1
|
Li Q, Sun M, Meng Y, Feng M, Wang M, Chang C, Dong H, Bu F, Xu C, Liu J, Ling Q, Qiao Y, Chen J. Kinesin family member 18B activates mTORC1 signaling via actin gamma 1 to promote the recurrence of human hepatocellular carcinoma. Oncogenesis 2023; 12:54. [PMID: 37957153 PMCID: PMC10643429 DOI: 10.1038/s41389-023-00499-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 10/08/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
The mechanistic target of rapamycin complex 1 (mTORC1) signaling pathway is frequently reported to be hyperactivated in hepatocellular carcinoma (HCC) and contributes to HCC recurrence. However, the underlying regulatory mechanisms of mTORC1 signaling in HCC are not fully understood. In the present study, we found that the expression of kinesin family member 18B (KIF18B) was positively correlated with mTORC1 signaling in HCC, and the upregulation of KIF18B and p-mTOR was associated with a poor prognosis and HCC recurrence. Utilizing in vitro and in vivo assays, we showed that KIF18B promoted HCC cell proliferation and migration through activating mTORC1 signaling. Mechanistically, we identified Actin gamma 1 (γ-Actin) as a binding partner of KIF18B. KIF18B and γ-Actin synergistically modulated lysosome positioning, promoted mTORC1 translocation to lysosome membrane, and prohibited p70 S6K from entering lysosomes for degradation, which finally led to the enhancement of mTORC1 signaling transduction. Moreover, we found that KIF18B was a direct target of Forkhead box M1, which explains the potential mechanism of KIF18B overexpression in HCC. Our study highlights the potential of KIF18B as a therapeutic target for the treatment of HCC.
Collapse
Affiliation(s)
- Qian Li
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Mengqing Sun
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Yao Meng
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Mengqing Feng
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Menglan Wang
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Cunjie Chang
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Heng Dong
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Fangtian Bu
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Chao Xu
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China
| | - Jing Liu
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China
| | - Qi Ling
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China.
| | - Yiting Qiao
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China.
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, NHC Key Laboratory of Combined Multi-organ Transplantation, Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, P. R. China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, 250000, P. R. China.
| | - Jianxiang Chen
- School of Pharmacy and Department of Hepatology, the Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou, 311121, P. R. China.
- Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, P. R. China.
- Laboratory of Cancer Genomics, Division of Cellular and Molecular Research, National Cancer Centre, Singapore, 169610, Singapore.
| |
Collapse
|
2
|
Turaga SM, Vishwakarma V, Hembruff SL, Gibbs BK, Sabu P, Puri RV, Pathak HB, Samuel G, Godwin AK. Inducing Mitotic Catastrophe as a Therapeutic Approach to Improve Outcomes in Ewing Sarcoma. Cancers (Basel) 2023; 15:4911. [PMID: 37894278 PMCID: PMC10605681 DOI: 10.3390/cancers15204911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/03/2023] [Accepted: 10/07/2023] [Indexed: 10/29/2023] Open
Abstract
Ewing sarcoma (EWS) is an aggressive pediatric malignancy of the bone and soft tissues in need of novel therapeutic options. To identify potential therapeutic targets, we focused on essential biological pathways that are upregulated by EWS-FLI1, the primary oncogenic driver of EWS, including mitotic proteins such as Aurora kinase A (AURKA) and kinesin family member 15 (KIF15) and its binding partner, targeting protein for Xklp2 (TPX2). KIF15/TPX2 cooperates with KIF11, a key mitotic kinesin essential for mitotic spindle orientation. Given the lack of clinical-grade KIF15/TPX2 inhibitors, we chose to target KIF11 (using SB-743921) in combination with AURKA (using VIC-1911) given that phosphorylation of KIF15S1169 by Aurora A is required for its targeting to the spindle. In vitro, the drug combination demonstrated strong synergy (Bliss score ≥ 10) at nanomolar doses. Colony formation assay revealed significant reduction in plating efficiency (1-3%) and increased percentage accumulation of cells in the G2/M phase with the combination treatment (45-52%) upon cell cycle analysis, indicating mitotic arrest. In vivo studies in EWS xenograft mouse models showed significant tumor reduction and overall effectiveness: drug combination vs. vehicle control (p ≤ 0.01), SB-743921 (p ≤ 0.01) and VIC-1911 (p ≤ 0.05). Kaplan-Meier curves demonstrated superior overall survival with the combination compared to vehicle or monotherapy arms (p ≤ 0.0001).
Collapse
Affiliation(s)
- Soumya M. Turaga
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (S.M.T.); (V.V.); (B.K.G.); (R.V.P.); (H.B.P.)
| | - Vikalp Vishwakarma
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (S.M.T.); (V.V.); (B.K.G.); (R.V.P.); (H.B.P.)
| | - Stacey L. Hembruff
- University of Kansas Cancer Center, Kansas City, KS 66160, USA; (S.L.H.); (P.S.)
| | - Benjamin K. Gibbs
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (S.M.T.); (V.V.); (B.K.G.); (R.V.P.); (H.B.P.)
| | - Priya Sabu
- University of Kansas Cancer Center, Kansas City, KS 66160, USA; (S.L.H.); (P.S.)
- Division of Gynecologic Oncology, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Rajni V. Puri
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (S.M.T.); (V.V.); (B.K.G.); (R.V.P.); (H.B.P.)
| | - Harsh B. Pathak
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (S.M.T.); (V.V.); (B.K.G.); (R.V.P.); (H.B.P.)
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
| | - Glenson Samuel
- Division of Pediatric Hematology Oncology and Bone Marrow Transplantation, Children’s Mercy Hospital, Kansas City, MO 64108, USA;
| | - Andrew K. Godwin
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA; (S.M.T.); (V.V.); (B.K.G.); (R.V.P.); (H.B.P.)
- University of Kansas Cancer Center, Kansas City, KS 66160, USA; (S.L.H.); (P.S.)
- Kansas Institute for Precision Medicine, University of Kansas Medical Center, Kansas City, KS 66160, USA
- Division of Genomic Diagnostics, Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, 3901 Rainbow Boulevard, MS 3040, Kansas City, KS 66160, USA
| |
Collapse
|
3
|
Shrestha S, Ems-McClung SC, Hazelbaker MA, Yount AL, Shaw SL, Walczak CE. Importin α/β promote Kif18B microtubule association and enhance microtubule destabilization activity. Mol Biol Cell 2023; 34:ar30. [PMID: 36790918 PMCID: PMC10092650 DOI: 10.1091/mbc.e22-03-0113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023] Open
Abstract
Tight regulation of microtubule (MT) dynamics is necessary for proper spindle assembly and chromosome segregation. The MT destabilizing Kinesin-8, Kif18B, controls astral MT dynamics and spindle positioning. Kif18B interacts with importin α/β as well as with the plus-tip tracking protein EB1, but how these associations modulate Kif18B is not known. We mapped the key binding sites on Kif18B, made residue-specific mutations, and assessed their impact on Kif18B function. Blocking EB1 interaction disrupted Kif18B MT plus-end accumulation and inhibited its ability to control MT length on monopolar spindles in cells. Blocking importin α/β interaction disrupted Kif18B localization without affecting aster size. In vitro, importin α/β increased Kif18B MT association by increasing the on-rate and decreasing the off-rate from MTs, which stimulated MT destabilization. In contrast, EB1 promoted MT destabilization without increasing lattice binding in vitro, which suggests that EB1 and importin α/β have distinct roles in the regulation of Kif18B-mediated MT destabilization. We propose that importin α/β spatially modulate Kif18B association with MTs to facilitate its MT destabilization activity. Our results suggest that Ran regulation is important not only to control molecular motor function near chromatin but also to provide a spatial control mechanism to modulate MT binding of nuclear localization signal-containing spindle assembly factors.
Collapse
Affiliation(s)
- Sanjay Shrestha
- Medical Sciences, Indiana School of Medicine-Bloomington, Bloomington, IN 47405
| | | | - Mark A Hazelbaker
- Medical Sciences, Indiana School of Medicine-Bloomington, Bloomington, IN 47405
| | - Amber L Yount
- Medical Sciences, Indiana School of Medicine-Bloomington, Bloomington, IN 47405
| | - Sidney L Shaw
- Department of Biology, Indiana University, Bloomington, IN 47405
| | - Claire E Walczak
- Medical Sciences, Indiana School of Medicine-Bloomington, Bloomington, IN 47405
| |
Collapse
|
4
|
McHugh T, Welburn JPI. Potent microtubule-depolymerizing activity of a mitotic Kif18b-MCAK-EB network. J Cell Sci 2023; 136:275263. [PMID: 35502670 DOI: 10.1242/jcs.260144] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 04/21/2022] [Indexed: 11/20/2022] Open
Abstract
The precise regulation of microtubule length during mitosis is essential to assemble and position the mitotic spindle and segregate chromosomes. The kinesin-13 Kif2C or MCAK acts as a potent microtubule depolymerase that diffuses short distances on microtubules, whereas the kinesin-8 Kif18b is a processive motor with weak depolymerase activity. However, the individual activities of these factors cannot explain the dramatic increase in microtubule dynamics in mitosis. Using in vitro reconstitution and single-molecule imaging, we demonstrate that Kif18b, MCAK and the plus-end tracking protein EB3 (also known as MAPRE3) act in an integrated manner to potently promote microtubule depolymerization at very low concentrations. We find that Kif18b can transport EB3 and MCAK and promotes their accumulation to microtubule plus ends through multivalent weak interactions. Together, our work defines the mechanistic basis for a cooperative Kif18b-MCAK-EB network at microtubule plus ends, that acts to efficiently shorten and regulate microtubules in mitosis, essential for correct chromosome segregation.
Collapse
Affiliation(s)
- Toni McHugh
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| | - Julie P I Welburn
- Wellcome Trust Centre for Cell Biology, School of Biological Sciences, University of Edinburgh, Edinburgh EH9 3BF, Scotland, UK
| |
Collapse
|
5
|
He Z, Wang J, Xu J, Jiang X, Liu X, Jiang J. Dynamic regulation of KIF15 phosphorylation and acetylation promotes focal adhesions disassembly in pancreatic cancer. Cell Death Dis 2022; 13:896. [PMID: 36280663 PMCID: PMC9592618 DOI: 10.1038/s41419-022-05338-y] [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: 04/23/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Pancreatic cancer (PC) is prone to distant metastasis in the early stage, which is attributed to the strong migration ability of tumor cells. Focal adhesion turnover is essential for cancer cell metastasis, and the integrin recycling process is a key activation pathway for focal adhesion depolymerization. To identify the key motor protein involving in the integrin β1 recycling, we screened kinesin proteins involved in integrin β1 recycling using a kinesin family siRNA library and identified kinesin family 15 (KIF15) as a key regulator. KIF15 was upregulated in metastasis PC tissues and promoted PC cell migration and invasion. We identified KIF15 as a key component mediating integrin β1/FAK signaling that accelerated FA disassembly in a FAK-Y397-dependent manner. KIF15 recruited PI3K-C2α to promote integrin β1/FAK signaling and FA disassembly in a RAB11A-dependent manner. The C-terminal tail of KIF15 is required for the PI3K-C2α interaction and RAB11A activation. In addition, we also found that SIRT1-mediated acetylation of KIF15 is essential for KIF15 phosphorylation, which is the key activation event in motor protein function. Together, these findings indicate that KIF15 interacts with PI3K-C2α to promote FA turnover in PC cells by controlling the endosome recycling of integrin β1 in a SIRT1 acetylation modification-dependent manner, eventually promoting focal adhesions turnover and distant metastasis in PC.
Collapse
Affiliation(s)
- Zhiwei He
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jie Wang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jian Xu
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xueyi Jiang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Xinyuan Liu
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| | - Jianxin Jiang
- grid.412632.00000 0004 1758 2270Department of Hepatic-Biliary Surgery, Renmin Hospital of Wuhan University, 430060 Wuhan, China
| |
Collapse
|
6
|
Schrock MS, Scarberry L, Stromberg BR, Sears C, Torres AE, Tallman D, Krupinski L, Chakravarti A, Summers MK. MKLP2 functions in early mitosis to ensure proper chromosome congression. J Cell Sci 2022; 135:275559. [PMID: 35638575 DOI: 10.1242/jcs.259560] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 05/23/2022] [Indexed: 11/20/2022] Open
Abstract
Mitotic kinesin-like protein 2 (MKLP2) is a motor protein with a well-established function in promoting cytokinesis. However, our results with siRNAs targeting MKLP2 and small molecule inhibitors of MKLP2 (MKLP2i) suggested a function earlier in mitosis, prior to anaphase. In this study we provide direct evidence that MKLP2 facilitates chromosome congression in prometaphase. We employed live imaging to observe HeLa cells with fluorescently tagged histones treated with MKLP2i and discovered a pronounced chromosome congression defect. We show that MKLP2 facilitates error correction as inhibited cells had a significant increase in unstable, syntelic kinetochore-microtubule attachments. We find that the aberrant attachments are accompanied by elevated Aurora Kinase (A/B) activity and phosphorylation of the downstream target, pHEC1 (Ser 55). Lastly, we show that MKLP2 inhibition results in aneuploidy, confirming that MKLP2 safeguards cells against chromosomal instability.
Collapse
Affiliation(s)
- Morgan S Schrock
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Luke Scarberry
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA.,Biomedical Sciences Graduate, Program The Ohio State University Columbus, OH, 43210, USA
| | - Benjamin R Stromberg
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA.,Biomedical Sciences Graduate, Program The Ohio State University Columbus, OH, 43210, USA
| | - Claire Sears
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA.,Undergraduate Studies, Kenyon College, Gambier, OH, 43022, USA
| | - Adrian E Torres
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - David Tallman
- Department of Internal Medicine, The Ohio State University, Columbus, OH, 43210, USA.,Molecular, Cellular, and Developmental Biology Graduate Program, The Ohio State University, Columbus, OH, 43210, USA
| | - Lucas Krupinski
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Arnab Chakravarti
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| | - Matthew K Summers
- Department of Radiation Oncology, Arthur G James Comprehensive Cancer Center and Richard L. Solove Research Institute, The Ohio State University, Columbus, OH, 43210, USA
| |
Collapse
|
7
|
Moreci RS, Lechler T. KIF18B is a cell type-specific regulator of spindle orientation in the epidermis. Mol Biol Cell 2021; 32:ar29. [PMID: 34432485 PMCID: PMC8693959 DOI: 10.1091/mbc.e21-06-0291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Proper spindle orientation is required for asymmetric cell division and the establishment of complex tissue architecture. In the developing epidermis, spindle orientation requires a conserved cortical protein complex of LGN/NuMA/dynein-dynactin. However, how microtubule dynamics are regulated to interact with this machinery and properly position the mitotic spindle is not fully understood. Furthermore, our understanding of the processes that link spindle orientation during asymmetric cell division to cell fate specification in distinct tissue contexts remains incomplete. We report a role for the microtubule catastrophe factor KIF18B in regulating microtubule dynamics to promote spindle orientation in keratinocytes. During mitosis, KIF18B accumulates at the cell cortex, colocalizing with the conserved spindle orientation machinery. In vivo we find that KIF18B is required for oriented cell divisions within the hair placode, the first stage of hair follicle morphogenesis, but is not essential in the interfollicular epidermis. Disrupting spindle orientation in the placode, using mutations in either KIF18B or NuMA, results in aberrant cell fate marker expression of hair follicle progenitor cells. These data functionally link spindle orientation to cell fate decisions during hair follicle morphogenesis. Taken together, our data demonstrate a role for regulated microtubule dynamics in spindle orientation in epidermal cells. This work also highlights the importance of spindle orientation during asymmetric cell division to dictate cell fate specification.
Collapse
Affiliation(s)
- Rebecca S Moreci
- Department of Dermatology and Department of Cell Biology, Duke University, Durham, NC 27710
| | - Terry Lechler
- Department of Dermatology and Department of Cell Biology, Duke University, Durham, NC 27710
| |
Collapse
|
8
|
Machado CB, DA Silva EL, Dias Nogueira BM, DA Silva JBS, DE Moraes Filho MO, Montenegro RC, DE Moraes MEA, Moreira-Nunes CA. The Relevance of Aurora Kinase Inhibition in Hematological Malignancies. CANCER DIAGNOSIS & PROGNOSIS 2021; 1:111-126. [PMID: 35399305 DOI: 10.21873/cdp.10016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 04/27/2021] [Indexed: 12/26/2022]
Abstract
Aurora kinases are a family of serine/threonine protein kinases that play a central role in eukaryotic cell division. Overexpression of aurora kinases in cancer and their role as major regulators of the cell cycle quickly inspired the idea that their inhibition might be a potential pathway when treating oncologic patients. Over the past couple of decades, the search for designing and testing of molecules capable of inhibiting aurora activities fueled many pre-clinical and clinical studies. In this study, data from the past 10 years of in vitro and in vivo investigations, as well as clinical trials, utilizing aurora kinase inhibitors as therapeutics for hematological malignancies were compiled and discussed, aiming to highlight potential uses of these inhibitors as a novel monotherapy model or alongside conventional chemotherapies. While there is still much to be elucidated, it is clear that these kinases play a key role in oncogenesis, and their manageable toxicity and potentially synergistic effects still render them a focus of interest for future investigations in combinatorial clinical trials.
Collapse
Affiliation(s)
- Caio Bezerra Machado
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Emerson Lucena DA Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Beatriz Maria Dias Nogueira
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Jean Breno Silveira DA Silva
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Manoel Odorico DE Moraes Filho
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | - Raquel Carvalho Montenegro
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| | | | - Caroline Aquino Moreira-Nunes
- Pharmacogenetics Laboratory, Drug Research and Development Center (NPDM),Federal University of Ceará, Fortaleza, CE, Brazil
| |
Collapse
|
9
|
Hansson K, Radke K, Aaltonen K, Saarela J, Mañas A, Sjölund J, Smith EM, Pietras K, Påhlman S, Wennerberg K, Gisselsson D, Bexell D. Therapeutic targeting of KSP in preclinical models of high-risk neuroblastoma. Sci Transl Med 2021; 12:12/562/eaba4434. [PMID: 32967973 DOI: 10.1126/scitranslmed.aba4434] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 05/11/2020] [Accepted: 07/30/2020] [Indexed: 01/05/2023]
Abstract
Neuroblastoma is a childhood malignancy with often dismal prognosis; relapse is common despite intense treatment. Here, we used human tumor organoids representing multiple MYCN-amplified high-risk neuroblastomas to perform a high-throughput drug screen with approved or emerging oncology drugs. Tumor-selective effects were calculated using drug sensitivity scores. Several drugs with previously unreported anti-neuroblastoma effects were identified by stringent selection criteria. ARRY-520, an inhibitor of kinesin spindle protein (KSP), was among those causing reduced viability. High expression of the KSP-encoding gene KIF11 was associated with poor outcome in neuroblastoma. Genome-scale loss-of-function screens in hundreds of human cancer cell lines across 22 tumor types revealed that KIF11 is particularly important for neuroblastoma cell viability. KSP inhibition in neuroblastoma patient-derived xenograft (PDX) cells resulted in the formation of abnormal monoastral spindles, mitotic arrest, up-regulation of mitosis-associated genes, and apoptosis. In vivo, KSP inhibition caused regression of MYCN-amplified neuroblastoma PDX tumors. Furthermore, treatment of mice harboring orthotopic neuroblastoma PDX tumors resulted in increased survival. Our results suggested that KSP inhibition could be a promising treatment strategy in children with high-risk neuroblastoma.
Collapse
Affiliation(s)
- Karin Hansson
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Katarzyna Radke
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Kristina Aaltonen
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Jani Saarela
- Institute for Molecular Medicine Finland, University of Helsinki, 00290 Helsinki, Finland
| | - Adriana Mañas
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Jonas Sjölund
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Emma M Smith
- Division of Molecular Medicine and Gene Therapy, Lund Strategic Center for Stem Cell Biology, Lund University, 221 84 Lund, Sweden
| | - Kristian Pietras
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Sven Påhlman
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden
| | - Krister Wennerberg
- Institute for Molecular Medicine Finland, University of Helsinki, 00290 Helsinki, Finland.,BRIC - Biotech Research and Innovation Centre, University of Copenhagen, 2200 Copenhagen, Denmark
| | - David Gisselsson
- Division of Clinical Genetics, Department of Laboratory Medicine, Lund University, 221 85 Lund, Sweden.,Department of Pathology, Laboratory Medicine, Medical Services, University Hospital, 221 84 Lund, Sweden
| | - Daniel Bexell
- Division of Translational Cancer Research, Department of Laboratory Medicine, Lund University, 223 81 Lund, Sweden.
| |
Collapse
|
10
|
Vanadocene dichloride induces apoptosis in HeLa cells through depolymerization of microtubules and inhibition of Eg5. J Biol Inorg Chem 2021; 26:511-531. [PMID: 34057639 DOI: 10.1007/s00775-021-01872-w] [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: 02/18/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022]
Abstract
Vanadocene dichloride (VDC), a vanadium containing metallocene dihalide exhibits promising anticancer activity. However, its mechanism of action remains elusive as several diverse targets and pathways have been proposed for its anticancer activity. In this study, we observed that VDC inhibited the proliferation of mammalian cancer cells and induced apoptotic cell death by altering the mitochondrial membrane potential and the expression of bcl2 and bax. Probing further into its anticancer mechanism, we found that VDC caused depolymerization of interphase microtubules and blocked the cells at mitosis with considerable proportion of cells exhibiting monopolar spindles. The reassembly of cold depolymerized microtubules was strongly inhibited in the presence of 10 μM VDC. VDC perturbed the microtubule-kinetochore interactions during mitosis as indicated by the absence of cold stable spindle microtubules in the cells treated with 20 μM VDC. Using goat brain tubulin, we found that VDC inhibited the steady-state polymer mass of microtubules and bound to tubulin at a novel site with a Kd of 9.71 ± 0.19 μM and perturbed the secondary structure of tubulin dimer. In addition, VDC was also found to bind to the mitotic kinesin Eg5 and inhibit its basal as well as microtubule stimulated ATPase activity. The results suggest that disruption of microtubule assembly dynamics and inhibition of the ATPase activity of Eg5 could be a plausible mechanism for the antiproliferative and antimitotic activity of VDC.Graphic abstract.
Collapse
|
11
|
Blengini CS, Ibrahimian P, Vaskovicova M, Drutovic D, Solc P, Schindler K. Aurora kinase A is essential for meiosis in mouse oocytes. PLoS Genet 2021; 17:e1009327. [PMID: 33901174 PMCID: PMC8102010 DOI: 10.1371/journal.pgen.1009327] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/06/2021] [Accepted: 04/08/2021] [Indexed: 12/18/2022] Open
Abstract
The Aurora protein kinases are well-established regulators of spindle building and chromosome segregation in mitotic and meiotic cells. In mouse oocytes, there is significant Aurora kinase A (AURKA) compensatory abilities when the other Aurora kinase homologs are deleted. Whether the other homologs, AURKB or AURKC can compensate for loss of AURKA is not known. Using a conditional mouse oocyte knockout model, we demonstrate that this compensation is not reciprocal because female oocyte-specific knockout mice are sterile, and their oocytes fail to complete meiosis I. In determining AURKA-specific functions, we demonstrate that its first meiotic requirement is to activate Polo-like kinase 1 at acentriolar microtubule organizing centers (aMTOCs; meiotic spindle poles). This activation induces fragmentation of the aMTOCs, a step essential for building a bipolar spindle. We also show that AURKA is required for regulating localization of TACC3, another protein required for spindle building. We conclude that AURKA has multiple functions essential to completing MI that are distinct from AURKB and AURKC.
Collapse
Affiliation(s)
- Cecilia S. Blengini
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- Human Genetics Institute of New Jersey; Piscataway, New Jersey, United States of America
| | - Patricia Ibrahimian
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
| | - Michaela Vaskovicova
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - David Drutovic
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Petr Solc
- Institute of Animal Physiology and Genetics of the Czech Academy of Sciences, Libechov, Czech Republic
| | - Karen Schindler
- Department of Genetics; Rutgers, The State University of New Jersey, Piscataway, New Jersey, United States of America
- Human Genetics Institute of New Jersey; Piscataway, New Jersey, United States of America
| |
Collapse
|
12
|
Priyanga J, Guha G, Bhakta-Guha D. Microtubule motors in centrosome homeostasis: A target for cancer therapy? Biochim Biophys Acta Rev Cancer 2021; 1875:188524. [PMID: 33582170 DOI: 10.1016/j.bbcan.2021.188524] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 01/02/2023]
Abstract
Cancer is a grievous concern to human health, owing to a massive heterogeneity in its cause and impact. Dysregulation (numerical, positional and/or structural) of centrosomes is one of the notable factors among those that promote onset and progression of cancers. In a normal dividing cell, a pair of centrosomes forms two poles, thereby governing the formation of a bipolar spindle assembly. A large number of cancer cells, however, harbor supernumerary centrosomes, which mimic the bipolar arrangement in normal cells by centrosome clustering (CC) into two opposite poles, thus developing a pseudo-bipolar spindle assembly. Manipulation of centrosome homeostasis is the paramount pre-requisite for the evasive strategy of CC in cancers. Out of the varied factors that uphold centrosome integrity, microtubule motors (MiMos) play a critical role. Categorized as dyneins and kinesins, MiMos are involved in cohesion of centrosomes, and also facilitate the maintenance of the numerical, positional and structural integrity of centrosomes. Herein, we elucidate the decisive mechanisms undertaken by MiMos to mediate centrosome homeostasis, and how dysregulation of the same might lead to CC in cancer cells. Understanding the impact of MiMos on CC might open up avenues toward a credible therapeutic target against diverse cancers.
Collapse
Affiliation(s)
- J Priyanga
- Cellular Dyshomeostasis Laboratory (CDHL), School of Chemical and Bio Technology, SASTRA University, Thanjavur 613 401, Tamil Nadu, India
| | - Gunjan Guha
- Cellular Dyshomeostasis Laboratory (CDHL), School of Chemical and Bio Technology, SASTRA University, Thanjavur 613 401, Tamil Nadu, India.
| | - Dipita Bhakta-Guha
- Cellular Dyshomeostasis Laboratory (CDHL), School of Chemical and Bio Technology, SASTRA University, Thanjavur 613 401, Tamil Nadu, India.
| |
Collapse
|
13
|
Zhao F, Feng Y, Zhang X, Liu X, Li A. Kinesin Superfamily Member 18B (KIF18B) Promotes Cell Proliferation in Colon Adenocarcinoma. Cancer Manag Res 2020; 12:12769-12778. [PMID: 33335427 PMCID: PMC7737937 DOI: 10.2147/cmar.s261894] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Accepted: 08/20/2020] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND The role of kinesin superfamily proteins (KIFs) has been reported in a variety of tumors and KIFs contributed to the proliferation of cancer cells. But few studies were focus on colon adenocarcinoma. METHODS Through bioinformatics analysis and immunohistochemistry (IHC) assays, the expression of KIF18B in colon adenocarcinoma tissues was determined. Stable KIF18B-depleted cell lines were constructed using lentivirus-mediated shRNA of KIF18B. Cell colony formation assay and CCK8 assay were performed to assess cell proliferation degree, and the expression level of KI67 and PCNA was used to indicate cell proliferation in vitro and verified using xenograft tumors in vivo. RESULTS KIF18B is highly expressed in colon adenocarcinoma tissues and has a negative correlation with the prognosis and tumor grade of colon adenocarcinoma. Interfering with KIF18B inhibits cell proliferation in vitro and in vivo. CONCLUSION KIF18B can be used as a prognostic marker for colon adenocarcinoma and may be a therapeutic target for colon adenocarcinoma treatment.
Collapse
Affiliation(s)
- Fei Zhao
- Department of Gastrointestinal Surgery, HanDan Central Hospital, Heibei, People's Republic of China
| | - Yunzhang Feng
- Department of Gastrointestinal Surgery, HanDan Central Hospital, Heibei, People's Republic of China
| | - Xueqiang Zhang
- HanDan Central Hospital President's Office, HanDan Central Hospital, Heibei, People's Republic of China
| | - Xiaohui Liu
- Department of Gastrointestinal Surgery, HanDan Central Hospital, Heibei, People's Republic of China
| | - Aili Li
- Department of Gynaecology, HanDan Central Hospital, Heibei, People’s Republic of China
| |
Collapse
|
14
|
Nuclear Isoforms of Neurofibromin Are Required for Proper Spindle Organization and Chromosome Segregation. Cells 2020; 9:cells9112348. [PMID: 33114250 PMCID: PMC7690890 DOI: 10.3390/cells9112348] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/30/2022] Open
Abstract
Mitotic spindles are highly organized, microtubule (MT)-based, transient structures that serve the fundamental function of unerring chromosome segregation during cell division and thus of genomic stability during tissue morphogenesis and homeostasis. Hence, a multitude of MT-associated proteins (MAPs) regulates the dynamic assembly of MTs in preparation for mitosis. Some tumor suppressors, normally functioning to prevent tumor development, have now emerged as significant MAPs. Among those, neurofibromin, the product of the Neurofibromatosis-1 gene (NF1), a major Ras GTPase activating protein (RasGAP) in neural cells, controls also the critical function of chromosome congression in astrocytic cellular contexts. Cell type- and development-regulated splicings may lead to the inclusion or exclusion of NF1exon51, which bears a nuclear localization sequence (NLS) for nuclear import at G2; yet the functions of the produced NLS and ΔNLS neurofibromin isoforms have not been previously addressed. By using a lentiviral shRNA system, we have generated glioblastoma SF268 cell lines with conditional knockdown of NLS or ΔNLS transcripts. In dissecting the roles of NLS or ΔNLS neurofibromins, we found that NLS-neurofibromin knockdown led to increased density of cytosolic MTs but loss of MT intersections, anastral spindles featuring large hollows and abnormal chromosome positioning, and finally abnormal chromosome segregation and increased micronuclei frequency. Therefore, we propose that NLS neurofibromin isoforms exert prominent mitotic functions.
Collapse
|
15
|
Stiff T, Echegaray-Iturra FR, Pink HJ, Herbert A, Reyes-Aldasoro CC, Hochegger H. Prophase-Specific Perinuclear Actin Coordinates Centrosome Separation and Positioning to Ensure Accurate Chromosome Segregation. Cell Rep 2020; 31:107681. [PMID: 32460023 PMCID: PMC7262599 DOI: 10.1016/j.celrep.2020.107681] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 02/11/2020] [Accepted: 05/01/2020] [Indexed: 12/30/2022] Open
Abstract
Centrosome separation in late G2/ early prophase requires precise spatial coordination that is determined by a balance of forces promoting and antagonizing separation. The major effector of centrosome separation is the kinesin Eg5. However, the identity and regulation of Eg5-antagonizing forces is less well characterized. By manipulating candidate components, we find that centrosome separation is reversible and that separated centrosomes congress toward a central position underneath the flat nucleus. This positioning mechanism requires microtubule polymerization, as well as actin polymerization. We identify perinuclear actin structures that form in late G2/early prophase and interact with microtubules emanating from the centrosomes. Disrupting these structures by breaking the interactions of the linker of nucleoskeleton and cytoskeleton (LINC) complex with perinuclear actin filaments abrogates this centrosome positioning mechanism and causes an increase in subsequent chromosome segregation errors. Our results demonstrate how geometrical cues from the cell nucleus coordinate the orientation of the emanating spindle poles before nuclear envelope breakdown.
Collapse
Affiliation(s)
- Tom Stiff
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Fabio R Echegaray-Iturra
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Harry J Pink
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | - Alex Herbert
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK
| | | | - Helfrid Hochegger
- Genome Damage and Stability Centre, School of Life Sciences, University of Sussex, Brighton BN19RQ, UK.
| |
Collapse
|
16
|
Omta WA, van Heesbeen RG, Shen I, de Nobel J, Robers D, van der Velden LM, Medema RH, Siebes APJM, Feelders AJ, Brinkkemper S, Klumperman JS, Spruit MR, Brinkhuis MJS, Egan DA. Combining Supervised and Unsupervised Machine Learning Methods for Phenotypic Functional Genomics Screening. SLAS DISCOVERY 2020; 25:655-664. [PMID: 32400262 DOI: 10.1177/2472555220919345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
There has been an increase in the use of machine learning and artificial intelligence (AI) for the analysis of image-based cellular screens. The accuracy of these analyses, however, is greatly dependent on the quality of the training sets used for building the machine learning models. We propose that unsupervised exploratory methods should first be applied to the data set to gain a better insight into the quality of the data. This improves the selection and labeling of data for creating training sets before the application of machine learning. We demonstrate this using a high-content genome-wide small interfering RNA screen. We perform an unsupervised exploratory data analysis to facilitate the identification of four robust phenotypes, which we subsequently use as a training set for building a high-quality random forest machine learning model to differentiate four phenotypes with an accuracy of 91.1% and a kappa of 0.85. Our approach enhanced our ability to extract new knowledge from the screen when compared with the use of unsupervised methods alone.
Collapse
Affiliation(s)
- Wienand A Omta
- Department of Cell Biology, Centre for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands.,Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands.,Core Life Analytics B.V., Utrecht, The Netherlands
| | - Roy G van Heesbeen
- Department of Cell Biology, NKI-AVL, Amsterdam, Noord-Holland, The Netherlands
| | - Ian Shen
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | - Jacob de Nobel
- Department of Cell Biology, Centre for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Desmond Robers
- Department of Cell Biology, Centre for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Lieke M van der Velden
- Department of Cell Biology, Centre for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - René H Medema
- Department of Cell Biology, NKI-AVL, Amsterdam, Noord-Holland, The Netherlands
| | - Arno P J M Siebes
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ad J Feelders
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | - Sjaak Brinkkemper
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | - Judith S Klumperman
- Department of Cell Biology, Centre for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
| | - Marco René Spruit
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | - Matthieu J S Brinkhuis
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | - David A Egan
- Core Life Analytics B.V., Utrecht, The Netherlands
| |
Collapse
|
17
|
Omta WA, van Heesbeen RG, Shen I, Feelders AJ, Brinkhuis M, Egan DA, Spruit MR. PurifyR: An R Package for Highly Automated, Reproducible Variable Extraction and Standardization. SYSTEMS MEDICINE 2020. [DOI: 10.1089/sysm.2019.0007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Affiliation(s)
- Wienand A. Omta
- Department of Cell Biology, Centre for Molecular Medicine, UMC Utrecht, Utrecht, The Netherlands
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
- Core Life Analytics B.V., Utrecht, The Netherlands
| | | | - Ian Shen
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | - Ad J. Feelders
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | - M.J.S. Brinkhuis
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| | | | - Marco R. Spruit
- Department of Information and Computing Sciences, Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
18
|
Bondaz A, Cirillo L, Meraldi P, Gotta M. Cell polarity-dependent centrosome separation in the C. elegans embryo. J Cell Biol 2019; 218:4112-4126. [PMID: 31645459 PMCID: PMC6891102 DOI: 10.1083/jcb.201902109] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 07/10/2019] [Accepted: 09/06/2019] [Indexed: 12/30/2022] Open
Abstract
Bondaz et al. show that in C. elegans embryos, the microtubule depolymerase KLP-7/MCAK is required for efficient centrosome separation in the somatic AB cell, but not the germline P1 cell. This difference in spindle assembly depends on cell polarity via the mitotic kinase PLK1. In animal cells, faithful chromosome segregation depends on the assembly of a bipolar spindle driven by the timely separation of the two centrosomes. Here we took advantage of the highly stereotypical cell divisions in Caenorhabditis elegans embryos to identify new regulators of centrosome separation. We find that at the two-cell stage, the somatic AB cell initiates centrosome separation later than the germline P1 cell. This difference is strongly exacerbated by the depletion of the kinesin-13 KLP-7/MCAK, resulting in incomplete centrosome separation at NEBD in AB but not P1. Our genetic and cell biology data indicate that this phenotype depends on cell polarity via the enrichment in AB of the mitotic kinase PLK-1, which itself limits the cortical localization of the dynein-binding NuMA orthologue LIN-5. We postulate that the timely separation of centrosomes is regulated in a cell type–dependent manner.
Collapse
Affiliation(s)
- Alexandra Bondaz
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Luca Cirillo
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Patrick Meraldi
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland .,Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Monica Gotta
- Department of Cell Physiology and Metabolism, Faculty of Medicine, University of Geneva, Geneva, Switzerland .,Translational Research Centre in Onco-hematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Swiss National Centre for Competence in Research in Chemical Biology, University of Geneva, Geneva, Switzerland
| |
Collapse
|
19
|
Myobatake Y, Kamisuki S, Tsukuda S, Higashi T, Chinen T, Takemoto K, Hachisuka M, Suzuki Y, Takei M, Tsurukawa Y, Maekawa H, Takeuchi T, Matsunaga TM, Sahara H, Usui T, Matsunaga S, Sugawara F. Pyrenocine A induces monopolar spindle formation and suppresses proliferation of cancer cells. Bioorg Med Chem 2019; 27:115149. [PMID: 31679979 DOI: 10.1016/j.bmc.2019.115149] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 10/25/2022]
Abstract
Pyrenocine A, a phytotoxin, was found to exhibit cytotoxicity against cancer cells with an IC50 value of 2.6-12.9 μM. Live cell imaging analysis revealed that pyrenocine A arrested HeLa cells at the M phase with characteristic ring-shaped chromosomes. Furthermore, as a result of immunofluorescence staining analysis, we found that pyrenocine A resulted in the formation of monopolar spindles in HeLa cells. Monopolar spindles are known to be induced by inhibitors of the kinesin motor protein Eg5 such as monastrol and STLC. Monastrol and STLC induce monopolar spindle formation and M phase arrest via inhibition of the ATPase activity of Eg5. Interestingly, our data revealed that pyrenocine A had no effect on the ATPase activity of Eg5 in vitro, which suggested the compound induces a monopolar spindle by an unknown mechanism. Structure-activity relationship analysis indicates that the enone structure of pyrenocine A is likely to be important for its cytotoxicity. An alkyne-tagged analog of pyrenocine A was synthesized and suppressed proliferation of HeLa cells with an IC50 value of 2.3 μM. We concluded that pyrenocine A induced monopolar spindle formation by a novel mechanism other than direct inhibition of Eg5 motor activity, and the activity of pyrenocine A may suggest a new anticancer mechanism.
Collapse
Affiliation(s)
- Yusuke Myobatake
- Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Shinji Kamisuki
- School of Veterinary Medicine, Azabu University, Fuchinobe, Sagamihara, Kanagawa, Japan.
| | - Senko Tsukuda
- Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Tsunehito Higashi
- Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Takumi Chinen
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Kenji Takemoto
- Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Masami Hachisuka
- School of Veterinary Medicine, Azabu University, Fuchinobe, Sagamihara, Kanagawa, Japan
| | - Yuka Suzuki
- School of Veterinary Medicine, Azabu University, Fuchinobe, Sagamihara, Kanagawa, Japan
| | - Maya Takei
- School of Veterinary Medicine, Azabu University, Fuchinobe, Sagamihara, Kanagawa, Japan
| | - Yukine Tsurukawa
- School of Veterinary Medicine, Azabu University, Fuchinobe, Sagamihara, Kanagawa, Japan
| | - Hiroaki Maekawa
- Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Toshifumi Takeuchi
- Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Tomoko M Matsunaga
- Research Institute for Science and Technology, Tokyo University of Science, Chiba, Japan
| | - Hiroeki Sahara
- School of Veterinary Medicine, Azabu University, Fuchinobe, Sagamihara, Kanagawa, Japan
| | - Takeo Usui
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Sachihiro Matsunaga
- Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| | - Fumio Sugawara
- Department of Applied Biological Science, Tokyo University of Science, Chiba, Japan
| |
Collapse
|
20
|
Dumas ME, Chen GY, Kendrick ND, Xu G, Larsen SD, Jana S, Waterson AG, Bauer JA, Hancock W, Sulikowski GA, Ohi R. Dual inhibition of Kif15 by oxindole and quinazolinedione chemical probes. Bioorg Med Chem Lett 2018; 29:148-154. [PMID: 30528696 DOI: 10.1016/j.bmcl.2018.12.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Revised: 11/30/2018] [Accepted: 12/04/2018] [Indexed: 11/29/2022]
Abstract
The mitotic spindle is a microtubule-based machine that segregates a replicated set of chromosomes during cell division. Many cancer drugs alter or disrupt the microtubules that form the mitotic spindle. Microtubule-dependent molecular motors that function during mitosis are logical alternative mitotic targets for drug development. Eg5 (Kinesin-5) and Kif15 (Kinesin-12), in particular, are an attractive pair of motor proteins, as they work in concert to drive centrosome separation and promote spindle bipolarity. Furthermore, we hypothesize that the clinical failure of Eg5 inhibitors may be (in part) due to compensation by Kif15. In order to test this idea, we screened a small library of kinase inhibitors and identified GW108X, an oxindole that inhibits Kif15 in vitro. We show that GW108X has a distinct mechanism of action compared with a commercially available Kif15 inhibitor, Kif15-IN-1 and may serve as a lead with which to further develop Kif15 inhibitors as clinically relevant agents.
Collapse
Affiliation(s)
- Megan E Dumas
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, United States
| | - Geng-Yuan Chen
- Department of Biomedical Engineering, Pennsylvania State University, State College, PA, United States
| | - Nicole D Kendrick
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN 37232, United States
| | - George Xu
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Scott D Larsen
- Department of Medicinal Chemistry, University of Michigan, Ann Arbor, MI 48109, United States
| | - Somnath Jana
- Vanderbilt Institute of Chemical Biology, Nashville, TN 37232, United States
| | - Alex G Waterson
- Vanderbilt Institute of Chemical Biology, Nashville, TN 37232, United States; Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States; Department of Pharmacology, Vanderbilt University, Nashville, TN 37232, United States
| | - Joshua A Bauer
- Department of Biochemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - William Hancock
- Department of Biomedical Engineering, Pennsylvania State University, State College, PA, United States
| | - Gary A Sulikowski
- Department of Chemistry, Vanderbilt University, Nashville, TN 37232, United States
| | - Ryoma Ohi
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
21
|
Parts list for a microtubule depolymerising kinesin. Biochem Soc Trans 2018; 46:1665-1672. [PMID: 30467119 PMCID: PMC6299235 DOI: 10.1042/bst20180350] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 10/11/2018] [Accepted: 10/15/2018] [Indexed: 12/12/2022]
Abstract
The Kinesin superfamily is a large group of molecular motors that use the turnover of ATP to regulate their interaction with the microtubule cytoskeleton. The coupled relationship between nucleotide turnover and microtubule binding is harnessed in various ways by these motors allowing them to carry out a variety of cellular functions. The Kinesin-13 family is a group of specialist microtubule depolymerising motors. Members of this family use their microtubule destabilising activity to regulate processes such as chromosome segregation, maintenance of cilia and neuronal development. Here, we describe the current understanding of the structure of this family of kinesins and the role different parts of these proteins play in their microtubule depolymerisation activity and in the wider function of this family of kinesins.
Collapse
|
22
|
Tang F, Pan MH, Wan X, Lu Y, Zhang Y, Sun SC. Kif18a regulates Sirt2-mediated tubulin acetylation for spindle organization during mouse oocyte meiosis. Cell Div 2018; 13:9. [PMID: 30459823 PMCID: PMC6234775 DOI: 10.1186/s13008-018-0042-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 10/26/2018] [Indexed: 12/17/2022] Open
Abstract
Background During oocyte meiosis, the cytoskeleton dynamics, especially spindle organization, are critical for chromosome congression and segregation. However, the roles of the kinesin superfamily in this process are still largely unknown. Results In the present study, Kif18a, a member of the kinesin-8 family, regulated spindle organization through its effects on tubulin acetylation in mouse oocyte meiosis. Our results showed that Kif18a is expressed and mainly localized in the spindle region. Knock down of Kif18a caused the failure of first polar body extrusion, dramatically affecting spindle organization and resulting in severe chromosome misalignment. Further analysis showed that the disruption of Kif18a caused an increase in acetylated tubulin level, which might be the reason for the spindle organization defects after Kif18a knock down in oocyte meiosis, and the decreased expression of deacetylase Sirt2 was found after Kif18a knock down. Moreover, microinjections of tubulin K40R mRNA, which could induce tubulin deacetylation, protected the oocytes from the effects of Kif18a downregulation, resulting in normal spindle morphology in Kif18a-knock down oocytes. Conclusions Taken together, our results showed that Kif18a affected Sirt2-mediated tubulin acetylation level for spindle organization during mouse oocyte meiosis. Our results not only revealed the critical effect of Kif18a on microtubule stability, but also extended our understanding of kinesin activity in meiosis.
Collapse
Affiliation(s)
- Feng Tang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Meng-Hao Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Xiang Wan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Yujie Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095 China
| |
Collapse
|
23
|
Manukyan A, Sargsyan L, Parsons SJ, Stukenberg PT. P190RhoGAP prevents mitotic spindle fragmentation and is required to activate Aurora A kinase at acentriolar poles. Chromosoma 2018; 127:375-386. [PMID: 29656322 DOI: 10.1007/s00412-018-0670-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Revised: 03/20/2018] [Accepted: 03/21/2018] [Indexed: 02/03/2023]
Abstract
Assembly of the mitotic spindle is essential for proper chromosome segregation during mitosis. Maintenance of spindle poles requires precise regulation of kinesin- and dynein-generated forces, and improper regulation of these forces disrupts pole integrity leading to pole fragmentation. The formation and function of the mitotic spindle are regulated by many proteins, including Aurora A kinase and the motor proteins Kif2a and Eg5. Here, we characterize a surprising role for the RhoA GTPase-activating protein, p190RhoGAP, in regulating the mitotic spindle. We show that cells depleted of p190RhoGAP arrest for long periods in mitosis during which cells go through multiple transitions between having bipolar and multipolar spindles. Most of the p190RhoGAP-depleted cells finally achieve a stable bipolar attachment and proceed through anaphase. The multipolar spindle phenotype can be rescued by low doses of an Eg5 inhibitor. Moreover, we show that p190RhoGAP-depleted multipolar cells localize Aurora A to all the poles, but the kinase is only activated at the two centriolar poles. Overall, our data identify an unappreciated connection between p190RhoGAP and the proteins that control spindle poles including Aurora A kinase and Eg5 that is required to prevent or correct spindle pole fragmentation.
Collapse
Affiliation(s)
- Arkadi Manukyan
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Lilit Sargsyan
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA
| | - Sarah J Parsons
- Department of Microbiology, Immunology and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - P Todd Stukenberg
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA, 22908, USA.
- , Charlottesville, USA.
| |
Collapse
|
24
|
Biochemical and Biophysical characterization of curcumin binding to human mitotic kinesin Eg5: Insights into the inhibitory mechanism of curcumin on Eg5. Int J Biol Macromol 2018; 109:1189-1208. [DOI: 10.1016/j.ijbiomac.2017.11.115] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2017] [Revised: 11/15/2017] [Accepted: 11/17/2017] [Indexed: 02/06/2023]
|
25
|
Kinetochore-microtubule interactions in chromosome segregation: lessons from yeast and mammalian cells. Biochem J 2017; 474:3559-3577. [PMID: 29046344 DOI: 10.1042/bcj20170518] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Revised: 08/24/2017] [Accepted: 09/11/2017] [Indexed: 02/06/2023]
Abstract
Chromosome congression and segregation require robust yet dynamic attachment of the kinetochore with the spindle microtubules. Force generated at the kinetochore-microtubule interface plays a vital role to drive the attachment, as it is required to move chromosomes and to provide signal to sense correct attachments. To understand the mechanisms underlying these processes, it is critical to describe how the force is generated and how the molecules at the kinetochore-microtubule interface are organized and assembled to withstand the force and respond to it. Research in the past few years or so has revealed interesting insights into the structural organization and architecture of kinetochore proteins that couple kinetochore attachment to the spindle microtubules. Interestingly, despite diversities in the molecular players and their modes of action, there appears to be architectural similarity of the kinetochore-coupling machines in lower to higher eukaryotes. The present review focuses on the most recent advances in understanding of the molecular and structural aspects of kinetochore-microtubule interaction based on the studies in yeast and vertebrate cells.
Collapse
|
26
|
Mann BJ, Balchand SK, Wadsworth P. Regulation of Kif15 localization and motility by the C-terminus of TPX2 and microtubule dynamics. Mol Biol Cell 2016; 28:65-75. [PMID: 27852894 PMCID: PMC5221630 DOI: 10.1091/mbc.e16-06-0476] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2016] [Revised: 11/04/2016] [Accepted: 11/07/2016] [Indexed: 12/31/2022] Open
Abstract
Mitotic motor proteins generate force to establish and maintain spindle bipolarity, but how they are temporally and spatially regulated in vivo is unclear. Prior work demonstrated that a microtubule-associated protein, TPX2, targets kinesin-5 and kinesin-12 motors to spindle microtubules. The C-terminal domain of TPX2 contributes to the localization and motility of the kinesin-5, Eg5, but it is not known whether this domain regulates kinesin-12, Kif15. We found that the C-terminal domain of TPX2 contributes to the localization of Kif15 to spindle microtubules in cells and suppresses motor walking in vitro. Kif15 and Eg5 are partially redundant motors, and overexpressed Kif15 can drive spindle formation in the absence of Eg5 activity. Kif15-dependent bipolar spindle formation in vivo requires the C-terminal domain of TPX2. In the spindle, fluorescent puncta of GFP-Kif15 move toward the equatorial region at a rate equivalent to microtubule growth. Reduction of microtubule growth with paclitaxel suppresses GFP-Kif15 motility, demonstrating that dynamic microtubules contribute to Kif15 behavior. Our results show that the C-terminal region of TPX2 regulates Kif15 in vitro, contributes to motor localization in cells, and is required for Kif15 force generation in vivo and further reveal that dynamic microtubules contribute to Kif15 behavior in vivo.
Collapse
Affiliation(s)
- Barbara J Mann
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Sai K Balchand
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| | - Patricia Wadsworth
- Department of Biology and Program in Molecular and Cellular Biology, University of Massachusetts Amherst, Amherst, MA 01003
| |
Collapse
|