MedicineNanchangPeople's Republic of China
| | - Lin Liu
- Department of NursingJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Jin‐Hui Cheng
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Jing‐Tang Li
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Zu‐Tai Huang
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| | - Min Ouyang
- Department of OrthopaedicsJiangxi Provincial People's Hospital, the First Affiliated Hospital of Nanchang Medical CollegeNanchangPeople's Republic of China
| |
Collapse
|
3
|
Pao YS, Liao KJ, Shiau YC, Chao MH, Li MC, Lin LM, Chang HH, Yeh HW, Chen YJ, Chiu YT, Pan MYC, Chang YH, Shen SY, Lin SY, Cheng HC, Lin YC, Sun YJ, Kuo CC, Hsieh HP, Wang LHC. KIF2C promotes paclitaxel resistance by depolymerizing polyglutamylated microtubules. Dev Cell 2025:S1534-5807(25)00151-0. [PMID: 40157365 DOI: 10.1016/j.devcel.2025.03.004] [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: 03/11/2024] [Revised: 11/27/2024] [Accepted: 03/04/2025] [Indexed: 04/01/2025]
Abstract
The long-term effectiveness of paclitaxel is limited by chemoresistance. In this study, we elucidate the molecular mechanism by which kinesin family member 2C (KIF2C), a well-known microtubule depolymerase, contributes to the development of chemoresistance in triple-negative breast cancer (TNBC). We observed elevated levels of KIF2C, tubulin tyrosination, and polyglutamylation in human and mouse breast cancer cells resistant to paclitaxel. Additionally, these chemoresistant cells possessed cross-resistance to diverse microtubule-targeting agents (MTAs). We demonstrated that KIF2C preferentially depolymerizes polyglutamylated tubulin, even in the presence of paclitaxel. To counter this, we developed 7S9, a chemical inhibitor of KIF2C, that prohibits the dissociation of KIF2C from microtubules. The combination of 7S9 and paclitaxel significantly reduced tumorigenesis in chemoresistant TNBC model in mice. Moreover, 7S9 diminished cancer cell chemoresistance to several clinically available MTAs. Our findings elucidate the molecular mechanism of KIF2C-mediated chemoresistance and highlight KIF2C as a promising target for combating cross-resistance in TNBC.
Collapse
Affiliation(s)
- Yuan-Shao Pao
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Kuan-Ju Liao
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Ya-Chia Shiau
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Ming-Hong Chao
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Mu-Chun Li
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei City 115202, Taiwan
| | - Li-Mei Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Hsin-Huei Chang
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Hung-Wei Yeh
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Yi-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei City 115202, Taiwan
| | - Yu-Ting Chiu
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Max Yu-Chen Pan
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Yu-Hsuan Chang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Shih-Yu Shen
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Shu-Yu Lin
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan
| | - Hui-Chun Cheng
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Yu-Chun Lin
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu City 300044, Taiwan; Department of Medical Sciences, National Tsing Hua University, Hsinchu City 300044, Taiwan
| | - Yuh-Ju Sun
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan.
| | - Ching-Chuan Kuo
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan.
| | - Hsing-Pang Hsieh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 350401, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu City 300044, Taiwan; Biomedical Translation Research Center, Academia Sinica, Taipei City 115202, Taiwan.
| | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu City 300044, Taiwan; Department of Medical Sciences, National Tsing Hua University, Hsinchu City 300044, Taiwan; School of Medicine, National Tsing Hua University, Hsinchu City 300044, Taiwan.
| |
Collapse
|
4
|
Chiolo I, Altmeyer M, Legube G, Mekhail K. Nuclear and genome dynamics underlying DNA double-strand break repair. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00828-1. [PMID: 40097581 DOI: 10.1038/s41580-025-00828-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/21/2025] [Indexed: 03/19/2025]
Abstract
Changes in nuclear shape and in the spatial organization of chromosomes in the nucleus commonly occur in cancer, ageing and other clinical contexts that are characterized by increased DNA damage. However, the relationship between nuclear architecture, genome organization, chromosome stability and health remains poorly defined. Studies exploring the connections between the positioning and mobility of damaged DNA relative to various nuclear structures and genomic loci have revealed nuclear and cytoplasmic processes that affect chromosome stability. In this Review, we discuss the dynamic mechanisms that regulate nuclear and genome organization to promote DNA double-strand break (DSB) repair, genome stability and cell survival. Genome dynamics that support DSB repair rely on chromatin states, repair-protein condensates, nuclear or cytoplasmic microtubules and actin filaments, kinesin or myosin motor proteins, the nuclear envelope, various nuclear compartments, chromosome topology, chromatin loop extrusion and diverse signalling cues. These processes are commonly altered in cancer and during natural or premature ageing. Indeed, the reshaping of the genome in nuclear space during DSB repair points to new avenues for therapeutic interventions that may take advantage of new cancer cell vulnerabilities or aim to reverse age-associated defects.
Collapse
Affiliation(s)
- Irene Chiolo
- Department of Molecular and Computational Biology, University of Southern California, Los Angeles, CA, USA.
| | - Matthias Altmeyer
- Department of Molecular Mechanisms of Disease, University of Zurich (UZH), Zurich, Switzerland.
| | - Gaëlle Legube
- MCD, Centre de Biologie Intégrative (CBI), CNRS, Université de Toulouse, UT3, Toulouse, France.
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
5
|
Solomon AD, Gouttia OG, Wang L, Zhu S, Wang F, Li Y, Paydar M, Bessho T, Kwok BH, Peng A. γ-tubulin mediates DNA double-strand break repair. J Cell Sci 2025; 138:jcs262255. [PMID: 40135584 PMCID: PMC12050090 DOI: 10.1242/jcs.262255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 12/20/2024] [Indexed: 03/27/2025] Open
Abstract
Double-strand breaks (DSBs) in DNA pose a critical threat to genomic integrity, potentially leading to the onset and progression of various diseases, including cancer. Cellular responses to such lesions entail sophisticated repair mechanisms primarily mediated by non-homologous end joining (NHEJ) and homologous recombination (HR). Interestingly, the efficient recruitment of repair proteins and completion of DSB repair likely involve complex, inter-organelle communication and coordination of cellular components. In this study, we report a role of γ-tubulin in DSB repair. γ-tubulin is a major microtubule nucleation factor governing microtubule dynamics. We show that γ-tubulin is recruited to the site of DNA damage and is required for efficient DSB repair via both NHEJ and HR. Suppression of γ-tubulin impedes DNA repair and exacerbates DNA damage accumulation. Furthermore, γ-tubulin mediates the mobilization and formation of DNA damage foci, which serve as repair centers, thereby facilitating the recruitment of HR and NHEJ repair proteins on damaged chromatin. Finally, pharmacological inhibition of γ-tubulin enhances the cytotoxic effect of DNA-damaging agents, consistent with the DNA repair function of γ-tubulin, and underscoring the potential of its therapeutic intervention in cancer therapy.
Collapse
Affiliation(s)
- Abhishikt David Solomon
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Odjo G. Gouttia
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Ling Wang
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Songli Zhu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Feifei Wang
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Yanqui Li
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Mohammadjavad Paydar
- Institute for Research in Immunology and Cancer (IRIC), Département de médecine, Université de Montréal, Montréal H3C 3J7, Canada
| | - Tadayoshi Bessho
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Benjamin H. Kwok
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE 68583, USA
| | - Aimin Peng
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| |
Collapse
|
6
|
Vaziri C, Forker K, Zhang X, Wu D, Zhou P, Bowser JL. Pathological modulation of genome maintenance by cancer/testes antigens (CTAs). DNA Repair (Amst) 2025; 147:103818. [PMID: 39983270 PMCID: PMC11923853 DOI: 10.1016/j.dnarep.2025.103818] [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: 12/22/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025]
Abstract
The Cancer Testis Antigens (CTAs) are a group of germ cell proteins that are absent from normal somatic cells yet aberrantly expressed in many cancer cells. When mis-expressed in cancer cells, many CTAs promote tumorigenic characteristics including genome instability, DNA damage tolerance and therapy resistance. Here we highlight some of the CTAs for which their roles in genome maintenance in cancer cells are well established. We consider three broad CTA categories: (1) Melanoma Antigens (MAGEs) (2) Mitotic CTAs and (3) CTAs with roles in meiotic homologous recombination. Many cancer cells rely on CTAs to tolerate intrinsic and therapy-induced genotoxic stress. Therefore, CTAs represent molecular vulnerabilities of cancer cells and may provide opportunities for therapy. Owing to their high-level expression in tumors and absence from normal somatic cells, CTA-directed therapies could have a high level of specificity and would likely be devoid of side-effect toxicity.
Collapse
Affiliation(s)
- Cyrus Vaziri
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| | - Karly Forker
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Xingyuan Zhang
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, USA
| | - Di Wu
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA; Department of Biostatistics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Pei Zhou
- Department of Biochemistry, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jessica L Bowser
- Department of Pathology and Laboratory Medicine, University of North Carolina, Chapel Hill, NC 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
7
|
Kreis NN, Moon HH, Wordeman L, Louwen F, Solbach C, Yuan J, Ritter A. KIF2C/MCAK a prognostic biomarker and its oncogenic potential in malignant progression, and prognosis of cancer patients: a systematic review and meta-analysis as biomarker. Crit Rev Clin Lab Sci 2024; 61:404-434. [PMID: 38344808 PMCID: PMC11815995 DOI: 10.1080/10408363.2024.2309933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/05/2023] [Accepted: 01/22/2024] [Indexed: 03/24/2024]
Abstract
KIF2C/MCAK (KIF2C) is the most well-characterized member of the kinesin-13 family, which is critical in the regulation of microtubule (MT) dynamics during mitosis, as well as interphase. This systematic review briefly describes the important structural elements of KIF2C, its regulation by multiple molecular mechanisms, and its broad cellular functions. Furthermore, it systematically summarizes its oncogenic potential in malignant progression and performs a meta-analysis of its prognostic value in cancer patients. KIF2C was shown to be involved in multiple crucial cellular processes including cell migration and invasion, DNA repair, senescence induction and immune modulation, which are all known to be critical during the development of malignant tumors. Indeed, an increasing number of publications indicate that KIF2C is aberrantly expressed in multiple cancer entities. Consequently, we have highlighted its involvement in at least five hallmarks of cancer, namely: genome instability, resisting cell death, activating invasion and metastasis, avoiding immune destruction and cellular senescence. This was followed by a systematic search of KIF2C/MCAK's expression in various malignant tumor entities and its correlation with clinicopathologic features. Available data were pooled into multiple weighted meta-analyses for the correlation between KIF2Chigh protein or gene expression and the overall survival in breast cancer, non-small cell lung cancer and hepatocellular carcinoma patients. Furthermore, high expression of KIF2C was correlated to disease-free survival of hepatocellular carcinoma. All meta-analyses showed poor prognosis for cancer patients with KIF2Chigh expression, associated with a decreased overall survival and reduced disease-free survival, indicating KIF2C's oncogenic potential in malignant progression and as a prognostic marker. This work delineated the promising research perspective of KIF2C with modern in vivo and in vitro technologies to further decipher the function of KIF2C in malignant tumor development and progression. This might help to establish KIF2C as a biomarker for the diagnosis or evaluation of at least three cancer entities.
Collapse
Affiliation(s)
- Nina-Naomi Kreis
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Ha Hyung Moon
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Linda Wordeman
- Department of Physiology and Biophysics, University of Washington School of Medicine, Seattle, WA, USA
| | - Frank Louwen
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Christine Solbach
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Juping Yuan
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| | - Andreas Ritter
- Obstetrics and Prenatal Medicine, Gynaecology and Obstetrics, University Hospital Frankfurt, J. W. Goethe-University, Frankfurt, Germany
| |
Collapse
|
8
|
Huang J, Ma K, Zhang J, Zhou J, Yi J, Qi W, Liao M. Pathogenicity and transmission of novel highly pathogenic H7N2 variants originating from H7N9 avian influenza viruses in chickens. Virology 2024; 597:110121. [PMID: 38917688 DOI: 10.1016/j.virol.2024.110121] [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: 03/06/2024] [Revised: 05/01/2024] [Accepted: 05/21/2024] [Indexed: 06/27/2024]
Abstract
The H7 subtype avian influenza viruses are circulating widely worldwide, causing significant economic losses to the poultry industry and posing a serious threat to human health. In 2019, H7N2 and H7N9 co-circulated in Chinese poultry, yet the risk of H7N2 remained unclear. We isolated and sequenced four H7N2 viruses from chickens, revealing them as novel reassortants with H7N9-derived HA, M, NS genes and H9N2-derived PB2, PB1, PA,NP, NA genes. To further explore the key segment of pathogenicity, H7N2-H7N9NA and H7N2-H9N2HA single-substitution were constructed. Pathogenicity study showed H7N2 isolates to be highly pathogenic in chickens, with H7N2-H7N9NA slightly weaker than H7N2-Wild type. Transcriptomic analysis suggested that H7N9-derived HA genes primarily drove the high pathogenicity of H7N2 isolates, eliciting a strong inflammatory response. These findings underscored the increased threat posed by reassorted H7N2 viruses to chickens, emphasizing the necessity of long-term monitoring of H7 subtype avian influenza viruses.
Collapse
Affiliation(s)
- Jinyu Huang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; National Avian Influenza Para-Reference Laboratory, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China
| | - Kaixiong Ma
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; National Avian Influenza Para-Reference Laboratory, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Jiahao Zhang
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; National Avian Influenza Para-Reference Laboratory, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China
| | - Jiangtao Zhou
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; National Avian Influenza Para-Reference Laboratory, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China
| | - Jiahui Yi
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; National Avian Influenza Para-Reference Laboratory, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China
| | - Wenbao Qi
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; National Avian Influenza Para-Reference Laboratory, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China; Key Laboratory of Zoonoses Prevention and Control of Guangdong Province, Guangzhou, 510642, China.
| | - Ming Liao
- State Key Laboratory for Animal Disease Control and Prevention, South China Agricultural University, Guangzhou, 510642, China; National Avian Influenza Para-Reference Laboratory, Guangzhou, 510642, China; Key Laboratory of Zoonoses, Ministry of Agriculture and Rural Affairs, Guangzhou, 510642, China; National and Regional Joint Engineering Laboratory for Medicament of Zoonoses Prevention and Control, Guangzhou, 510642, China; College of Animal Science and Technology, Zhongkai University of Agriculture and Engineering, Guangzhou, China.
| |
Collapse
|
9
|
Gudivada IP, Amajala KC. Integrative Bioinformatics Analysis for Targeting Hub Genes in Hepatocellular Carcinoma Treatment. Curr Genomics 2024; 26:48-80. [PMID: 39911278 PMCID: PMC11793067 DOI: 10.2174/0113892029308243240709073945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/24/2024] [Accepted: 06/11/2024] [Indexed: 02/07/2025] Open
Abstract
Background The damage in the liver and hepatocytes is where the primary liver cancer begins, and this is referred to as Hepatocellular Carcinoma (HCC). One of the best methods for detecting changes in gene expression of hepatocellular carcinoma is through bioinformatics approaches. Objective This study aimed to identify potential drug target(s) hubs mediating HCC progression using computational approaches through gene expression and protein-protein interaction datasets. Methodology Four datasets related to HCC were acquired from the GEO database, and Differentially Expressed Genes (DEGs) were identified. Using Evenn, the common genes were chosen. Using the Fun Rich tool, functional associations among the genes were identified. Further, protein-protein interaction networks were predicted using STRING, and hub genes were identified using Cytoscape. The selected hub genes were subjected to GEPIA and Shiny GO analysis for survival analysis and functional enrichment studies for the identified hub genes. The up-regulating genes were further studied for immunohistopathological studies using HPA to identify gene/protein expression in normal vs HCC conditions. Drug Bank and Drug Gene Interaction Database were employed to find the reported drug status and targets. Finally, STITCH was performed to identify the functional association between the drugs and the identified hub genes. Results The GEO2R analysis for the considered datasets identified 735 upregulating and 284 downregulating DEGs. Functional gene associations were identified through the Fun Rich tool. Further, PPIN network analysis was performed using STRING. A comparative study was carried out between the experimental evidence and the other seven data evidence in STRING, revealing that most proteins in the network were involved in protein-protein interactions. Further, through Cytoscape plugins, the ranking of the genes was analyzed, and densely connected regions were identified, resulting in the selection of the top 20 hub genes involved in HCC pathogenesis. The identified hub genes were: KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Further, GEPIA and Shiny GO analyses provided insights into survival ratios and functional enrichment studied for the hub genes. The HPA database studies further found that upregulating genes were involved in changes in protein expression in Normal vs HCC tissues. These findings indicated that hub genes were certainly involved in the progression of HCC. STITCH database studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, could be used as leads to identify novel drugs, and identified hub genes could also be considered as potential and promising drug targets as they are involved in the gene-chemical interaction networks. Conclusion The present study involved various integrated bioinformatics approaches, analyzing gene expression and protein-protein interaction datasets, resulting in the identification of 20 top-ranked hubs involved in the progression of HCC. They are KIF2C, CDK1, TPX2, CEP55, MELK, TTK, BUB1, NCAPG, ASPM, KIF11, CCNA2, HMMR, BUB1B, TOP2A, CENPF, KIF20A, NUSAP1, DLGAP5, PBK, and CCNB2. Gene-chemical interaction network studies uncovered that existing drug molecules, including sorafenib, regorafenib, cabozantinib, and lenvatinib, can be used as leads to identify novel drugs, and the identified hub genes can be promising drug targets. The current study underscores the significance of targeting these hub genes and utilizing existing molecules to generate new molecules to combat liver cancer effectively and can be further explored in terms of drug discovery research to develop treatments for HCC.
Collapse
Affiliation(s)
- Indu Priya Gudivada
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| | - Krishna Chaitanya Amajala
- Department of Biochemistry and Bioinformatics, GITAM School of Science, GITAM (Deemed to be University), Visakhapatnam, 530045, Andhra Pradesh, India
| |
Collapse
|
10
|
Li RQ, Yang Y, Qiao L, Yang L, Shen DD, Zhao XJ. KIF2C: An important factor involved in signaling pathways, immune infiltration, and DNA damage repair in tumorigenesis. Biomed Pharmacother 2024; 171:116173. [PMID: 38237349 DOI: 10.1016/j.biopha.2024.116173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 01/02/2024] [Accepted: 01/13/2024] [Indexed: 02/08/2024] Open
Abstract
BACKGROUNDS Poorly regulated mitosis and chromosomal instability are common characteristics in malignant tumor cells. Kinesin family member 2 C (KIF2C), also known as mitotic centromere-associated kinesin (MCAK) is an essential component during mitotic regulation. In recent years, KIF2C was shown to be dysregulated in several tumors and was involved in many aspects of tumor self-regulation. Research on KIF2C may be a new direction and target for anti-tumor therapy. OBJECT The article aims at reviewing current literatures and summarizing the research status of KIF2C in malignant tumors as well as the oncogenic signaling pathways associated with KIF2C and its role in immune infiltration. RESULT In this review, we summarize the KIF2C mechanisms and signaling pathways in different malignant tumors, and briefly describe its involvement in pathways related to classical chemotherapeutic drug resistance, such as MEK/ERK, mTOR, Wnt/β-catenin, P53 and TGF-β1/Smad pathways. KIF2C upregulation was shown to promote tumor cell migration, invasion, chemotherapy resistance and inhibit DNA damage repair. It was also highly correlated with microRNAs, and CD4 +T cell and CD8 +T cell tumor immune infiltration. CONCLUSION This review shows that KIF2C may function as a new anticancer drug target with great potential for malignant tumor treatment and the mitigation of chemotherapy resistance.
Collapse
Affiliation(s)
- Rui-Qing Li
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yan Yang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, China
| | - Lin Qiao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Li Yang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Zhengzhou Key Laboratory of Endometrial Disease Prevention and Treatment, Zhengzhou, China.
| | - Dan-Dan Shen
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Jing Zhao
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Simon MN, Dubrana K, Palancade B. On the edge: how nuclear pore complexes rule genome stability. Curr Opin Genet Dev 2024; 84:102150. [PMID: 38215626 DOI: 10.1016/j.gde.2023.102150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 11/10/2023] [Accepted: 12/18/2023] [Indexed: 01/14/2024]
Abstract
Nuclear organization has emerged as a critical layer in the coordination of DNA repair activities. Distinct types of DNA lesions have notably been shown to relocate at the vicinity of nuclear pore complexes (NPCs), where specific repair pathways are favored, ultimately safeguarding genome integrity. Here, we review the most recent progress in this field, notably highlighting the increasingly diverse types of DNA structures undergoing repositioning, and the signaling pathways involved. We further discuss our growing knowledge of the molecular mechanisms underlying the choice of repair pathways at NPCs, and their conservation - or divergences. Intriguingly, a series of recent findings suggest that DNA metabolism may be coupled to NPC biogenesis and specialization, challenging our initial vision of these processes.
Collapse
Affiliation(s)
- Marie-Noëlle Simon
- Marseille Cancer Research Center (CRCM), U1068 Inserm, UMR7258 CNRS, Institut Paoli-Calmettes, Equipe Labélisée Ligue, Aix Marseille University, Marseille, France. https://twitter.com/@IJMonod
| | - Karine Dubrana
- Université Paris Cité, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, F-92260 Fontenay-aux-Roses, France; Université Paris-Saclay, Inserm, CEA, Stabilité Génétique Cellules Souches et Radiations, F-92260 Fontenay-aux-Roses, France. https://twitter.com/@DubranaLab
| | - Benoit Palancade
- Université Paris Cité, CNRS, Institut Jacques Monod, F-75013 Paris, France.
| |
Collapse
|
12
|
Tankrathok A, Mahong B, Roytrakul S, Daduang S, Temsiripong Y, Klaynongsruang S, Jangpromma N. Proteomic analysis of crocodile white blood cells reveals insights into the mechanism of the innate immune system. Heliyon 2024; 10:e24583. [PMID: 38312682 PMCID: PMC10835162 DOI: 10.1016/j.heliyon.2024.e24583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 02/06/2024] Open
Abstract
Crocodiles have a particularly powerful innate immune system because their blood contains high levels of antimicrobial peptides. They can survive injuries that would be fatal to other animals, and they are rarely afflicted with diseases. To better understand the crocodile's innate immune response, proteomic analysis was performed on the white blood cells (WBC) of an Aeromonas hydrophila-infected crocodile. Levels of WBC and red blood cells (RBC) rapidly increased within 1 h. In WBC, there were 109 up-regulated differentially expressed proteins (DEP) that were up-regulated. Fifty-nine DEPs dramatically increased expression from 1 h after inoculation, whereas 50 up-regulated DEPs rose after 24 h. The most abundant DEPs mainly had two biological functions, 1) gene expression regulators, for example, zinc finger proteins and histone H1 family, and 2) cell mechanical forces such as actin cytoskeleton proteins and microtubule-binding proteins. This finding illustrates the characteristic effective innate immune response mechanism of crocodiles that might occur via boosted transcription machinery proteins to accelerate cytoskeletal protein production for induction of phagocytosis, along with the increment of trafficking proteins to transport essential molecules for combating pathogens. The findings of this study provide new insights into the mechanisms of the crocodile's innate immune system.
Collapse
Affiliation(s)
- Anupong Tankrathok
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Biotechnology, Faculty of Agricultural Technology, Kalasin University, Kalasin, 46000, Thailand
| | - Bancha Mahong
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Sittiruk Roytrakul
- Proteomics Laboratory, Genome Technology Research Unit, National Center for Genetic Engineering and Biotechnology, Pathumthani, 12120, Thailand
| | - Sakda Daduang
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
- Division of Pharmacognosy and Toxicology, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen, 40002, Thailand
| | | | - Sompong Klaynongsruang
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
| | - Nisachon Jangpromma
- Protein and Proteomics Research Center for Commercial and Industrial Purposes (ProCCI), Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
- Department of Biochemistry, Faculty of Science, Khon Kaen University, Khon Kaen, 40002, Thailand
| |
Collapse
|
13
|
Deng H, Gong X, Ji G, Li C, Cheng S. KIF2C promotes clear cell renal cell carcinoma progression via activating JAK2/STAT3 signaling pathway. Mol Cell Probes 2023; 72:101938. [PMID: 37863123 DOI: 10.1016/j.mcp.2023.101938] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/14/2023] [Accepted: 10/16/2023] [Indexed: 10/22/2023]
Abstract
BACKGROUND Clear cell renal cell carcinoma (ccRCC) is one of the most common malignant tumors that can be highly aggressive. Despite advances in the exploration of its underlying molecular biology, the clinical outcome for advanced ccRCC is still unsatisfied. Recently, more attention was paid to the functions of Kinesin family member 2C (KIF2C) in cancer progression, while the specific function of KIF2C in ccRCC has not been sufficiently elucidated. The present study aims to investigate the role of KIF2C in the progression of ccRCC and reveal potential mechanisms. METHODS Expression of KIF2C in ccRCC tissues and adjacent normal tissue was compared and the association of KIF2C expression level with tumor grade, stage, and metastasis were analyzed using online web tool. Kaplan-Meier survival was performed to detect the association of KIF2C expression and patient' prognosis. Stably cell lines with KIF2C knockdown or overexpression were constructed by lentivirus infection. CCK-8, colony formation, scratch healing, and transwell invasion assays were carried out to explore the effect of KIF2C knockdown or overexpression on the proliferation, migration, and invasion of ccRCC cells. Gene set enrichment analysis (GSEA) was conducted to reveal signaling pathways associated with KIF2C expression. The effect of KIF2C on JAK2/STAT3 signaling pathway were explored by western blot assay. RESULTS KIF2C expression was significantly upregulated in ccRCC tissues and was higher with the increase of tumor grade, stage, and metastasis. Higher expression of KIF2C was correlated with worse overall survival and diseases free survival in ccRCC patients. Silence of KIF2C inhibited proliferation, migration, and invasion in ccRCC cells. Conversely, overexpression of KIF2C had the opposite effect. GSEA results showed that JAK/STAT signaling pathway was markedly enriched in KIF2Chigh group. Pearson' correlation revealed that KIF2C expression was significantly associated with genes in JAK2/STAT3 signaling. Western blot results showed that KIF2C knockdown decreased protein expression of p-JAK2 and p-STAT3, and KIF2C overexpression increased the phosphorylation of JAK2 and STAT3. AG490, a JAK2/STAT3 signaling inhibitor, could partly impair the tumor-promoting effects of KIF2C in ccRCC. CONCLUSION KIF2C expression was significantly upregulated in ccRCC and correlated with tumor grade, stage, metastasis, and patients' prognosis. KIF2C promoted ccRCC progression via activating JAK2/STAT3 signaling pathway, and KIF2C might be a novel target in ccRCC therapy.
Collapse
Affiliation(s)
- Hao Deng
- Department of Urology, The First People's Hospital of Jingzhou, Jingzhou, 434000, PR China
| | - Xiaobo Gong
- Department of Urology, The First People's Hospital of Jingzhou, Jingzhou, 434000, PR China
| | - Guanghai Ji
- Department of Urology, The First People's Hospital of Jingzhou, Jingzhou, 434000, PR China
| | - Chenglong Li
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, PR China.
| | - Shaoping Cheng
- Department of Urology, The First People's Hospital of Jingzhou, Jingzhou, 434000, PR China.
| |
Collapse
|
14
|
Dey G, Bharti R, Braley C, Alluri R, Esakov E, Crean-Tate K, McCrae K, Joehlin-Price A, Rose PG, Lathia J, Gong Z, Reizes O. LCK facilitates DNA damage repair by stabilizing RAD51 and BRCA1 in the nucleus of chemoresistant ovarian cancer. J Ovarian Res 2023; 16:122. [PMID: 37370140 PMCID: PMC10294509 DOI: 10.1186/s13048-023-01194-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 05/24/2023] [Indexed: 06/29/2023] Open
Abstract
Poly-ADP Ribose Polymerase (PARP) targeted therapy is clinically approved for the treatment of homologous recombination (HR) repair deficient tumors. The remarkable success of this therapy in the treatment of HR repair deficient cancers has not translated to HR-proficient cancers. Our studies identify the novel role of non-receptor lymphocyte-specific protein tyrosine kinase (LCK) in the regulation of HR repair in endometrioid epithelial ovarian cancer (eEOC) model. We show that DNA damage leads to direct interaction of LCK with the HR repair proteins RAD51 and BRCA1 in a kinase dependent manner RAD51 and BRCA1 stabilization. LCK expression is induced and activated in the nucleus in response to DNA damage insult. Disruption of LCK expression attenuates RAD51, BRCA1, and BRCA2 protein expression by hampering there stability and results in inhibition of HR-mediated DNA repair including suppression of RAD51 foci formation, and augmentation of γH2AX foci formation. In contrast LCK overexpression leads to increased RAD51 and BRCA1 expression with a concomitant increase in HR DNA damage repair. Importantly, attenuation of LCK sensitizes HR-proficient eEOC cells to PARP inhibitor in cells and pre-clinical mouse studies. Collectively, our findings identify a novel therapeutic strategy to expand the utility of PARP targeted therapy in HR proficient ovarian cancer.
Collapse
Affiliation(s)
- Goutam Dey
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Rashmi Bharti
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Chad Braley
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Ravi Alluri
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Emily Esakov
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
| | - Katie Crean-Tate
- Division of Gynecologic Cancer, Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Keith McCrae
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | | | - Peter G Rose
- Division of Gynecologic Cancer, Women's Health Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Justin Lathia
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Zihua Gong
- Case Comprehensive Cancer Center, Cleveland, OH, USA
- Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Ofer Reizes
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
- Case Comprehensive Cancer Center, Cleveland, OH, USA.
| |
Collapse
|
15
|
Smith JC, Husted S, Pilrose J, Ems-McClung SC, Stout JR, Carpenter RL, Walczak CE. MCAK Inhibitors Induce Aneuploidy in Triple-Negative Breast Cancer Models. Cancers (Basel) 2023; 15:3309. [PMID: 37444419 PMCID: PMC10340532 DOI: 10.3390/cancers15133309] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 06/16/2023] [Accepted: 06/19/2023] [Indexed: 07/15/2023] Open
Abstract
Standard of care for triple-negative breast cancer (TNBC) involves the use of microtubule poisons such as paclitaxel, which are proposed to work by inducing lethal levels of aneuploidy in tumor cells. While these drugs are initially effective in treating cancer, dose-limiting peripheral neuropathies are common. Unfortunately, patients often relapse with drug-resistant tumors. Identifying agents against targets that limit aneuploidy may be a valuable approach for therapeutic development. One potential target is the microtubule depolymerizing kinesin, MCAK, which limits aneuploidy by regulating microtubule dynamics during mitosis. Using publicly available datasets, we found that MCAK is upregulated in triple-negative breast cancer and is associated with poorer prognoses. Knockdown of MCAK in tumor-derived cell lines caused a two- to five-fold reduction in the IC50 for paclitaxel, without affecting normal cells. Using FRET and image-based assays, we screened compounds from the ChemBridge 50 k library and discovered three putative MCAK inhibitors. These compounds reproduced the aneuploidy-inducing phenotype of MCAK loss, reduced clonogenic survival of TNBC cells regardless of taxane-resistance, and the most potent of the three, C4, sensitized TNBC cells to paclitaxel. Collectively, our work shows promise that MCAK may serve as both a biomarker of prognosis and as a therapeutic target.
Collapse
Affiliation(s)
- John C. Smith
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| | - Stefan Husted
- LabCorp Drug Development Indianapolis, Indianapolis, IN 46214, USA;
| | - Jay Pilrose
- Catalent Pharma Solutions Bloomington, Bloomington, IN 47403, USA;
| | - Stephanie C. Ems-McClung
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| | - Jane R. Stout
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| | - Richard L. Carpenter
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| | - Claire E. Walczak
- Medical Sciences, Indiana School of Medicine—Bloomington, Bloomington, IN 47405, USA; (J.C.S.); (S.C.E.-M.); (J.R.S.); (R.L.C.)
| |
Collapse
|
16
|
Smith JC, Husted S, Pilrose J, Ems-McClung SC, Stout JR, Carpenter RL, Walczak CE. MCAK Inhibitors Induce Aneuploidy in Triple Negative Breast Cancer Models. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.31.543118. [PMID: 37397990 PMCID: PMC10312595 DOI: 10.1101/2023.05.31.543118] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Standard of care for triple negative breast cancer (TNBC) involves the use of microtubule poisons like paclitaxel, which are proposed to work by inducing lethal levels of aneuploidy in tumor cells. While these drugs are initially effective in treating cancer, dose-limiting peripheral neuropathies are common. Unfortunately, patients often relapse with drug resistant tumors. Identifying agents against targets that limit aneuploidy may be a valuable approach for therapeutic development. One potential target is the microtubule depolymerizing kinesin, MCAK, which limits aneuploidy by regulating microtubule dynamics during mitosis. Using publicly available datasets, we found that MCAK is upregulated in triple negative breast cancer and is associated with poorer prognoses. Knockdown of MCAK in tumor-derived cell lines caused a two- to five-fold reduction in the IC 50 for paclitaxel, without affecting normal cells. Using FRET and image-based assays, we screened compounds from the ChemBridge 50k library and discovered three putative MCAK inhibitors. These compounds reproduced the aneuploidy-inducing phenotype of MCAK loss, reduced clonogenic survival of TNBC cells regardless of taxane-resistance, and the most potent of the three, C4, sensitized TNBC cells to paclitaxel. Collectively, our work shows promise that MCAK may serve as both a biomarker of prognosis and as a therapeutic target. Simple Summary Triple negative breast cancer (TNBC) is the most lethal breast cancer subtype with few treatment options available. Standard of care for TNBC involves the use of taxanes, which are initially effective, but dose limiting toxicities are common, and patients often relapse with resistant tumors. Specific drugs that produce taxane-like effects may be able to improve patient quality of life and prognosis. In this study we identify three novel inhibitors of the Kinesin-13 MCAK. MCAK inhibition induces aneuploidy; similar to cells treated with taxanes. We demonstrate that MCAK is upregulated in TNBC and is associated with poorer prognoses. These MCAK inhibitors reduce the clonogenic survival of TNBC cells, and the most potent of the three inhibitors, C4, sensitizes TNBC cells to taxanes, similar to the effects of MCAK knockdown. This work will expand the field of precision medicine to include aneuploidy-inducing drugs that have the potential to improve patient outcomes.
Collapse
|
17
|
Yang JH, Brandão HB, Hansen AS. DNA double-strand break end synapsis by DNA loop extrusion. Nat Commun 2023; 14:1913. [PMID: 37024496 PMCID: PMC10079674 DOI: 10.1038/s41467-023-37583-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/23/2023] [Indexed: 04/08/2023] Open
Abstract
DNA double-strand breaks (DSBs) occur every cell cycle and must be efficiently repaired. Non-homologous end joining (NHEJ) is the dominant pathway for DSB repair in G1-phase. The first step of NHEJ is to bring the two DSB ends back into proximity (synapsis). Although synapsis is generally assumed to occur through passive diffusion, we show that passive diffusion is unlikely to produce the synapsis speed observed in cells. Instead, we hypothesize that DNA loop extrusion facilitates synapsis. By combining experimentally constrained simulations and theory, we show that a simple loop extrusion model constrained by previous live-cell imaging data only modestly accelerates synapsis. Instead, an expanded loop extrusion model with targeted loading of loop extruding factors (LEFs), a small portion of long-lived LEFs, and LEF stabilization by boundary elements and DSB ends achieves fast synapsis with near 100% efficiency. We propose that loop extrusion contributes to DSB repair by mediating fast synapsis.
Collapse
Affiliation(s)
- Jin H Yang
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA
- The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
- Koch Institute for Integrative Cancer Research, Cambridge, MA, 02142, USA
| | - Hugo B Brandão
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Koch Institute for Integrative Cancer Research, Cambridge, MA, 02142, USA.
- Illumina Inc., San Diego, CA, 92122, USA.
| | - Anders S Hansen
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, 02139, USA.
- The Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
- Koch Institute for Integrative Cancer Research, Cambridge, MA, 02142, USA.
| |
Collapse
|
18
|
Liu S, Ye Z, Xue VW, Sun Q, Li H, Lu D. KIF2C is a prognostic biomarker associated with immune cell infiltration in breast cancer. BMC Cancer 2023; 23:307. [PMID: 37016301 PMCID: PMC10071625 DOI: 10.1186/s12885-023-10788-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Accepted: 03/29/2023] [Indexed: 04/06/2023] Open
Abstract
BACKGROUND The kinesin-13 family member 2C (KIF2C) is a versatile protein participating in many biological processes. KIF2C is frequently up-regulated in multiple types of cancer and is associated with cancer development. However, the role of KIF2C in immune cell infiltration of tumor microenvironment and immunotherapy in breast cancer remains unclear. METHODS The expression of KIF2C was analyzed using Tumor Immune Estimation Resource (TIMER) database and further verified by immunohistochemical staining in human breast cancer tissues. The correlation between KIF2C expression and clinical parameters, the impact of KIF2C on clinical prognosis and independent prognostic factors were analyzed by using TCGA database, the Kaplan-Meier plotter, and Univariate and multivariate Cox analyses, respectively. The nomograms were constructed according to independent prognostic factors and validated with C-index, calibration curves, ROC curves, and decision curve analysis. A gene set enrichment analysis (GSEA) was performed to explore the underlying molecular mechanisms of KIF2C. The degree of immune infiltration was assessed by the Estimation of Stromal and Immune cells in Malignant Tumor tissues using the Expression (ESTIMATE) algorithm and the single sample GSEA (ssGSEA). The Tumor mutational burden and Tumor Immune Dysfunction and Rejection (TIDE) were used to analyze immunotherapeutic efficiency. Finally, the KIF2C-related competing endogenous RNA (ceRNA) network was constructed to predict the putative regulatory mechanisms of KIF2C. RESULTS KIF2C was remarkably up-regulated in 18 different types of cancers, including breast cancer. Kaplan-Meier survival analysis showed that high KIF2C expression was associated with poor overall survival (OS). KIF2C expression was associated with clinical parameters such as age, TMN stage, T status, and molecular subtypes. We identified age, stage, estrogen receptor (ER) and KIF2C expression as OS-related independent prognosis factors for breast cancer. An OS-related nomogram was developed based on these independent prognosis factors and displayed good predicting ability for OS of breast cancer patients. Finally, our results revealed that KIF2C was significantly related to immune cell infiltration, tumor mutational burden, and immunotherapy in patients with breast cancer. CONCLUSION KIF2C was overexpressed in breast cancer and was positively correlated with immune cell infiltration and immunotherapy response. Therefore, KIF2C can serve as a potential biomarker for prognosis and immunotherapy in breast cancer.
Collapse
Affiliation(s)
- Shanshan Liu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology, Carson International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong, 518055, China
| | - Ziwei Ye
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology, Carson International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong, 518055, China
| | - Vivian Weiwen Xue
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology, Carson International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong, 518055, China
| | - Qi Sun
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology, Carson International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong, 518055, China
| | - Huan Li
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology, Carson International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong, 518055, China
| | - Desheng Lu
- Guangdong Provincial Key Laboratory of Regional Immunity and Diseases, Department of Pharmacology, Carson International Cancer Center, Shenzhen University Medical School, Shenzhen, Guangdong, 518055, China.
| |
Collapse
|
19
|
Zhang J, An L, Zhao R, Shi R, Zhou X, Wei S, Zhang Q, Zhang T, Feng D, Yu Z, Wang H. KIF4A promotes genomic stability and progression of endometrial cancer through regulation of TPX2 protein degradation. Mol Carcinog 2023; 62:303-318. [PMID: 36468837 DOI: 10.1002/mc.23487] [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: 07/20/2022] [Revised: 10/07/2022] [Accepted: 11/04/2022] [Indexed: 12/12/2022]
Abstract
Kinesin family member 4A (KIF4A) belongs to the kinesin superfamily proteins, which are closely associated with mitophagy. Nonetheless, the role of KIF4A in endometrial cancer (EC) remains poorly characterized. The present study showed that KIF4A not only was upregulated but also predicted poor prognosis in patients with EC. KIF4A knockdown in EC cells resulted in attenuated proliferative capacity in vitro and in vivo. Transcriptome sequencing and gene function analysis revealed that KIF4A contributed to the maintenance of EC cells' genomic stability and that KIF4A knockdown induced the DNA damage response, cell cycle arrest, and apoptosis. Mechanistically, KIF4A interacted with TPX2 (a protein involved in DNA damage repair to cope with the replication pressure) to enhance its stability via inhibition of TPX2 ubiquitination and eventually ensured the genomic stability of EC cells during mitosis. Taken together, our results indicated that KIF4A functions as a tumor oncogene that facilitates EC progression via the maintenance of genomic stability. Therefore, targeting the KIF4A/TPX2 axis may provide new concepts and strategies for the treatment of patients with EC.
Collapse
Affiliation(s)
- Jun Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lanfen An
- Division of Life Science and Medicine, Clinical Center of Reproductive Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, China
| | - Rong Zhao
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Rui Shi
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xing Zhou
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Sitian Wei
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Qi Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Tangansu Zhang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Dilu Feng
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhicheng Yu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Hongbo Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.,Clinical Research Center of Cancer Immunotherapy, Wuhan, Hubei, China
| |
Collapse
|
20
|
Huang X, Zhao F, Wu Q, Wang Z, Ren H, Zhang Q, Wang Z, Xu J. KIF2C Facilitates Tumor Growth and Metastasis in Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2023; 15:1502. [PMID: 36900292 PMCID: PMC10000478 DOI: 10.3390/cancers15051502] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 02/16/2023] [Accepted: 02/19/2023] [Indexed: 03/06/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a highly lethal cancer with a poor prognosis. For PDAC, an increase in the survival time of patients and a reduction mortality have not yet successfully been achieved. In many research works, Kinesin family member 2C (KIF2C) is highly expressed in several tumors. Nevertheless, the role of KIF2C in pancreatic cancer is unknown. In this study, we found that KIF2C expression is significantly upregulated in human PDAC tissues and cell lines such as ASPC-1 and MIA-PaCa2. Moreover, KIF2C upregulation is associated with a poor prognosis when combining the expression of KIF2C with clinical information. Through cell functional assays and the construction of animal models, we showed that KIF2C promotes PDAC cell proliferation, migration, invasion, and metastasis, both in vitro and in vivo. Finally, the results of sequencing showed that the overexpression of KIF2C causes a decrease in some proinflammatory factors and chemokines. The cell cycle detection indicated that the pancreatic cancer cells in the overexpressed group had abnormal proliferation in the G2 and S phases. These results revealed the potential of KIF2C as a therapeutic target for the treatment of PDAC.
Collapse
Affiliation(s)
- Xing Huang
- Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Feng Zhao
- Department of Stem Cells and Regenerative Medicine, China Medical University, Shenyang 110122, China
| | - Quan Wu
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Zitong Wang
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Haiyue Ren
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Qiqi Zhang
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Zhe Wang
- Department of Pathology, Shengjing Hospital, China Medical University, Shenyang 110004, China
| | - Jin Xu
- Department of General Surgery, Shengjing Hospital, China Medical University, Shenyang 110004, China
| |
Collapse
|
21
|
Dullovi A, Ozgencil M, Rajvee V, Tse WY, Cutillas PR, Martin SA, Hořejší Z. Microtubule-associated proteins MAP7 and MAP7D1 promote DNA double-strand break repair in the G1 cell cycle phase. iScience 2023; 26:106107. [PMID: 36852271 PMCID: PMC9958362 DOI: 10.1016/j.isci.2023.106107] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 12/12/2022] [Accepted: 01/27/2023] [Indexed: 02/04/2023] Open
Abstract
The DNA-damage response is a complex signaling network that guards genomic integrity. The microtubule cytoskeleton is involved in the repair of DNA double-strand breaks; however, little is known about which cytoskeleton-related proteins are involved in DNA repair and how. Using quantitative proteomics, we discovered that microtubule associated proteins MAP7 and MAP7D1 interact with several DNA repair proteins including DNA double-strand break repair proteins RAD50, BRCA1 and 53BP1. We observed that downregulation of MAP7 and MAP7D1 leads to increased phosphorylation of p53 after γ-irradiation. Moreover, we determined that the downregulation of MAP7D1 leads to a strong G1 arrest and that the downregulation of MAP7 and MAP7D1 in G1 arrested cells negatively affects DNA repair, recruitment of RAD50 to chromatin and localization of 53BP1 to the sites of damage. These findings describe for the first time a novel function of MAP7 and MAP7D1 in cell cycle regulation and repair of DNA double-strand breaks.
Collapse
Affiliation(s)
- Arlinda Dullovi
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Meryem Ozgencil
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Vinothini Rajvee
- Mass Spectrometry Laboratory, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Wai Yiu Tse
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Pedro R. Cutillas
- Centre for Genomics & Computational Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Sarah A. Martin
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK
| | - Zuzana Hořejší
- Centre for Cancer Cell & Molecular Biology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London EC1M 6BQ, UK,Corresponding author
| |
Collapse
|
22
|
Zhang B, Liu P, Li Y, Hu Q, Li H, Pang X, Wu H. Multi-omics analysis of kinesin family member 2C in human tumors: novel prognostic biomarker and tumor microenvironment regulator. Am J Cancer Res 2022; 12:4954-4976. [PMID: 36504885 PMCID: PMC9729912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 10/26/2022] [Indexed: 12/15/2022] Open
Abstract
Kinesin family member 2C (KIF2C) is the best-characterized member of the kinesin-13 family and is involved in accurately fine-tuned dynamics of mitotic spindles. As KIF2C is involved in both spindle formation and regulation of DNA double-strand breaks, precise regulation of KIF2C is essential to prevent malignant transformation associated with gains and losses of DNA content. In the present study, we initially reviewed The Cancer Genome Atlas database and observed that KIF2C is abundantly expressed in most tumor types. We then analyzed the gene alteration profile, protein expression, prognosis, and immune reactivities of KIF2C in more than 10,000 samples from several well-established databases. In addition, we conducted a gene enrichment set analysis to investigate the potential mechanisms underlying the role of KIF2C in tumorigenesis. Multi-omics analysis of KIF2C demonstrated significant statistical correlations between KIF2C expression and clinical prognosis, oncogenic signature gene sets, myeloid-derived suppressor cell infiltration, ImmunoScore, immune checkpoints, microsatellite instability, and tumor mutational burden across multiple tumors. Single-cell data showed that KIF2C is abundantly expressed in malignant cells. The experimental validation demonstrated that KIF2C is highly expressed in gastric cancer cell lines, gastric adenocarcinoma, and hepatocelluar carcinoma. The findings of this study provide important insight for understanding the role and mechanisms of KIF2C in tumorigenesis and immunotherapy in a variety of cancers.
Collapse
Affiliation(s)
- Bixi Zhang
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal UniversityChangsha, Hunan, China
| | - Peng Liu
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Yanchun Li
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal UniversityChangsha, Hunan, China
| | - Qing Hu
- Department of Pathology, Hunan Provincial People’s Hospital, Hunan Normal UniversityChangsha, Hunan, China
| | - Huan Li
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Xiaoyang Pang
- Department of Orthopaedics, Xiangya Hospital, Central South UniversityChangsha, Hunan, China
| | - Hao Wu
- Department of Gastroenterology, Third Xiangya Hospital, Central South UniversityChangsha, Hunan, China
- Center for Precision Medicine, University of Missouri School of MedicineColumbia, MO, USA
| |
Collapse
|
23
|
García Fernández F, Almayrac E, Carré Simon À, Batrin R, Khalil Y, Boissac M, Fabre E. Global chromatin mobility induced by a DSB is dictated by chromosomal conformation and defines the HR outcome. eLife 2022; 11:78015. [PMID: 36125964 PMCID: PMC9489209 DOI: 10.7554/elife.78015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 09/08/2022] [Indexed: 12/16/2022] Open
Abstract
Repair of DNA double-strand breaks (DSBs) is crucial for genome integrity. A conserved response to DSBs is an increase in chromatin mobility that can be local, at the site of the DSB, or global, at undamaged regions of the genome. Here, we address the function of global chromatin mobility during homologous recombination (HR) of a single, targeted, controlled DSB. We set up a system that tracks HR in vivo over time and show that two types of DSB-induced global chromatin mobility are involved in HR, depending on the position of the DSB. Close to the centromere, a DSB induces global mobility that depends solely on H2A(X) phosphorylation and accelerates repair kinetics, but is not essential. In contrast, the global mobility induced by a DSB away from the centromere becomes essential for HR repair and is triggered by homology search through a mechanism that depends on H2A(X) phosphorylation, checkpoint progression, and Rad51. Our data demonstrate that global mobility is governed by chromosomal conformation and differentially coordinates repair by HR.
Collapse
Affiliation(s)
| | - Etienne Almayrac
- Université de Paris, IRSL, INSERM, U944, CNRS, UMR7212, Paris, France
| | - Ànnia Carré Simon
- Université de Paris, IRSL, INSERM, U944, CNRS, UMR7212, Paris, France
| | - Renaud Batrin
- Université de Paris, IRSL, INSERM, U944, CNRS, UMR7212, Paris, France
| | - Yasmine Khalil
- Université de Paris, IRSL, INSERM, U944, CNRS, UMR7212, Paris, France
| | - Michel Boissac
- Université de Paris, IRSL, INSERM, U944, CNRS, UMR7212, Paris, France
| | - Emmanuelle Fabre
- Université de Paris, IRSL, INSERM, U944, CNRS, UMR7212, Paris, France
| |
Collapse
|
24
|
Kim JM. Molecular Link between DNA Damage Response and Microtubule Dynamics. Int J Mol Sci 2022; 23:ijms23136986. [PMID: 35805981 PMCID: PMC9266319 DOI: 10.3390/ijms23136986] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 11/16/2022] Open
Abstract
Microtubules are major components of the cytoskeleton that play important roles in cellular processes such as intracellular transport and cell division. In recent years, it has become evident that microtubule networks play a role in genome maintenance during interphase. In this review, we highlight recent advances in understanding the role of microtubule dynamics in DNA damage response and repair. We first describe how DNA damage checkpoints regulate microtubule organization and stability. We then highlight how microtubule networks are involved in the nuclear remodeling following DNA damage, which leads to changes in chromosome organization. Lastly, we discuss how microtubule dynamics participate in the mobility of damaged DNA and promote consequent DNA repair. Together, the literature indicates the importance of microtubule dynamics in genome organization and stability during interphase.
Collapse
Affiliation(s)
- Jung Min Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 58128, Korea
| |
Collapse
|
25
|
Zhang P, Gao H, Ye C, Yan R, Yu L, Xia C, Yang D. Large-Scale Transcriptome Data Analysis Identifies KIF2C as a Potential Therapeutic Target Associated With Immune Infiltration in Prostate Cancer. Front Immunol 2022; 13:905259. [PMID: 35720323 PMCID: PMC9203693 DOI: 10.3389/fimmu.2022.905259] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 05/06/2022] [Indexed: 12/24/2022] Open
Abstract
Prostate cancer (PCa) is one of the most prevalent cancers of the urinary system. In previous research, Kinesin family member 2C (KIF2C), as an oncogene, has been demonstrated to have a key role in the incidence and progression of different cancers. However, KIF2C has not been reported in PCa. We combined data from different databases, including The Cancer Genome Atlas, the Cancer Cell Line Encyclopedia, Genotype Tissue-Expression, cBioPortal, and the Genomics of Drug Sensitivity in Cancer database, to explore the potential oncogenic role of KIF2C in PCa through a series of bioinformatics approaches, including analysis of the association between KIF2C and prognosis, clinicopathological features, gene mutations, DNA methylation, immune cell infiltration, and drug resistance. The results showed that KIF2C was significantly up-regulated in PCa. High KIF2C expression was associated with age, pathological stage, lymph node metastases, prostate-specific antigen (PSA), and Gleason score and significantly predicted an unfavorable prognosis in PCa patients. Results from Gene Set Enrichment Analysis (GSEA) suggested that KIF2C was involved in the cell cycle and immune response. KIF2C DNA methylation was reduced in PCa and was inversely linked with KIF2C expression. KIF2C was shown to have a strong relationship with the tumor microenvironment (TME), infiltrating cells, and immune checkpoint genes. Furthermore, high KIF2C expression was significantly resistant to a variety of MAPK signaling pathway-related inhibitors. Our study reveals that KIF2C may be a possible predictive biomarker for assessing prognosis in PCa patients with immune infiltration.
Collapse
Affiliation(s)
- Pingxin Zhang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hang Gao
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chunwei Ye
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Ruping Yan
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Lu Yu
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Chengxing Xia
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Delin Yang
- Department of Urology, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
26
|
Ren K, Wang L, Wang Y, An G, Du Q, Cao J, Jin Q, Yun K, Guo Z, Wang Y, Liang Q, Sun J. Wound age estimation based on next-generation sequencing: Fitting the optimal index system using machine learning. Forensic Sci Int Genet 2022; 59:102722. [DOI: 10.1016/j.fsigen.2022.102722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 04/30/2022] [Accepted: 05/10/2022] [Indexed: 11/04/2022]
|
27
|
In Silico Analysis of the Correlation of KIF2C with Prognosis and Immune Infiltration in Glioma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6320828. [PMID: 35387222 PMCID: PMC8977321 DOI: 10.1155/2022/6320828] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 01/03/2022] [Accepted: 01/20/2022] [Indexed: 11/17/2022]
Abstract
Glioblastoma (GBM) is one of the most commonly pivotal malignant caners. Numerous reports have revealed the crucial roles of immune infiltration in the initiation and progression of GBM. In this study, we first identified differentially expressed genes (DEGs) in the progression of GBM using CGGA databases. Totally, 156 upregulated DEGs and 251 downregulated DEGs were revealed. By constructing a protein-protein interaction network, KIF2C was identified as a hub gene in GBM. Further analysis revealed an evidently positive association existing in KIF2C expression and the advanced stages of gliomas. Higher expression of KIF2C was in WHO grade IV samples relative to that in grade III and grade II samples. In addition, our results showed that KIF2C was higher in IDH1 wild-type samples than IDH1 mutant glioma samples, in 1p/19q noncodel samples than 1p/19q code glioma samples, and in recurrent samples than primary glioma samples. Moreover, our results showed that higher expression of KIF2C correlated with shorter survival time in both primary and recurrent gliomas and could act as a potential biomarker for the prognosis of GBM. Further analysis demonstrated that higher expression of KIF2C was related to higher levels of endothelial cell, T cell CD8+ naïve, common lymphoid progenitor, T cell CD4+ Th2, T cell CD4+ Th2, macrophage, macrophage M1, T cell CD4+ memory, and T cell CD4+ effector memory, but was related to lower levels of NK cell, B cell plasma, T cell CD4+ Th1, T cell regulatory (Tregs), neutrophil, and T cell NK. We thought this study could provide potential biomarkers for the prediction of prognosis and immune infiltration of gliomas.
Collapse
|
28
|
Yuan Y, Chen Z, Cai X, He S, Li D, Zhao W. Identification of Hub Genes Correlated With Poor Prognosis for Patients With Uterine Corpus Endometrial Carcinoma by Integrated Bioinformatics Analysis and Experimental Validation. Front Oncol 2021; 11:766947. [PMID: 34868993 PMCID: PMC8639584 DOI: 10.3389/fonc.2021.766947] [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: 08/30/2021] [Accepted: 10/29/2021] [Indexed: 12/13/2022] Open
Abstract
Uterine Corpus Endometrial Carcinoma (UCEC) is one of the most common malignancies of the female genital tract and there remains a major public health problem. Although significant progress has been made in explaining the progression of UCEC, it is still warranted that molecular mechanisms underlying the tumorigenesis of UCEC are to be elucidated. The aim of the current study was to investigate key modules and hub genes related to UCEC pathogenesis, and to explore potential biomarkers and therapeutic targets for UCEC. The RNA-seq dataset and corresponding clinical information for UCEC patients were obtained from the Cancer Genome Atlas (TCGA) database. Differentially expressed genes (DEGs) were screened between 23 paired UCEC tissues and adjacent non-cancerous tissues. Subsequently, the co-expression network of DEGs was determined via weighted gene co-expression network analysis (WGCNA). The Blue and Brown modules were identified to be significantly positively associated with neoplasm histologic grade. The highly connected genes of the two modules were then investigated as potential key factors related to tumor differentiation. Additionally, a protein-protein interaction (PPI) network for all genes in the two modules was constructed to obtain key modules and nodes. 10 genes were identified by both WGCNA and PPI analyses, and it was shown by Kaplan-Meier curve analysis that 6 out of the 10 genes were significantly negatively related to the 5-year overall survival (OS) in patients (AURKA, BUB1, CDCA8, DLGAP5, KIF2C, TPX2). Besides, according to the DEGs from the two modules, lncRNA-miRNA-mRNA and lncRNA-TF-mRNA networks were constructed to explore the molecular mechanism of UCEC-related lncRNAs. 3 lncRNAs were identified as being significantly negatively related to the 5-year OS (AC015849.16, DUXAP8 and DGCR5), with higher expression in UCEC tissues compared to non-tumor tissues. Finally, quantitative Real-time PCR was applied to validate the expression patterns of hub genes. Cell proliferation and colony formation assays, as well as cell cycle distribution and apoptosis analysis, were performed to test the effects of representative hub genes. Altogether, this study not only promotes our understanding of the molecular mechanisms for the pathogenesis of UCEC but also identifies several promising biomarkers in UCEC development, providing potential therapeutic targets for UCEC.
Collapse
Affiliation(s)
- Yi Yuan
- Department of Laboratory Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhengzheng Chen
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xushan Cai
- Department of Clinical Laboratory, Maternal and Child Health Hospital of Jiading District, Shanghai, China.,School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shengxiang He
- School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Dong Li
- Department of Laboratory Medicine, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Weidong Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
29
|
Starvation-induced proteasome assemblies in the nucleus link amino acid supply to apoptosis. Nat Commun 2021; 12:6984. [PMID: 34848715 PMCID: PMC8633328 DOI: 10.1038/s41467-021-27306-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 11/09/2021] [Indexed: 11/15/2022] Open
Abstract
Eukaryotic cells have evolved highly orchestrated protein catabolic machineries responsible for the timely and selective disposal of proteins and organelles, thereby ensuring amino acid recycling. However, how protein degradation is coordinated with amino acid supply and protein synthesis has remained largely elusive. Here we show that the mammalian proteasome undergoes liquid-liquid phase separation in the nucleus upon amino acid deprivation. We termed these proteasome condensates SIPAN (Starvation-Induced Proteasome Assemblies in the Nucleus) and show that these are a common response of mammalian cells to amino acid deprivation. SIPAN undergo fusion events, rapidly exchange proteasome particles with the surrounding milieu and quickly dissolve following amino acid replenishment. We further show that: (i) SIPAN contain K48-conjugated ubiquitin, (ii) proteasome inhibition accelerates SIPAN formation, (iii) deubiquitinase inhibition prevents SIPAN resolution and (iv) RAD23B proteasome shuttling factor is required for SIPAN formation. Finally, SIPAN formation is associated with decreased cell survival and p53-mediated apoptosis, which might contribute to tissue fitness in diverse pathophysiological conditions.
Collapse
|
30
|
Mast Cells Resting-Related Prognostic Signature in Hepatocellular Carcinoma. JOURNAL OF ONCOLOGY 2021; 2021:4614257. [PMID: 34840569 PMCID: PMC8616659 DOI: 10.1155/2021/4614257] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/03/2021] [Accepted: 11/05/2021] [Indexed: 01/24/2023]
Abstract
The immune microenvironment of liver cancer is of great significance for the treatment of liver cancer. After evaluating the content of mast cells resting in the transcriptome data of The Cancer Genome Atlas database by CIBERSORT analysis, this study aimed to group the samples according to the content of mast cells resting in different samples to find the differentially expressed genes in the two groups. Significant prognostic differences were found between high and low mast cells resting infiltration groups. The prognostic model was constructed according to the differentially expressed genes. The model was validated using external independent datasets. The results revealed that the constructed model was reliable. It could well distinguish the prognostic differences of patients in different characteristic groups. The high-risk group was mainly concentrated in metabolic pathways. The risk score of this model was closely related to some immune cells, immune function, and immune checkpoints. Therefore, this model may provide new ideas for immunotherapy of hepatocellular carcinoma.
Collapse
|
31
|
Li Y, Kardell MB, Wang F, Wang L, Zhu S, Bessho T, Peng A. The Sm core components of small nuclear ribonucleoproteins promote homologous recombination repair. DNA Repair (Amst) 2021; 108:103244. [PMID: 34768043 DOI: 10.1016/j.dnarep.2021.103244] [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: 04/19/2021] [Revised: 10/01/2021] [Accepted: 10/29/2021] [Indexed: 11/17/2022]
Abstract
DNA Double strand breaks (DSBs) are highly hazardous to the cell, and are repaired predominantly via non-homologous end joining (NHEJ) and homologous recombination (HR). Using DSB-mimicking DNA templates, our proteomic studies identified a group of Sm core proteins of small nuclear ribonucleoproteins (snRNPs) as potential DSB-associated proteins. We further confirmed that these Sm proteins were recruited to laser-induced DNA damage sites, and co-localized with established DNA damage repair factors. Depletion of Sm-D3 or Sm-B induced accumulation of γ-H2AX, and impaired the repair efficiency of HR, but not NHEJ. Furthermore, disruption of Sm-D3 reduced the protein level of HR factors, especially RAD51 and CHK1, but caused no change in the expression of repair factors involved in NHEJ. Mechanistically, Sm-D3 proteins bound RAD51, suppressed the ubiquitination of RAD51, and mediated the stabilization of RAD51; Sm-D3 depletion particularly impacted the level of RAD51 and CHK1 on damaged chromatin. As such, our studies characterized a role of Sm proteins in HR repair, via a new mechanism that is distinct from their conventional functions in RNA processing and gene regulation, but consistent with their direct recruitment to DNA damage sites and association with repair factors.
Collapse
Affiliation(s)
- Yanqiu Li
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Mary Bridget Kardell
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Feifei Wang
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Ling Wang
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Songli Zhu
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA
| | - Tadayoshi Bessho
- The Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, USA
| | - Aimin Peng
- Department of Oral Biology, University of Nebraska Medical Center, Lincoln, NE, USA.
| |
Collapse
|
32
|
Lin Q, Qi Q, Hou S, Chen Z, Jiang N, Zhang L, Lin C. Activation of the TGF-β1/Smad signaling by KIF2C contributes to the malignant phenotype of thyroid carcinoma cells. Tissue Cell 2021; 73:101655. [PMID: 34624565 DOI: 10.1016/j.tice.2021.101655] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 09/09/2021] [Accepted: 09/23/2021] [Indexed: 12/21/2022]
Abstract
Kinesin family member 2C (KIF2C) has been identified as a potential oncogene in various types of human cancers; however, the role of KIF2C in thyroid cancer has not yet been elucidated. Quantitative real-time polymerase chain reaction and western blotting were employed for gene expression analysis. Cell Counting Kit-8 and ethynyl-2'-deoxyuridine assays were performed to examine cell proliferation. Cell migration and invasion were assessed by wound-healing and transwell invasion assays. Results showed that KIF2C expression was upregulated in thyroid carcinoma cell lines. In addition, upregulation of KIF2C promoted the proliferation, migration, and invasion of thyroid carcinoma cells, while downregulation of KIF2C exerted the opposite effects. Overexpression of KIF2C induced the activation of transforming growth factor-β1 (TGF-β1)/Smad signaling in thyroid carcinoma cells. However, inhibition of TGF-β1/Smad signaling through silencing TGF-β1 attenuated the promoting effects of KIF2C overexpression on the malignant phenotype of thyroid carcinoma cells. Besides, overexpression of TGF-β1 suppressed the inhibitory effect of KIF2C knockdown on the proliferation and metastasis of thyroid carcinoma cells. In conclusion, our findings demonstrated that KIF2C contributed to the malignant phenotype of thyroid carcinoma cells by inducing the activation of TGF-β1/Smad signaling, thus uncovering a novel mechanism for thyroid carcinoma progression.
Collapse
Affiliation(s)
- Qiuyu Lin
- Nuclear Medicine Department, The First Hospital of Jilin University, China
| | - Qianle Qi
- Nuclear Medicine Department, The First Hospital of Jilin University, China
| | - Sen Hou
- Nuclear Medicine Department, The First Hospital of Jilin University, China
| | - Zhen Chen
- Chengdu Xinke Pharmaceutical Co., LTD, China
| | - Nan Jiang
- Nuclear Medicine Department, The First Hospital of Jilin University, China
| | - Laney Zhang
- College of Biological Sciences, Cornell University, United States
| | - Chenghe Lin
- Nuclear Medicine Department, The First Hospital of Jilin University, China.
| |
Collapse
|
33
|
Schweiggert J, Habeck G, Hess S, Mikus F, Beloshistov R, Meese K, Hata S, Knobeloch K, Melchior F. SCF Fbxw5 targets kinesin-13 proteins to facilitate ciliogenesis. EMBO J 2021; 40:e107735. [PMID: 34368969 PMCID: PMC8441365 DOI: 10.15252/embj.2021107735] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 06/29/2021] [Accepted: 07/12/2021] [Indexed: 11/23/2022] Open
Abstract
Microtubule depolymerases of the kinesin-13 family play important roles in various cellular processes and are frequently overexpressed in different cancer types. Despite the importance of their correct abundance, remarkably little is known about how their levels are regulated in cells. Using comprehensive screening on protein microarrays, we identified 161 candidate substrates of the multi-subunit ubiquitin E3 ligase SCFFbxw5 , including the kinesin-13 member Kif2c/MCAK. In vitro reconstitution assays demonstrate that MCAK and its closely related orthologs Kif2a and Kif2b become efficiently polyubiquitylated by neddylated SCFFbxw5 and Cdc34, without requiring preceding modifications. In cells, SCFFbxw5 targets MCAK for proteasomal degradation predominantly during G2 . While this seems largely dispensable for mitotic progression, loss of Fbxw5 leads to increased MCAK levels at basal bodies and impairs ciliogenesis in the following G1 /G0 , which can be rescued by concomitant knockdown of MCAK, Kif2a or Kif2b. We thus propose a novel regulatory event of ciliogenesis that begins already within the G2 phase of the preceding cell cycle.
Collapse
Affiliation(s)
- Jörg Schweiggert
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Gregor Habeck
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Sandra Hess
- Institute of NeuropathologyFaculty of MedicineUniversity of FreiburgFreiburgGermany
- Faculty of BiologyUniversity of FreiburgFreiburgGermany
| | - Felix Mikus
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Roman Beloshistov
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Klaus Meese
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | - Shoji Hata
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| | | | - Frauke Melchior
- Zentrum für Molekulare Biologie der Universität Heidelberg (ZMBH)University of HeidelbergDKFZ ‐ ZMBH AllianceHeidelbergGermany
| |
Collapse
|
34
|
The nuclear kinesin KIF18B promotes 53BP1-mediated DNA double-strand break repair. Cell Rep 2021; 35:109306. [PMID: 34192545 DOI: 10.1016/j.celrep.2021.109306] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 04/29/2021] [Accepted: 06/03/2021] [Indexed: 12/13/2022] Open
Abstract
53BP1 is recruited to chromatin in the vicinity of DNA double-strand breaks (DSBs). We identify the nuclear kinesin, KIF18B, as a 53BP1-interacting protein and define its role in 53BP1-mediated DSB repair. KIF18B is a molecular motor protein involved in destabilizing astral microtubules during mitosis. It is primarily nuclear throughout the interphase and is constitutively chromatin bound. Our observations indicate a nuclear function during the interphase for a kinesin previously implicated in mitosis. We identify a central motif in KIF18B, which we term the Tudor-interacting motif (TIM), because of its interaction with the Tudor domain of 53BP1. TIM enhances the interaction between the 53BP1 Tudor domain and dimethylated lysine 20 of histone H4. TIM and the motor function of KIF18B are both required for efficient 53BP1 focal recruitment in response to damage and for fusion of dysfunctional telomeres. Our data suggest a role for KIF18B in efficient 53BP1-mediated end-joining of DSBs.
Collapse
|
35
|
Shokrollahi M, Mekhail K. Interphase microtubules in nuclear organization and genome maintenance. Trends Cell Biol 2021; 31:721-731. [PMID: 33902985 DOI: 10.1016/j.tcb.2021.03.014] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 03/10/2021] [Accepted: 03/23/2021] [Indexed: 12/11/2022]
Abstract
Microtubules are major cytoskeletal components mediating fundamental cellular processes, including cell division. Recent evidence suggests that microtubules also regulate the nucleus during the cell cycle's interphase stage. Deciphering such roles of microtubules should uncover direct crosstalk between the nucleus and cytoplasm, impacting genome function and organismal health. Here, we review emerging roles for microtubules in interphase genome regulation. We explore how microtubules exert cytoplasmic forces on the nucleus or transport molecular cargo, including DNA, into or within the nucleus. We also describe how microtubules perform these functions by establishing transient or stable connections with nuclear envelope elements. Lastly, we discuss how the regulation of the nucleus by microtubules impacts genome organization and repair. Together, the literature indicates that interphase microtubules are critical regulators of nuclear structure and genome stability.
Collapse
Affiliation(s)
- Mitra Shokrollahi
- Department of Laboratory Medicine and Pathobiology, MaRS Centre, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Karim Mekhail
- Department of Laboratory Medicine and Pathobiology, MaRS Centre, Faculty of Medicine, University of Toronto, Toronto, ON, Canada; Canada Research Chairs Program, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
36
|
Cobb AM, Yusoff S, Hayward R, Ahmad S, Sun M, Verhulst A, D'Haese PC, Shanahan CM. Runx2 (Runt-Related Transcription Factor 2) Links the DNA Damage Response to Osteogenic Reprogramming and Apoptosis of Vascular Smooth Muscle Cells. Arterioscler Thromb Vasc Biol 2021; 41:1339-1357. [PMID: 33356386 DOI: 10.1161/atvbaha.120.315206] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Accepted: 12/08/2020] [Indexed: 01/08/2023]
Abstract
[Figure: see text].
Collapse
MESH Headings
- Animals
- Apoptosis
- Cells, Cultured
- Cellular Reprogramming
- Core Binding Factor Alpha 1 Subunit/genetics
- Core Binding Factor Alpha 1 Subunit/metabolism
- DNA Damage
- Disease Models, Animal
- Female
- Histones/metabolism
- Humans
- Male
- Mice, Inbred C57BL
- Mice, Knockout
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Osteogenesis
- Phosphorylation
- Rats, Wistar
- Signal Transduction
- Vascular Calcification/genetics
- Vascular Calcification/metabolism
- Vascular Calcification/pathology
- Mice
- Rats
Collapse
Affiliation(s)
- Andrew M Cobb
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, The James Black Centre, United Kingdom (A.M.C., S.Y., R.H., S.A., M.S., C.M.S.)
| | - Syabira Yusoff
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, The James Black Centre, United Kingdom (A.M.C., S.Y., R.H., S.A., M.S., C.M.S.)
| | - Robert Hayward
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, The James Black Centre, United Kingdom (A.M.C., S.Y., R.H., S.A., M.S., C.M.S.)
| | - Sadia Ahmad
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, The James Black Centre, United Kingdom (A.M.C., S.Y., R.H., S.A., M.S., C.M.S.)
| | - Mengxi Sun
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, The James Black Centre, United Kingdom (A.M.C., S.Y., R.H., S.A., M.S., C.M.S.)
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium (A.V., P.C.D.)
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Wilrijk, Belgium (A.V., P.C.D.)
| | - Catherine M Shanahan
- BHF Centre of Research Excellence, School of Cardiovascular Medicine and Sciences, King's College London, The James Black Centre, United Kingdom (A.M.C., S.Y., R.H., S.A., M.S., C.M.S.)
| |
Collapse
|
37
|
Zhou P, Wang J, Mishail D, Wang CY. Recent advancements in PARP inhibitors-based targeted cancer therapy. PRECISION CLINICAL MEDICINE 2020; 3:187-201. [PMID: 32983586 PMCID: PMC7501589 DOI: 10.1093/pcmedi/pbaa030] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 08/27/2020] [Accepted: 08/27/2020] [Indexed: 12/11/2022] Open
Abstract
Poly(ADP-ribose) polymerase inhibitors (PARPi) are a new class of agents with unparalleled clinical achievement for driving synthetic lethality in BRCA-deficient cancers. Recent FDA approval of PARPi has motivated clinical trials centered around the optimization of PARPi-associated therapies in a variety of BRCA-deficient cancers. This review highlights recent advancements in understanding the molecular mechanisms of PARP ‘trapping’ and synthetic lethality. Particular attention is placed on the potential extension of PARPi therapies from BRCA-deficient patients to populations with other homologous recombination-deficient backgrounds, and common characteristics of PARPi and non-homologous end-joining have been elucidated. The synergistic antitumor effect of combining PARPi with various immune checkpoint blockades has been explored to evaluate the potential of combination therapy in attaining greater therapeutic outcome. This has shed light onto the differing classifications of PARPi as well as the factors that result in altered PARPi activity. Lastly, acquired chemoresistance is a crucial issue for clinical application of PARPi. The molecular mechanisms underlying PARPi resistance and potential overcoming strategies are discussed.
Collapse
Affiliation(s)
- Ping Zhou
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, CA 90095, USA
| | - Justin Wang
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, CA 90095, USA
| | - Daniel Mishail
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, CA 90095, USA
| | - Cun-Yu Wang
- Laboratory of Molecular Signaling, Division of Oral Biology and Medicine, School of Dentistry, UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
38
|
Abstract
The presence of actin in the nucleus has historically been a highly contentious issue. It is now, however, well accepted that actin has physiologically important roles in the nucleus. In this Review, we describe the evolution of our thinking about actin in the nucleus starting with evidence supporting its involvement in transcription, chromatin remodeling and intranuclear movements. We also review the growing literature on the mechanisms that regulate the import and export of actin and how post-translational modifications of actin could regulate nuclear actin. We end with an extended discussion of the role of nuclear actin in the repair of DNA double stranded breaks.
Collapse
Affiliation(s)
- Leonid Serebryannyy
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, United States
| | - Primal de Lanerolle
- Department of Physiology and Biophysics, University of Illinois at Chicago, Chicago, IL 60612, United States.
| |
Collapse
|
39
|
Oh CK, Kang JW, Lee Y, Myung K, Ha M, Kang J, Kwon EJ, Kim Y, Oh SO, Heo HJ, Kim S, Kim YH. Role of kif2c, A Gene Related to ALL Relapse, in Embryonic Hematopoiesis in Zebrafish. Int J Mol Sci 2020; 21:3127. [PMID: 32354205 PMCID: PMC7246619 DOI: 10.3390/ijms21093127] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/17/2020] [Accepted: 04/27/2020] [Indexed: 12/18/2022] Open
Abstract
Relapse of acute lymphoblastic leukemia (ALL) is dangerous and it worsens the prognosis of patients; however, prognostic markers or therapeutic targets for ALL remain unknown. In the present study, using databases such as TARGET, GSE60926 and GSE28460, we determined that KIF2C and its binding partner, KIF18B are overexpressed in patients with relapsed ALL compared to that in patients diagnosed with ALL for the first time. As 50% of the residues are exactly the same and the signature domain of KIF2C is highly conserved between human and zebrafish, we used zebrafish embryos as a model to investigate the function of kif2c in vivo. We determined that kif2c is necessary for lymphopoiesis in zebrafish embryos. Additionally, we observed that kif2c is not related to differentiation of HSCs; however, it is important for the maintenance of HSCs as it provides survival signals to HSCs. These results imply that the ALL relapse-related gene KIF2C is linked to the survival of HSCs. In conclusion, we suggest that KIF2C can serve as a novel therapeutic target for relapsed ALL.
Collapse
Affiliation(s)
- Chang-Kyu Oh
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea; (C.-K.O.); (Y.L.); (K.M.)
| | - Ji Wan Kang
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Yoonsung Lee
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea; (C.-K.O.); (Y.L.); (K.M.)
| | - Kyungjae Myung
- Center for Genomic Integrity, Institute for Basic Science (IBS), Ulsan 44919, Korea; (C.-K.O.); (Y.L.); (K.M.)
| | - Mihyang Ha
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Junho Kang
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Eun Jung Kwon
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Youngjoo Kim
- Interdisciplinary Program of Genomic Science, Pusan National University, Yangsan 50612, Korea; (J.W.K.); (M.H.); (J.K.); (E.J.K.); (Y.K.)
| | - Sae-Ock Oh
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Korea; (S.-O.O.); (H.J.H.)
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Korea; (S.-O.O.); (H.J.H.)
| | - Shin Kim
- Department of Immunology, School of Medicine, Keimyung University, Dalseo-gu, Daegu 42601, Korea
- Institute of Medical Science, Keimyung University, Dalseo-gu, Daegu 42601, Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Korea; (S.-O.O.); (H.J.H.)
- Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan 50612, Korea
| |
Collapse
|