1
|
Lee SH, Kwon MS, Lee T, Hohng S, Lee H. Kinesin-like protein KIF18A is required for faithful coordination of chromosome congression with cytokinesis. FEBS J 2025. [PMID: 39954259 DOI: 10.1111/febs.70019] [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: 09/06/2024] [Revised: 12/17/2024] [Accepted: 01/31/2025] [Indexed: 02/17/2025]
Abstract
The maintenance of genetic integrity in proliferating cells requires the coordinated regulation of DNA replication, chromosome segregation, and cytokinetic abscission. Chromosome-microtubule interactions regulate mitosis, while interactions between the actin cytoskeleton and Myosin IIA dictate cytokinetic abscission. This process, crucial for the equal distribution of the duplicated genome into two daughter cells, occurs perpendicular to the axis of chromosome segregation. However, the mechanism of how microtubule-driven mitosis and actin-associated cytokinesis are precisely coordinated remains poorly understood. This study highlights the role of KIF18A, a kinesin-like protein, in linking kinetochore-microtubule dynamics to cytokinetic axis formation. KIF18A's localization changes through the cell division cycle, from the metaphase plate during chromosome congression to the central spindle in late anaphase, and finally to the spindle midbody in telophase. KIF18A depletion leads to chromosome congression failures and anaphase onset delays. Notably, cells attempting to undergo division in the absence of KIF18A exhibited disruptions in the parallel structure of the central spindle, causing mislocalization of the centralspindlin complex, such as kinesin-like protein KIF23 (also known as MKLP1) and Rac GTPase-activating protein 1 (RACGAP1). These disruptions impair cleavage furrow establishment, causing incomplete cytokinesis and the formation of mononuclear or binucleated cells. Our findings suggest that KIF18A is crucial for coordinating chromosome congression and cytokinesis by regulating the spatial and temporal assembly of the central spindle during late anaphase.
Collapse
Affiliation(s)
- Su Hyun Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Korea
| | - Mi-Sun Kwon
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Korea
| | - Taerim Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Korea
| | - Sungchul Hohng
- Department of Physics and Astronomy, Seoul National University, Korea
| | - Hyunsook Lee
- Department of Biological Sciences, Institute of Molecular Biology and Genetics, Seoul National University, Korea
| |
Collapse
|
2
|
Fujishiro S, Sasai M. Three-dimensional memory of nuclear organization through cell cycles. J Chem Phys 2025; 162:065103. [PMID: 39945369 DOI: 10.1063/5.0242859] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 01/25/2025] [Indexed: 05/09/2025] Open
Abstract
The genome in the cell nucleus is organized by a dynamic process influenced by structural memory from mitosis. In this study, we develop a model of human genome dynamics through cell cycles by extending the previously developed whole-genome model to cover the mitotic phase. With this extension, we focus on the role of mitotic and cell cycle memory in genome organization. The simulation progresses from mitosis to interphase and the subsequent mitosis, leading to successive cell cycles. During mitosis, our model describes microtubule dynamics, showing how forces orchestrate the assembly of chromosomes into a rosette ring structure at metaphase. The model explains how the positioning of chromosomes depends on their size in metaphase. The memory of the metaphase configuration persists through mitosis and into interphase in dimensions perpendicular to the cell division axis, effectively guiding the distribution of chromosome territories over multiple cell cycles. At the onset of each G1 phase, phase separation of active and inactive chromatin domains occurs, leading to A/B compartmentalization. Our cycling simulations show that the compartments are unaffected by structural memory from previous cycles and are consistently established in each cell cycle. The genome model developed in this study highlights the interplay between chromosome dynamics and structural memory across cell cycles, providing insights for the analyses of cellular processes.
Collapse
Affiliation(s)
- Shin Fujishiro
- Fukui Institute for Fundamental Chemistry, Kyoto University, Kyoto 606-8103, Japan
| | - Masaki Sasai
- Fukui Institute for Fundamental Chemistry, Kyoto University, Kyoto 606-8103, Japan
- Department of Complex Systems Science, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
3
|
Yildiz A. Mechanism and regulation of kinesin motors. Nat Rev Mol Cell Biol 2025; 26:86-103. [PMID: 39394463 DOI: 10.1038/s41580-024-00780-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/13/2024]
Abstract
Kinesins are a diverse superfamily of microtubule-based motors that perform fundamental roles in intracellular transport, cytoskeletal dynamics and cell division. These motors share a characteristic motor domain that powers unidirectional motility and force generation along microtubules, and they possess unique tail domains that recruit accessory proteins and facilitate oligomerization, regulation and cargo recognition. The location, direction and timing of kinesin-driven processes are tightly regulated by various cofactors, adaptors, microtubule tracks and microtubule-associated proteins. This Review focuses on recent structural and functional studies that reveal how members of the kinesin superfamily use the energy of ATP hydrolysis to transport cargoes, depolymerize microtubules and regulate microtubule dynamics. I also survey how accessory proteins and post-translational modifications regulate the autoinhibition, cargo binding and motility of some of the best-studied kinesins. Despite much progress, the mechanism and regulation of kinesins are still emerging, and unresolved questions can now be tackled using newly developed approaches in biophysics and structural biology.
Collapse
Affiliation(s)
- Ahmet Yildiz
- Physics Department, University of California at Berkeley, Berkeley, CA, USA.
- Department of Molecular and Cellular Biology, University of California at Berkeley, Berkeley, CA, USA.
| |
Collapse
|
4
|
Phillips AF, Zhang R, Jaffe M, Schulz R, Carty MC, Verma A, Feinberg TY, Arensman MD, Chiu A, Letso R, Bosco N, Queen KA, Racela AR, Stumpff J, Andreu-Agullo C, Bettigole SE, Depetris RS, Drutman S, Su SM, Cogan DA, Eng CH. Targeting chromosomally unstable tumors with a selective KIF18A inhibitor. Nat Commun 2025; 16:307. [PMID: 39747049 PMCID: PMC11697083 DOI: 10.1038/s41467-024-55300-z] [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: 02/28/2023] [Accepted: 12/05/2024] [Indexed: 01/04/2025] Open
Abstract
Chromosome instability is a prevalent vulnerability of cancer cells that has yet to be fully exploited therapeutically. To identify genes uniquely essential to chromosomally unstable cells, we mined the Cancer Dependency Map for genes essential in tumor cells with high levels of copy number aberrations. We identify and validate KIF18A, a mitotic kinesin, as a vulnerability of chromosomally unstable cancer cells. Knockdown of KIF18A leads to mitotic defects and reduction of tumor growth. Screening of a chemical library for inhibitors of KIF18A enzymatic activity identified a hit that was optimized to yield VLS-1272, which is orally bioavailable, potent, ATP non-competitive, microtubule-dependent, and highly selective for KIF18A versus other kinesins. Inhibition of KIF18A's ATPase activity prevents KIF18A translocation across the mitotic spindle, resulting in chromosome congression defects, mitotic cell accumulation, and cell death. Profiling VLS-1272 across >100 cancer cell lines demonstrates that the specificity towards cancer cells with chromosome instability differentiates KIF18A inhibition from other clinically tested anti-mitotic drugs. Treatment of tumor xenografts with VLS-1272 results in mitotic defects leading to substantial, dose-dependent inhibition of tumor growth. The strong biological rationale, robust preclinical data, and optimized compound properties enable the clinical development of a KIF18A inhibitor in cancers with high chromosomal instability.
Collapse
Affiliation(s)
| | | | - Mia Jaffe
- Volastra Therapeutics, New York, NY, USA
| | | | | | | | | | | | - Alan Chiu
- Volastra Therapeutics, New York, NY, USA
| | - Reka Letso
- Volastra Therapeutics, New York, NY, USA
| | | | - Katelyn A Queen
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Allison R Racela
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, USA
| | | | | | | | | | | | | | | |
Collapse
|
5
|
Wang Y, Zhou B, Lian X, Yu S, Huang B, Wu X, Wen L, Zhu C. KIF18A Is a Novel Target of JNK1/c-Jun Signaling Pathway Involved in Cervical Tumorigenesis. J Cell Physiol 2025; 240:e31516. [PMID: 39749722 DOI: 10.1002/jcp.31516] [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: 06/12/2024] [Revised: 11/21/2024] [Accepted: 12/13/2024] [Indexed: 01/04/2025]
Abstract
Cervical cancer remains a significant global health concern. KIF18A, a kinesin motor protein regulating microtubule dynamics during mitosis, is frequently overexpressed in various cancers, but its regulatory mechanisms are poorly understood. This study investigates KIF18A's role in cervical cancer and its regulation by the JNK1/c-Jun signaling pathway. Cell growth was assessed in vitro using MTT and colony formation assays, and in vivo using a nude mouse xenograft model with KIF18A knockdown HeLa cells. The Genomic Data Commons (GDC) data portal was used to identify KIF18A-related protein kinases in cervical cancer. Western blot analysis was employed to analyze phosphor-c-Jun, c-Jun, and KIF18A expression levels following JNK1 inhibition, c-Jun knockdown/overexpression, and KIF18A knockdown in cervical cancer cells. Chromatin immunoprecipitation (ChIP) and luciferase reporter assays were performed to assess c-Jun binding and transcriptional activity of the KIF18A promoter. KIF18A knockdown significantly impaired cervical cancer cell growth both in vitro and in vivo. A strong positive correlation was observed between JNK1 and KIF18A expression in cervical and other cancers. JNK1 inhibition decreased both KIF18A expression and c-Jun phosphorylation. c-Jun was found to directly bind to and activate the KIF18A promoter. Furthermore, c-Jun knockdown inhibited cervical cancer cell growth, and this effect was partially rescued by KIF18A overexpression. This study demonstrates that the JNK1/c-Jun pathway activates KIF18A expression, which is essential for cervical cancer cell growth. Targeting the JNK/c-Jun/KIF18A axis may represent a promising novel therapeutic strategy for cancer treatment.
Collapse
Affiliation(s)
- Yajie Wang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Bowen Zhou
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Xiaoying Lian
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Siqi Yu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Baihai Huang
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Xinyue Wu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Lianpu Wen
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| | - Changjun Zhu
- Tianjin Key Laboratory of Animal and Plant Resistance, College of Life Sciences, Tianjin Normal University, Tianjin, China
| |
Collapse
|
6
|
Nesbit C, Martin W, Czechanski A, Byers C, Raghupathy N, Ferraj A, Stumpff J, Reinholdt L. Anapc5 and Anapc7 as genetic modifiers of KIF18A function in fertility and mitotic progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626395. [PMID: 39677807 PMCID: PMC11642851 DOI: 10.1101/2024.12.03.626395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
The kinesin family member 18A (KIF18A) is an essential regulator of microtubule dynamics and chromosome alignment during mitosis. Functional dependency on KIF18A varies by cell type and genetic context but the heritable factors that influence this dependency remain unknown. To address this, we took advantage of the variable penetrance observed in different mouse strain backgrounds to screen for loci that modulate germ cell depletion in the absence of KIF18A. We found a significant association at a Chr5 locus where anaphase promoting complex subunits 5 (Anapc5) and 7 (Anapc7) were the top candidate genes. We found that both genes were differentially expressed in a sensitive strain background when compared to resistant strain background at key timepoints in gonadal development. We also identified a novel retroviral insertion in Anapc7 that may in part explain the observed expression differences. In cell line models, we found that depletion of KIF18A induced mitotic arrest, which was partially rescued by co-depletion of ANAPC7 (APC7) and exacerbated by co-depletion of ANAPC5 (APC5). These findings suggest that differential expression and activity of Anapc5 and Anapc7 may influence sensitivity to KIF18A depletion in germ cells and CIN cells, with potential implications for optimizing antineoplastic therapies.
Collapse
Affiliation(s)
- Carleigh Nesbit
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT
| | | | | | - Candice Byers
- The Roux Institute at Northeastern University, Portland, ME
| | | | | | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT
| | | |
Collapse
|
7
|
Mohd Amin AS, Eastwood S, Pilcher C, Truong JQ, Foitzik R, Boag J, Gorringe KL, Holien JK. KIF18A inhibition: the next big player in the search for cancer therapeutics. Cancer Metastasis Rev 2024; 44:3. [PMID: 39580563 DOI: 10.1007/s10555-024-10225-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 10/17/2024] [Indexed: 11/25/2024]
Abstract
Kinesin-like protein 18A (KIF18A) is a member of the kinesin family of molecular motor proteins, which utilise energy from the hydrolysis of adenosine triphosphate (ATP) to regulate critical cellular processes such as chromosome movement and microtubule dynamics. KIF18A plays a vital role in controlling microtubule length, which is crucial for maintaining proper cell function and division. Notably, increased expression levels of KIF18A have been observed in various types of cancer, indicating its potential involvement in tumour progression. Although preclinical studies have demonstrated that KIF18A is not essential for normal somatic cell division, it appears to be crucial for the survival and division of cancer cells, particularly those exhibiting chromosomal instability. This dependency makes KIF18A a promising target for developing new therapeutic strategies aimed at treating chromosomally unstable cancers. This review delves into the structural and functional aspects of KIF18A, and its role in cancer development, and evaluates current and emerging approaches to targeting KIF18A with innovative cancer treatments.
Collapse
Affiliation(s)
| | - Sarah Eastwood
- STEM College, RMIT University, 225-245 Plenty Rd, Bundoora, VIC, 3083, Australia
| | - Courtney Pilcher
- STEM College, RMIT University, 225-245 Plenty Rd, Bundoora, VIC, 3083, Australia
| | - Jia Q Truong
- STEM College, RMIT University, 225-245 Plenty Rd, Bundoora, VIC, 3083, Australia
| | - Richard Foitzik
- Oncology One Pty Ltd, 305 Grattan St, Melbourne, VIC, 3000, Australia
- Inosi Therapeutics Pty Ltd, 655 Elizabeth St, Melbourne, VIC, 3000, Australia
| | - Joanne Boag
- Oncology One Pty Ltd, 305 Grattan St, Melbourne, VIC, 3000, Australia
- The Walter and Eliza Hall Institute of Medical Research, 4 Research Avenue, Bundoora, VIC, 3083, Australia
- Ternarx Pty Ltd, 4 Research Avenue, Bundoora, VIC, 3083, Australia
| | - Kylie L Gorringe
- Peter MacCallum Cancer Centre, 305 Grattan St, Melbourne, VIC, 3000, Australia
- Department of Oncology, Sir Peter MacCallum, The University of Melbourne, Grattan St, Parkville, VIC, 3010, Australia
| | - Jessica K Holien
- STEM College, RMIT University, 225-245 Plenty Rd, Bundoora, VIC, 3083, Australia.
- The Faculty of Medicine, Dentistry and Health Science, The University of Melbourne, Grattan St, Parkville, VIC, 3010, Australia.
- St Vincent's Institute of Medical Research, 9 Princes St, Fitzroy, VIC, 3052, Australia.
| |
Collapse
|
8
|
Risteski P, Martinčić J, Jagrić M, Tintor E, Petelinec A, Tolić IM. Microtubule poleward flux as a target for modifying chromosome segregation errors. Proc Natl Acad Sci U S A 2024; 121:e2405015121. [PMID: 39541344 PMCID: PMC11588092 DOI: 10.1073/pnas.2405015121] [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: 03/13/2024] [Accepted: 10/07/2024] [Indexed: 11/16/2024] Open
Abstract
Cancer cells often display errors in chromosome segregation, some of which result from improper chromosome alignment at the spindle midplane. Chromosome alignment is facilitated by different rates of microtubule poleward flux between sister kinetochore fibers. However, the role of the poleward flux in supporting mitotic fidelity remains unknown. Here, we introduce the hypothesis that the finely tuned poleward flux safeguards against lagging chromosomes and micronuclei at mitotic exit by promoting chromosome alignment in metaphase. We used human untransformed RPE-1 cells depleted of KIF18A/kinesin-8 as a system with reduced mitotic fidelity, which we rescued by three mechanistically independent treatments, comprising low-dose taxol or codepletion of the spindle proteins HAUS8 or NuMA. The rescue of mitotic errors was due to shortening of the excessively long overlaps of antiparallel microtubules, serving as a platform for motor proteins that drive the flux, which in turn slowed down the overly fast flux and improved chromosome alignment. In contrast to the prevailing view, the rescue was not accompanied by reduction of overall microtubule growth rates. Instead, speckle microscopy revealed that the improved chromosome alignment in the rescue treatments was associated with slower growth and flux of kinetochore microtubules. In a similar manner, a low-dose taxol treatment rescued mitotic errors in a high-grade serous ovarian carcinoma cell line OVKATE. Collectively, our results highlight the potential of targeting microtubule poleward flux to modify chromosome instability and provide insight into the mechanism through which low doses of taxol rescue certain mitotic errors in cancer cells.
Collapse
Affiliation(s)
- Patrik Risteski
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb10000, Croatia
| | - Jelena Martinčić
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb10000, Croatia
| | - Mihaela Jagrić
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb10000, Croatia
| | - Erna Tintor
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb10000, Croatia
| | - Ana Petelinec
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb10000, Croatia
| | - Iva M. Tolić
- Laboratory of Cell Biophysics, Division of Molecular Biology, Ruđer Bošković Institute, Zagreb10000, Croatia
| |
Collapse
|
9
|
Serpico AF, Pisauro C, Trano A, Grieco D. Chromosome alignment and Kif18A action rely on spindle-localized control of Cdk1 activity. Front Cell Dev Biol 2024; 12:1490781. [PMID: 39610707 PMCID: PMC11602486 DOI: 10.3389/fcell.2024.1490781] [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: 09/03/2024] [Accepted: 10/29/2024] [Indexed: 11/30/2024] Open
Abstract
Introduction During mitosis, chromosome alignment at the mitotic spindle equator grants correct chromosome segregation and proper nuclei formation in daughter cells. The kinesin 8 family member Kif18A plays a crucial role for chromosome alignment by localizing at the kinetochore-microtubule (K-MT) plus ends to dampen MT dynamics and stabilize K-MT attachments. Kif18A action is directly antagonized by the master mitotic kinase cyclin B-dependent kinase 1 (Cdk1) and is promoted by protein phosphatase 1 (PP1). Since chromosome alignment precedes Cdk1 inactivation by cyclin B proteolysis, it is unclear how Kif18A evades Cdk1 inhibition. Methods We analyzed chromosome alignment and Kif18A in mitotic cells upon genetic perturbation of the phosphorylation-dependent inhibitory control of Cdk1 activity by immunofluorescence and cell fractionation experiments. Results We show here that chromosome alignment in human cells relies on a recently identified fraction of Cdk1 that is inhibited by Wee1-dependent phosphorylation in mitosis (i-Cdk1, standing for inhibited/inactive-Cdk1) and that localized at spindle structures where it promotes proper spindle assembly. Indeed, the reduction of i-Cdk1 led to several spindle defects including spindles with misaligned, bipolarly attached chromosomes showing poor Kif18A localization at their K-MT plus ends. Restoring i-Cdk1 reversed both alignment defects and Kif18A localization. In cells with lowered i-Cdk1, expressing a phosphonull Kif18A mutant version at the sites that serve as Cdk1 substrate significantly rescued the alignment defects. Discussion Mechanistically, our evidence suggests that i-Cdk1 and active PP1 facilitated the dephosphorylation and reactivation of spindle-localized Kif18A. Considering the relevance of Kif18A for survival of aneuploid cancer cells and the potential therapeutic targeting of both Kif18A and Wee1, these findings could also be relevant for cancer therapy.
Collapse
Affiliation(s)
- Angela Flavia Serpico
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), University of Naples “Federico II”, Naples, Italy
| | | | - Asia Trano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), University of Naples “Federico II”, Naples, Italy
| | - Domenico Grieco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche (DMMBM), University of Naples “Federico II”, Naples, Italy
- CEINGE Biotecnologie Avanzate “Franco Salvatore”, Naples, Italy
| |
Collapse
|
10
|
Perez-Bertoldi JM, Zhao Y, Thawani A, Yildiz A, Nogales E. HURP regulates Kif18A recruitment and activity to synergistically control microtubule dynamics. Nat Commun 2024; 15:9687. [PMID: 39516196 PMCID: PMC11549086 DOI: 10.1038/s41467-024-53691-7] [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: 04/10/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
During mitosis, microtubule dynamics are regulated to ensure proper alignment and segregation of chromosomes. The dynamics of kinetochore-attached microtubules are regulated by hepatoma-upregulated protein (HURP) and the mitotic kinesin-8 Kif18A, but the underlying mechanism remains elusive. Using single-molecule imaging in vitro, we demonstrate that Kif18A motility is regulated by HURP. While sparse decoration of HURP activates the motor, higher concentrations hinder processive motility. To shed light on this behavior, we determine the binding mode of HURP to microtubules using cryo-EM. The structure helps rationalize why HURP functions as a microtubule stabilizer. Additionally, HURP partially overlaps with the microtubule-binding site of the Kif18A motor domain, indicating that excess HURP inhibits Kif18A motility by steric exclusion. We also observe that HURP and Kif18A function together to suppress dynamics of the microtubule plus-end, providing a mechanistic basis for how they collectively serve in microtubule length control.
Collapse
Affiliation(s)
| | - Yuanchang Zhao
- Physics Department, University of California, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Akanksha Thawani
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - Ahmet Yildiz
- Biophysics Graduate Group, University of California, Berkeley, CA, USA.
- Physics Department, University of California, Berkeley, CA, USA.
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
| | - Eva Nogales
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Laboratory, Berkeley, CA, USA.
- Howard Hughes Medical Institute, University of California, Berkeley, CA, USA.
| |
Collapse
|
11
|
Biswas L, Tyc KM, Aboelenain M, Sun S, Dundović I, Vukušić K, Liu J, Guo V, Xu M, Scott RT, Tao X, Tolić IM, Xing J, Schindler K. Maternal genetic variants in kinesin motor domains prematurely increase egg aneuploidy. Proc Natl Acad Sci U S A 2024; 121:e2414963121. [PMID: 39475646 PMCID: PMC11551467 DOI: 10.1073/pnas.2414963121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 09/27/2024] [Indexed: 11/06/2024] Open
Abstract
The female reproductive lifespan is highly dependent on egg quality, especially the presence of a normal number of chromosomes in an egg, known as euploidy. Mistakes in meiosis leading to egg aneuploidy are frequent in humans. Yet, knowledge of the precise genetic landscape that causes egg aneuploidy in women is limited, as phenotypic data on the frequency of human egg aneuploidy are difficult to obtain and therefore absent in public genetic datasets. Here, we identify genetic determinants of reproductive aging via egg aneuploidy in women using a biobank of individual maternal exomes linked with maternal age and embryonic aneuploidy data. Using the exome data, we identified 404 genes bearing variants enriched in individuals with pathologically elevated egg aneuploidy rates. Analysis of the gene ontology and protein-protein interaction network implicated genes encoding the kinesin protein family in egg aneuploidy. We interrogate the causal relationship of the human variants within candidate kinesin genes via experimental perturbations and demonstrate that motor domain variants increase aneuploidy in mouse oocytes. Finally, using a knock-in mouse model, we validate that a specific variant in kinesin KIF18A accelerates reproductive aging and diminishes fertility. These findings reveal additional functional mechanisms of reproductive aging and shed light on how genetic variation underlies individual heterogeneity in the female reproductive lifespan, which might be leveraged to predict reproductive longevity. Together, these results lay the groundwork for the noninvasive biomarkers for egg quality, a first step toward personalized fertility medicine.
Collapse
Affiliation(s)
- Leelabati Biswas
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Rutgers Robert Wood Johnson Medical School, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Katarzyna M. Tyc
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Mansour Aboelenain
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Department of Theriogenology, Faculty of Veterinary Medicine, Mansoura University, Mansoura35516, Egypt
| | - Siqi Sun
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Iva Dundović
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Kruno Vukušić
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Jason Liu
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | | | - Min Xu
- Department of Statistics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | | | - Xin Tao
- Juno Genetics US, Basking Ridge, NJ07920
| | - Iva M. Tolić
- Department of Molecular Biology, Ruđer Bošković Institute, Zagreb1000, Croatia
| | - Jinchuan Xing
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| | - Karen Schindler
- Department of Genetics, Rutgers, The State University of New Jersey, Piscataway, NJ08854
- Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ08854
| |
Collapse
|
12
|
Lim Y, Cho YB, Seo YJ. Emerging roles of cytoskeletal transport and scaffold systems in human viral propagation. Anim Cells Syst (Seoul) 2024; 28:506-518. [PMID: 39439927 PMCID: PMC11494721 DOI: 10.1080/19768354.2024.2418332] [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: 05/17/2024] [Revised: 09/13/2024] [Accepted: 10/08/2024] [Indexed: 10/25/2024] Open
Abstract
Viruses have long been recognized as significant pathogens, contributing to multiple global pandemics throughout human history. Recent examples include the 2009 influenza pandemic and the COVID-19 pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in 2019. Despite ongoing experimental and clinical efforts, the development of effective antiviral treatments and vaccines remains challenging due to the high mutation rates of many human pathogenic viruses including influenza virus and SARS-CoV-2. As an alternative approach, antiviral strategies targeting host factors shared by multiple viruses could provide a more universally applicable solution. Emerging evidence suggests that viruses exploit the host cytoskeletal network to facilitate efficient viral replication and propagation. Therefore, a comprehensive understanding of the interactions between viral components and the cytoskeletal machinery may offer valuable insights for the development of broad-spectrum antiviral therapeutics. This review compiles and discusses current knowledge on the interactions between viruses and cytoskeletal elements, including kinesin, dynein, myosin, and vimentin, and explores their potential as therapeutic targets. The potential for these cytoskeletal components to serve as targets for new antiviral interventions is discussed in the context of diverse human viruses, including influenza virus, SARS-CoV-2, herpes simplex virus, human papillomavirus, and human immunodeficiency virus.
Collapse
Affiliation(s)
- Younghyun Lim
- Department of Life Science, Chung-Ang University, Dongjak-gu, Republic of Korea
| | - Yong-Bin Cho
- Department of Life Science, Chung-Ang University, Dongjak-gu, Republic of Korea
| | - Young-Jin Seo
- Department of Life Science, Chung-Ang University, Dongjak-gu, Republic of Korea
| |
Collapse
|
13
|
Chen Q, Le X, Li Q, Liu S, Chen Z. Exploration of inhibitors targeting KIF18A with ploidy-specific lethality. Drug Discov Today 2024; 29:104142. [PMID: 39168405 DOI: 10.1016/j.drudis.2024.104142] [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: 06/03/2024] [Revised: 08/06/2024] [Accepted: 08/14/2024] [Indexed: 08/23/2024]
Abstract
Currently, various antimitotic inhibitors applied in tumor therapy. However, these inhibitors exhibit targeted toxicity to some extent. As a motor protein, kinesin family member 18A (KIF18A) is crucial to spindle formation and is associated with tumors exhibiting ploidy-specific characteristics such as chromosomal aneuploidy, whole-genome doubling (WGD), and chromosomal instability (CIN). Differing from traditional antimitotic targets, KIF18A exhibits tumor-specific selectivity. The functional loss or attenuation of KIF18A results in vulnerability of tumor cells with ploidy-specific characteristics, with lesser effects on diploid cells. Research on inhibitors targeting KIF18A with ploidy-specific lethality holds significant importance. This review provides a brief overview of the regulatory mechanisms of the ploidy-specific lethality target KIF18A and the research advancements in its inhibitors, aiming to facilitate the development of KIF18A inhibitors.
Collapse
Affiliation(s)
- Qingsong Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, Hunan, China
| | - Xiangyang Le
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, Hunan, China
| | - Qianbin Li
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, Hunan, China
| | - Suyou Liu
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, Hunan, China
| | - Zhuo Chen
- Department of Medicinal Chemistry, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, Hunan, China; Hunan Key Laboratory of Small Molecules for Diagnosis and Treatment of Chronic Disease, Changsha 410013, Hunan, China; Hunan Key Laboratory of Organ Fibrosis, Changsha 410013, Hunan, China.
| |
Collapse
|
14
|
Wu T, Luo Y, Zhang M, Chen B, Du X, Gu H, Xie S, Pan Z, Yu R, Hai R, Niu X, Hao G, Jin L, Shi J, Sun X, Kuang Y, Li W, Sang Q, Wang L. Mechanisms of minor pole-mediated spindle bipolarization in human oocytes. Science 2024; 385:eado1022. [PMID: 39172836 DOI: 10.1126/science.ado1022] [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: 01/18/2024] [Revised: 04/29/2024] [Accepted: 07/02/2024] [Indexed: 08/24/2024]
Abstract
Spindle bipolarization, the process of a microtubule mass transforming into a bipolar spindle, is a prerequisite for accurate chromosome segregation. In contrast to mitotic cells, the process and mechanism of spindle bipolarization in human oocytes remains unclear. Using high-resolution imaging in more than 1800 human oocytes, we revealed a typical state of multipolar intermediates that form during spindle bipolarization and elucidated the mechanism underlying this process. We found that the minor poles formed in multiple kinetochore clusters contribute to the generation of multipolar intermediates. We further determined the essential roles of HAUS6, KIF11, and KIF18A in spindle bipolarization and identified mutations in these genes in infertile patients characterized by oocyte or embryo defects. These results provide insights into the physiological and pathological mechanisms of spindle bipolarization in human oocytes.
Collapse
Affiliation(s)
- Tianyu Wu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Yuxi Luo
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Meiling Zhang
- Center for Reproductive Medicine and Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Biaobang Chen
- NHC Key Lab of Reproduction Regulation, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Fudan University, Shanghai 200032, China
| | - Xingzhu Du
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Hao Gu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Siyuan Xie
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Zhiqi Pan
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Ran Yu
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Ruiqi Hai
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Xiangli Niu
- Reproductive Hospital of Guangxi Zhuang Autonomous Region, Nanning 530029, China
| | - Guimin Hao
- Hebei Clinical Research Center for Birth Defects, Department of Reproductive Medicine, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, China
| | - Liping Jin
- Laboratory for Reproductive Immunology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Juanzi Shi
- Assisted Reproduction Center, Northwest Women's and Children's Hospital, Xi'an 710003, China
| | - Xiaoxi Sun
- Shanghai JIAI Genetics and IVF Institute, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200011, China
| | - Yanping Kuang
- Department of Assisted Reproduction, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Wen Li
- Center for Reproductive Medicine and Fertility Preservation Program, International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200030, China
| | - Qing Sang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| | - Lei Wang
- Institute of Pediatrics, Children's Hospital of Fudan University, State Key Laboratory of Genetic Engineering, Institutes of Biomedical Sciences, Shanghai Key Laboratory of Medical Epigenetics, Fudan University, Shanghai 200032, China
| |
Collapse
|
15
|
Thomas A, Meraldi P. Centrosome age breaks spindle size symmetry even in cells thought to divide symmetrically. J Cell Biol 2024; 223:e202311153. [PMID: 39012627 PMCID: PMC11252449 DOI: 10.1083/jcb.202311153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/14/2024] [Accepted: 05/03/2024] [Indexed: 07/17/2024] Open
Abstract
Centrosomes are the main microtubule-organizing centers in animal cells. Due to the semiconservative nature of centrosome duplication, the two centrosomes differ in age. In asymmetric stem cell divisions, centrosome age can induce an asymmetry in half-spindle lengths. However, whether centrosome age affects the symmetry of the two half-spindles in tissue culture cells thought to divide symmetrically is unknown. Here, we show that in human epithelial and fibroblastic cell lines centrosome age imposes a mild spindle asymmetry that leads to asymmetric cell daughter sizes. At the mechanistic level, we show that this asymmetry depends on a cenexin-bound pool of the mitotic kinase Plk1, which favors the preferential accumulation on old centrosomes of the microtubule nucleation-organizing proteins pericentrin, γ-tubulin, and Cdk5Rap2, and microtubule regulators TPX2 and ch-TOG. Consistently, we find that old centrosomes have a higher microtubule nucleation capacity. We postulate that centrosome age breaks spindle size symmetry via microtubule nucleation even in cells thought to divide symmetrically.
Collapse
Affiliation(s)
- Alexandre Thomas
- 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
| |
Collapse
|
16
|
Biswas L, Tyc KM, Aboelenain M, Sun S, Dundović I, Vukušić K, Liu J, Guo V, Xu M, Scott RT, Tao X, Tolić IM, Xing J, Schindler K. Maternal genetic variants in kinesin motor domains prematurely increase egg aneuploidy. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.07.04.24309950. [PMID: 39006445 PMCID: PMC11245073 DOI: 10.1101/2024.07.04.24309950] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The female reproductive lifespan depends on egg quality, particularly euploidy. Mistakes in meiosis leading to egg aneuploidy are common, but the genetic landscape causing this is not well understood due to limited phenotypic data. We identify genetic determinants of reproductive aging via egg aneuploidy using a biobank of maternal exomes linked with maternal age and embryonic aneuploidy data. We found 404 genes with variants enriched in individuals with high egg aneuploidy rates and implicate kinesin protein family genes in aneuploidy risk. Experimental perturbations showed that motor domain variants in these genes increase aneuploidy in mouse oocytes. A knock-in mouse model validated that a specific variant in kinesin KIF18A accelerates reproductive aging and diminishes fertility. These findings suggest potential non-invasive biomarkers for egg quality, aiding personalized fertility medicine. One sentence summary The study identifies novel genetic determinants of reproductive aging linked to egg aneuploidy by analyzing maternal exomes and demonstrates that variants in kinesin genes, specifically KIF18A , contribute to increased aneuploidy and accelerated reproductive aging, offering potential for personalized fertility medicine.
Collapse
|
17
|
Kufleitner M, Haiber LM, Li S, Surendran H, Mayer TU, Zumbusch A, Wittmann V. Next-Generation Metabolic Glycosylation Reporters Enable Detection of Protein O-GlcNAcylation in Living Cells without S-Glyco Modification. Angew Chem Int Ed Engl 2024; 63:e202320247. [PMID: 38501674 DOI: 10.1002/anie.202320247] [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: 12/30/2023] [Revised: 03/08/2024] [Accepted: 03/18/2024] [Indexed: 03/20/2024]
Abstract
Protein O-GlcNAcylation is a ubiquitous posttranslational modification of cytosolic and nuclear proteins involved in numerous fundamental regulation processes. Investigation of O-GlcNAcylation by metabolic glycoengineering (MGE) has been carried out for two decades with peracetylated N-acetylglucosamine (GlcNAc) and N-acetylgalactosamine derivatives modified with varying reporter groups. Recently, it has been shown that these derivatives can result in non-specific protein labeling termed S-glyco modification. Here, we report norbornene-modified GlcNAc derivatives with a protected phosphate at the anomeric position and their application in MGE. These derivatives overcome two limitations of previously used O-GlcNAc reporters. They do not lead to detectable S-glyco modification, and they efficiently react in the inverse-electron-demand Diels-Alder (IEDDA) reaction, which can be carried out even within living cells. Using a derivative with an S-acetyl-2-thioethyl-protected phosphate, we demonstrate the protein-specific detection of O-GlcNAcylation of several proteins and the protein-specific imaging of O-GlcNAcylation inside living cells by Förster resonance energy transfer (FRET) visualized by confocal fluorescence lifetime imaging microscopy (FLIM).
Collapse
Affiliation(s)
- Markus Kufleitner
- Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Lisa Maria Haiber
- Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Shuang Li
- Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Harsha Surendran
- Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Thomas U Mayer
- Department of Biology, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Andreas Zumbusch
- Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| | - Valentin Wittmann
- Department of Chemistry, University of Konstanz, Universitätsstraße 10, 78464, Konstanz, Germany
| |
Collapse
|
18
|
Conway PJ, Dao J, Kovalskyy D, Mahadevan D, Dray E. Polyploidy in Cancer: Causal Mechanisms, Cancer-Specific Consequences, and Emerging Treatments. Mol Cancer Ther 2024; 23:638-647. [PMID: 38315992 PMCID: PMC11174144 DOI: 10.1158/1535-7163.mct-23-0578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/19/2023] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Drug resistance is the major determinant for metastatic disease and fatalities, across all cancers. Depending on the tissue of origin and the therapeutic course, a variety of biological mechanisms can support and sustain drug resistance. Although genetic mutations and gene silencing through epigenetic mechanisms are major culprits in targeted therapy, drug efflux and polyploidization are more global mechanisms that prevail in a broad range of pathologies, in response to a variety of treatments. There is an unmet need to identify patients at risk for polyploidy, understand the mechanisms underlying polyploidization, and to develop strategies to predict, limit, and reverse polyploidy thus enhancing efficacy of standard-of-care therapy that improve better outcomes. This literature review provides an overview of polyploidy in cancer and offers perspective on patient monitoring and actionable therapy.
Collapse
Affiliation(s)
- Patrick J Conway
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Department of Molecular Immunology & Microbiology, University of Texas Health San Antonio, San Antonio, Texas
| | - Jonathan Dao
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
| | - Dmytro Kovalskyy
- Greehey Children's Cancer Research Institute, University of Texas Health San Antonio, San Antonio, Texas
| | - Daruka Mahadevan
- Mays Cancer Center, University of Texas Health San Antonio, San Antonio, Texas
- Department of Molecular Immunology & Microbiology, University of Texas Health San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| | - Eloise Dray
- Long School of Medicine, University of Texas Health San Antonio, San Antonio, Texas
- Department of Biochemistry and Structural Biology, University of Texas Health San Antonio, San Antonio, Texas
| |
Collapse
|
19
|
Perez-Bertoldi JM, Zhao Y, Thawani A, Yildiz A, Nogales E. Molecular interplay between HURP and Kif18A in mitotic spindle regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.11.589088. [PMID: 38645125 PMCID: PMC11030443 DOI: 10.1101/2024.04.11.589088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2024]
Abstract
During mitosis, microtubule dynamics are regulated to ensure proper alignment and segregation of chromosomes. The dynamics of kinetochore-attached microtubules are regulated by hepatoma-upregulated protein (HURP) and the mitotic kinesin-8 Kif18A, but the underlying mechanism remains elusive. Using single-molecule imaging in vitro , we demonstrate that Kif18A motility is regulated by HURP. While sparse decoration of HURP activates the motor, higher concentrations hinder processive motility. To shed light on this behavior, we determined the binding mode of HURP to microtubules using Cryo-EM. The structure reveals that one HURP motif spans laterally across β-tubulin, while a second motif binds between adjacent protofilaments. HURP partially overlaps with the microtubule-binding site of the Kif18A motor domain, indicating that excess HURP inhibits Kif18A motility by steric exclusion. We also observed that HURP and Kif18A function together to suppress dynamics of the microtubule plus-end, providing a mechanistic basis for how they collectively serve in spindle length control.
Collapse
|
20
|
Gliech CR, Yeow ZY, Tapias-Gomez D, Yang Y, Huang Z, Tijhuis AE, Spierings DC, Foijer F, Chung G, Tamayo N, Bahrami-Nejad Z, Collins P, Nguyen TT, Plata Stapper A, Hughes PE, Payton M, Holland AJ. Weakened APC/C activity at mitotic exit drives cancer vulnerability to KIF18A inhibition. EMBO J 2024; 43:666-694. [PMID: 38279026 PMCID: PMC10907621 DOI: 10.1038/s44318-024-00031-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 12/23/2023] [Accepted: 01/02/2024] [Indexed: 01/28/2024] Open
Abstract
The efficacy of current antimitotic cancer drugs is limited by toxicity in highly proliferative healthy tissues. A cancer-specific dependency on the microtubule motor protein KIF18A therefore makes it an attractive therapeutic target. Not all cancers require KIF18A, however, and the determinants underlying this distinction remain unclear. Here, we show that KIF18A inhibition drives a modest and widespread increase in spindle assembly checkpoint (SAC) signaling from kinetochores which can result in lethal mitotic delays. Whether cells arrest in mitosis depends on the robustness of the metaphase-to-anaphase transition, and cells predisposed with weak basal anaphase-promoting complex/cyclosome (APC/C) activity and/or persistent SAC signaling through metaphase are uniquely sensitive to KIF18A inhibition. KIF18A-dependent cancer cells exhibit hallmarks of this SAC:APC/C imbalance, including a long metaphase-to-anaphase transition, and slow mitosis overall. Together, our data reveal vulnerabilities in the cell division apparatus of cancer cells that can be exploited for therapeutic benefit.
Collapse
Affiliation(s)
- Colin R Gliech
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Zhong Y Yeow
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Daniel Tapias-Gomez
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Yuchen Yang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Zhaoyu Huang
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Andréa E Tijhuis
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, AV, 9713, The Netherlands
| | - Diana Cj Spierings
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, AV, 9713, The Netherlands
| | - Floris Foijer
- European Research Institute for the Biology of Ageing, University of Groningen, University Medical Center Groningen, Groningen, AV, 9713, The Netherlands
| | - Grace Chung
- Oncology Research, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - Nuria Tamayo
- Medicinal Chemistry, Amgen Research, Thousand Oaks, CA, 91320, USA
| | | | - Patrick Collins
- Genome Analysis Unit, Amgen Research, South San Francisco, CA, 94084, USA
| | - Thong T Nguyen
- Genome Analysis Unit, Amgen Research, South San Francisco, CA, 94084, USA
| | - Andres Plata Stapper
- Center for Research Acceleration by Digital Innovation, Amgen Research, South San Francisco, CA, 94084, USA
| | - Paul E Hughes
- Oncology Research, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - Marc Payton
- Oncology Research, Amgen Research, Thousand Oaks, CA, 91320, USA
| | - Andrew J Holland
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
21
|
Schutt KL, Queen KA, Fisher K, Budington O, Mao W, Liu W, Gu X, Xiao Y, Aswad F, Joseph J, Stumpff J. Identification of the KIF18A alpha-4 helix as a therapeutic target for chromosomally unstable tumor cells. Front Mol Biosci 2024; 11:1328077. [PMID: 38410188 PMCID: PMC10896213 DOI: 10.3389/fmolb.2024.1328077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 01/08/2024] [Indexed: 02/28/2024] Open
Abstract
Background: The mitotic kinesin, KIF18A, is required for proliferation of cancer cells that exhibit chromosome instability (CIN), implicating it as a promising target for treatment of a subset of aggressive tumor types. Determining regions of the KIF18A protein to target for inhibition will be important for the design and optimization of effective small molecule inhibitors. Methods: In this study, we used cultured cell models to investigate the effects of mutating S284 within the alpha-4 helix of KIF18A, which was previously identified as a phosphorylated residue. Results: Mutations in S284 cause relocalization of KIF18A from the plus-ends of spindle microtubules to the spindle poles. Furthermore, KIF18A S284 mutants display loss of KIF18A function and fail to support proliferation in CIN tumor cells. Interestingly, similar effects on KIF18A localization and function were seen after treatment of CIN cells with KIF18A inhibitory compounds that are predicted to interact with residues within the alpha-4 helix. Conclusion: These data implicate the KIF18A alpha-4 helix as an effective target for inhibition and demonstrate that small molecules targeting KIF18A selectively limit CIN tumor cell proliferation and result in phenotypically similar effects on mitosis at the single cell level compared to genetic perturbations.
Collapse
Affiliation(s)
- Katherine L Schutt
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, United States
| | - Katelyn A Queen
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, United States
| | - Kira Fisher
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, United States
| | - Olivia Budington
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, United States
| | | | - Wei Liu
- Apeiron Therapeutics, Shanghai, China
| | | | | | - Fred Aswad
- Apeiron Therapeutics, Burlingame, CA, United States
| | - James Joseph
- Apeiron Therapeutics, Burlingame, CA, United States
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, United States
| |
Collapse
|
22
|
Li L, Ran J. Regulation of ciliary homeostasis by intraflagellar transport-independent kinesins. Cell Death Dis 2024; 15:47. [PMID: 38218748 PMCID: PMC10787775 DOI: 10.1038/s41419-024-06428-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/15/2024]
Abstract
Cilia are highly conserved eukaryotic organelles that protrude from the cell surface and are involved in sensory perception, motility, and signaling. Their proper assembly and function rely on the bidirectional intraflagellar transport (IFT) system, which involves motor proteins, including antegrade kinesins and retrograde dynein. Although the role of IFT-mediated transport in cilia has been extensively studied, recent research has highlighted the contribution of IFT-independent kinesins in ciliary processes. The coordinated activities and interplay between IFT kinesins and IFT-independent kinesins are crucial for maintaining ciliary homeostasis. In this comprehensive review, we aim to delve into the specific contributions and mechanisms of action of the IFT-independent kinesins in cilia. By shedding light on their involvement, we hope to gain a more holistic perspective on ciliogenesis and ciliopathies.
Collapse
Affiliation(s)
- Lin Li
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China
| | - Jie Ran
- Center for Cell Structure and Function, Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
23
|
Adegoke A, Ribeiro JMC, Smith R, Karim S. Tick innate immune responses to hematophagy and Ehrlichia infection at single-cell resolution. Front Immunol 2024; 14:1305976. [PMID: 38274813 PMCID: PMC10808623 DOI: 10.3389/fimmu.2023.1305976] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction Ticks rely on robust cellular and humoral responses to control microbial infection. However, several aspects of the tick's innate immune system remain uncharacterized, most notably that of the immune cells (called hemocytes), which are known to play a significant role in cellular and humoral responses. Despite the importance of hemocytes in regulating microbial infection, our understanding of their basic biology and molecular mechanisms remains limited. Therefore, we believe that a more detailed understanding of the role of hemocytes in the interactions between ticks and tick-borne microbes is crucial to illuminating their function in vector competence and to help identify novel targets for developing new strategies to block tick-borne pathogen transmission. Methods This study examined hemocytes from the lone star tick (Amblyomma americanum) at the transcriptomic level using the 10X genomics single-cell RNA sequencing platform to analyze hemocyte populations from unfed, partially blood-fed, and Ehrlichia chaffeensis-infected ticks. The functional role of differentially expressed hemocyte markers in hemocyte proliferation and Ehrlichia dissemination was determined using an RNA interference approach. Results and discussion Our data exhibit the identification of fourteen distinct hemocyte populations. Our results uncover seven distinct lineages present in uninfected and Ehrlichia-infected hemocyte clusters. The functional characterization of hemocytin, cystatin, fibronectin, and lipocalin demonstrate their role in hemocyte population changes, proliferation, and Ehrlichia dissemination. Conclusion Our results uncover the tick immune responses to Ehrlichia infection and hematophagy at a single-cell resolution. This work opens a new field of tick innate immunobiology to understand the role of hemocytes, particularly in response to prolonged blood-feeding (hematophagy), and tick-microbial interactions.
Collapse
Affiliation(s)
- Abdulsalam Adegoke
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| | - Jose M. C. Ribeiro
- Vector Biology Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Ryan C. Smith
- Department of Plant Pathology, Entomology, and Microbiology, Iowa State University, Ames, IA, United States
| | - Shahid Karim
- School of Biological, Environmental, and Earth Sciences, The University of Southern Mississippi, Hattiesburg, MS, United States
| |
Collapse
|
24
|
Li T, Gachet Y, Tournier S. MAARS Software for Automatic and Quantitative Analysis of Mitotic Progression. Methods Mol Biol 2024; 2740:275-293. [PMID: 38393482 DOI: 10.1007/978-1-0716-3557-5_17] [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] [Indexed: 02/25/2024]
Abstract
In this chapter, we describe a software called MAARS (Mitotic Analysis And Recording System) that enables automatic and quantitative analysis of mitotic progression on an open-source platform. This computer-assisted analysis of cell division allows the unbiased acquisition of multiple parameters such as cell shape or size, metaphase or anaphase delays, as well as various mitotic abnormalities. This chapter describes the power of such an expert system to highlight the complexity of the mechanisms required to prevent mitotic chromosome segregation errors, leading to aneuploidy.
Collapse
Affiliation(s)
- Tong Li
- MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse Cedex, France
- Wellcome Sanger Institute, Cambridge, UK
| | - Yannick Gachet
- MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse Cedex, France.
| | - Sylvie Tournier
- MCD, Centre de Biologie Intégrative, Université de Toulouse, CNRS, UPS, Toulouse Cedex, France.
| |
Collapse
|
25
|
Queen KA, Cario A, Berger CL, Stumpff J. Modification of the neck-linker of KIF18A alters Microtubule subpopulation preference. Mol Biol Cell 2024; 35:ar3. [PMID: 37903223 PMCID: PMC10881168 DOI: 10.1091/mbc.e23-05-0167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/01/2023] Open
Abstract
Kinesins support many diverse cellular processes, including facilitating cell division through mechanical regulation of the mitotic spindle. However, how kinesin activity is controlled to facilitate this process is not well understood. Interestingly, posttranslational modifications have been identified within the enzymatic region of all 45 mammalian kinesins, but the significance of these modifications has gone largely unexplored. Given the critical role of the enzymatic region in facilitating nucleotide and microtubule binding, it may serve as a primary site for kinesin regulation. Consistent with this idea, a phosphomimetic mutation at S357 in the neck-linker of KIF18A alters the localization of KIF18A within the spindle from kinetochore microtubules to nonkinetochore microtubules at the periphery of the spindle. Changes in localization of KIF18A-S357D are accompanied by defects in mitotic spindle positioning and the ability to promote mitotic progression. This altered localization pattern is mimicked by a shortened neck-linker mutant, suggesting that KIF18A-S357D may cause the motor to adopt a shortened neck-linker-like state that decreases KIF18A accumulation at the plus-ends of kinetochore microtubules. These findings demonstrate that posttranslational modifications in the enzymatic region of kinesins could be important for biasing their localization to particular microtubule subpopulations.
Collapse
Affiliation(s)
- Katelyn A. Queen
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05401
| | - Alisa Cario
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05401
| | - Christopher L. Berger
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05401
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05401
| |
Collapse
|
26
|
Payton M, Belmontes B, Hanestad K, Moriguchi J, Chen K, McCarter JD, Chung G, Ninniri MS, Sun J, Manoukian R, Chambers S, Ho SM, Kurzeja RJM, Edson KZ, Dahal UP, Wu T, Wannberg S, Beltran PJ, Canon J, Boghossian AS, Rees MG, Ronan MM, Roth JA, Minocherhomji S, Bourbeau MP, Allen JR, Coxon A, Tamayo NA, Hughes PE. Small-molecule inhibition of kinesin KIF18A reveals a mitotic vulnerability enriched in chromosomally unstable cancers. NATURE CANCER 2024; 5:66-84. [PMID: 38151625 PMCID: PMC10824666 DOI: 10.1038/s43018-023-00699-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 11/30/2023] [Indexed: 12/29/2023]
Abstract
Chromosomal instability (CIN) is a hallmark of cancer, caused by persistent errors in chromosome segregation during mitosis. Aggressive cancers like high-grade serous ovarian cancer (HGSOC) and triple-negative breast cancer (TNBC) have a high frequency of CIN and TP53 mutations. Here, we show that inhibitors of the KIF18A motor protein activate the mitotic checkpoint and selectively kill chromosomally unstable cancer cells. Sensitivity to KIF18A inhibition is enriched in TP53-mutant HGSOC and TNBC cell lines with CIN features, including in a subset of CCNE1-amplified, CDK4-CDK6-inhibitor-resistant and BRCA1-altered cell line models. Our KIF18A inhibitors have minimal detrimental effects on human bone marrow cells in culture, distinct from other anti-mitotic agents. In mice, inhibition of KIF18A leads to robust anti-cancer effects with tumor regression observed in human HGSOC and TNBC models at well-tolerated doses. Collectively, our results provide a rational therapeutic strategy for selective targeting of CIN cancers via KIF18A inhibition.
Collapse
Affiliation(s)
- Marc Payton
- Oncology Research, Amgen Research, Thousand Oaks, CA, USA.
| | | | - Kelly Hanestad
- Oncology Research, Amgen Research, Thousand Oaks, CA, USA
| | - Jodi Moriguchi
- Oncology Research, Amgen Research, Thousand Oaks, CA, USA
| | - Kui Chen
- Lead Discovery and Characterization, Amgen Research, Thousand Oaks, CA, USA
| | - John D McCarter
- Lead Discovery and Characterization, Amgen Research, Thousand Oaks, CA, USA
| | - Grace Chung
- Oncology Research, Amgen Research, Thousand Oaks, CA, USA
| | | | - Jan Sun
- Oncology Research, Amgen Research, Thousand Oaks, CA, USA
| | | | | | - Seok-Man Ho
- Research Biomics, Amgen Research, San Francisco, CA, USA
| | | | | | | | - Tian Wu
- Pre-Pivotal Drug Product, Amgen Process Development, Thousand Oaks, CA, USA
| | | | | | - Jude Canon
- Oncology Research, Amgen Research, Thousand Oaks, CA, USA
| | | | | | | | | | - Sheroy Minocherhomji
- Translational Safety and Bioanalytical Sciences, Amgen Research, Thousand Oaks, CA, USA
| | | | | | - Angela Coxon
- Oncology Research, Amgen Research, Thousand Oaks, CA, USA
| | - Nuria A Tamayo
- Medicinal Chemistry, Amgen Research, Thousand Oaks, CA, USA
| | - Paul E Hughes
- Oncology Research, Amgen Research, Thousand Oaks, CA, USA
| |
Collapse
|
27
|
Mihajlović AI, Byers C, Reinholdt L, FitzHarris G. Spindle assembly checkpoint insensitivity allows meiosis-II despite chromosomal defects in aged eggs. EMBO Rep 2023; 24:e57227. [PMID: 37795949 PMCID: PMC10626445 DOI: 10.15252/embr.202357227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 09/07/2023] [Accepted: 09/19/2023] [Indexed: 10/06/2023] Open
Abstract
Chromosome segregation errors in mammalian oocyte meiosis lead to developmentally compromised aneuploid embryos and become more common with advancing maternal age. Known contributors include age-related chromosome cohesion loss and spindle assembly checkpoint (SAC) fallibility in meiosis-I. But how effective the SAC is in meiosis-II and how this might contribute to age-related aneuploidy is unknown. Here, we developed genetic and pharmacological approaches to directly address the function of the SAC in meiosis-II. We show that the SAC is insensitive in meiosis-II oocytes and that as a result misaligned chromosomes are randomly segregated. Whilst SAC ineffectiveness in meiosis-II is not age-related, it becomes most prejudicial in oocytes from older females because chromosomes that prematurely separate by age-related cohesion loss become misaligned in meiosis-II. We show that in the absence of a robust SAC in meiosis-II these age-related misaligned chromatids are missegregated and lead to aneuploidy. Our data demonstrate that the SAC fails to prevent cell division in the presence of misaligned chromosomes in oocyte meiosis-II, which explains how age-related cohesion loss can give rise to aneuploid embryos.
Collapse
Affiliation(s)
| | - Candice Byers
- The Institute for Experiential AI, Roux InstituteNortheastern UniversityPortlandMEUSA
| | | | | |
Collapse
|
28
|
Kumar C, Mylavarapu SVS. Nucleolin is required for multiple centrosome-associated functions in early vertebrate mitosis. Chromosoma 2023; 132:305-315. [PMID: 37615728 DOI: 10.1007/s00412-023-00808-4] [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: 02/07/2023] [Revised: 06/10/2023] [Accepted: 08/11/2023] [Indexed: 08/25/2023]
Abstract
Nucleolin is a multifunctional RNA-binding protein that resides predominantly not only in the nucleolus, but also in multiple other subcellular pools in the cytoplasm in mammalian cells, and is best known for its roles in ribosome biogenesis, RNA stability, and translation. During early mitosis, nucleolin is required for equatorial mitotic chromosome alignment prior to metaphase. Using high resolution fluorescence imaging, we reveal that nucleolin is required for multiple centrosome-associated functions at the G2-prophase boundary. Nucleolin depletion led to dissociation of the centrosomes from the G2 nuclear envelope, a delay in the onset of nuclear envelope breakdown, reduced inter-centrosome separation, and longer metaphase spindles. Our results reveal novel roles for nucleolin in early mammalian mitosis, establishing multiple important functions for nucleolin during mammalian cell division.
Collapse
Affiliation(s)
- Chandan Kumar
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, -121001, India
| | - Sivaram V S Mylavarapu
- Laboratory of Cellular Dynamics, Regional Centre for Biotechnology, NCR Biotech Science Cluster, 3rd Milestone, Faridabad-Gurgaon Expressway, Faridabad, Haryana, -121001, India.
| |
Collapse
|
29
|
Schutt K, Queen KA, Fisher K, Budington O, Mao W, Liu W, Xiao Y, Aswad F, Joseph J, Stumpff J. Identification of the KIF18A alpha-4 helix as a therapeutic target for chromosomally unstable tumor cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.16.562576. [PMID: 37905069 PMCID: PMC10614886 DOI: 10.1101/2023.10.16.562576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
The mitotic kinesin, KIF18A, is required for proliferation of cancer cells that exhibit chromosome instability (CIN), implicating it as a promising target for treatment of a subset of aggressive tumor types. Determining regions of the KIF18A protein to target for inhibition will be important for the design and optimization of effective small molecule inhibitors. In this study, we investigated the effects of mutating S284 within the alpha-4 helix of KIF18A, which was previously identified as a phosphorylated residue. Mutations in S284 cause relocalization of KIF18A from the plus-ends of spindle microtubules to the spindle poles. Furthermore, KIF18A S284 mutants display loss of KIF18A function and fail to support proliferation in CIN tumor cells. Interestingly, similar effects on KIF18A localization and function were seen after treatment of CIN cells with KIF18A inhibitory compounds that are predicted to interact with residues within the alpha-4 helix. These data implicate the KIF18A alpha-4 helix as an effective target for inhibition and demonstrate that small molecules targeting KIF18A selectively limit CIN tumor cell proliferation and result in phenotypically similar effects on mitosis at the single cell level compared to genetic perturbations.
Collapse
Affiliation(s)
- Katherine Schutt
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT
| | - Katelyn A Queen
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT
| | - Kira Fisher
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT
| | - Olivia Budington
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT
| | | | - Wei Liu
- Apeiron Therapeutics, Shanghai, CN
| | | | | | | | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT
| |
Collapse
|
30
|
Shah S, Mittal P, Kumar D, Mittal A, Ghosh SK. Evidence of kinesin motors involved in stable kinetochore assembly during early meiosis. Mol Biol Cell 2023; 34:ar107. [PMID: 37556230 PMCID: PMC10559306 DOI: 10.1091/mbc.e22-12-0569] [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: 01/03/2023] [Revised: 07/27/2023] [Accepted: 08/02/2023] [Indexed: 08/11/2023] Open
Abstract
During mitosis, the budding yeast, kinetochores remain attached to microtubules, except for a brief period during S phase. Sister-kinetochores separate into two clusters (bilobed organization) upon stable end-on attachment to microtubules emanating from opposite spindle poles. However, in meiosis, the outer kinetochore protein (Ndc80) reassembles at the centromeres much later after prophase I, establishing new kinetochore-microtubule attachments. Perhaps due to this, despite homolog bi-orientation, we observed that the Ndc80 are linearly dispersed between spindle poles during metaphase I of meiosis. The presence of end-on attachment marker Dam1 as a cluster near each pole suggests one of the other possibilities that the pole-proximal and pole-distal kinetochores are attached end-on and laterally to the microtubules, respectively. Colocalization studies of kinetochores and kinesin motors suggest that budding yeast kinesin 5, Cin8, and Kip1 perhaps localize to the end-on attached kinetochores while kinesin 8 and Kip3 resides at all the kinetochores. Our findings, including kinesin 5 and Ndc80 coappearance after prophase I and reduced Ndc80 levels in cin8 null mutant, suggest that kinesin motors are crucial for kinetochore reassembly and stability during early meiosis. Thus, this work reports yet another meiosis specific function of kinesin motors.
Collapse
Affiliation(s)
- Seema Shah
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Priyanka Mittal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Deepanshu Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Anjani Mittal
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| | - Santanu K. Ghosh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai-400076, India
| |
Collapse
|
31
|
Liu X, Liu H, Gu N, Pei J, Lin X, Zhao W. Preeclampsia promotes autism in offspring via maternal inflammation and fetal NFκB signaling. Life Sci Alliance 2023; 6:e202301957. [PMID: 37290815 PMCID: PMC10250690 DOI: 10.26508/lsa.202301957] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/28/2023] [Accepted: 05/30/2023] [Indexed: 06/10/2023] Open
Abstract
Preeclampsia (PE) is a risk factor for autism spectrum disorder (ASD) in offspring. However, the exact mechanisms underlying the impact of PE on progeny ASD are not fully understood, which hinders the development of effective therapeutic approaches. This study shows the offspring born to a PE mouse model treated by Nω-nitro-L-arginine methyl ester (L-NAME) exhibit ASD-like phenotypes, including neurodevelopment deficiency and behavioral abnormalities. Transcriptomic analysis of the embryonic cortex and adult offspring hippocampus suggested the expression of ASD-related genes was dramatically changed. Furthermore, the level of inflammatory cytokines TNFα in maternal serum and nuclear factor kappa B (NFκB) signaling in the fetal cortex were elevated. Importantly, TNFα neutralization during pregnancy enabled to ameliorate ASD-like phenotypes and restore the NFκB activation level in the offspring exposed to PE. Furthermore, TNFα/NFκB signaling axis, but not L-NAME, caused deficits in neuroprogenitor cell proliferation and synaptic development. These experiments demonstrate that offspring exposed to PE phenocopies ASD signatures reported in humans and indicate therapeutic targeting of TNFα decreases the likelihood of bearing children with ASD phenotypes from PE mothers.
Collapse
Affiliation(s)
- Xueyuan Liu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
| | - Haiyan Liu
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Nihao Gu
- International Peace Maternity & Child Health Hospital Affiliated to Shanghai Jiao Tong University School of Medicine and Shanghai Key Laboratory for Embryo-Feta Original Adult Disease, Shanghai Jiao Tong University, Shanghai, China
| | - Jiangnan Pei
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Xianhua Lin
- Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Wenlong Zhao
- Environmental and Occupational Health Science Institute, Rutgers University, Piscataway, NJ, USA
- International Peace Maternity & Child Health Hospital Affiliated to Shanghai Jiao Tong University School of Medicine and Shanghai Key Laboratory for Embryo-Feta Original Adult Disease, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
32
|
Normandin K, Coulombe-Huntington J, St-Denis C, Bernard A, Bourouh M, Bertomeu T, Tyers M, Archambault V. Genetic enhancers of partial PLK1 inhibition reveal hypersensitivity to kinetochore perturbations. PLoS Genet 2023; 19:e1010903. [PMID: 37639469 PMCID: PMC10491399 DOI: 10.1371/journal.pgen.1010903] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 09/08/2023] [Accepted: 08/06/2023] [Indexed: 08/31/2023] Open
Abstract
Polo-like kinase 1 (PLK1) is a serine/threonine kinase required for mitosis and cytokinesis. As cancer cells are often hypersensitive to partial PLK1 inactivation, chemical inhibitors of PLK1 have been developed and tested in clinical trials. However, these small molecule inhibitors alone are not completely effective. PLK1 promotes numerous molecular and cellular events in the cell division cycle and it is unclear which of these events most crucially depend on PLK1 activity. We used a CRISPR-based genome-wide screening strategy to identify genes whose inactivation enhances cell proliferation defects upon partial chemical inhibition of PLK1. Genes identified encode proteins that are functionally linked to PLK1 in multiple ways, most notably factors that promote centromere and kinetochore function. Loss of the kinesin KIF18A or the outer kinetochore protein SKA1 in PLK1-compromised cells resulted in mitotic defects, activation of the spindle assembly checkpoint and nuclear reassembly defects. We also show that PLK1-dependent CENP-A loading at centromeres is extremely sensitive to partial PLK1 inhibition. Our results suggest that partial inhibition of PLK1 compromises the integrity and function of the centromere/kinetochore complex, rendering cells hypersensitive to different kinetochore perturbations. We propose that KIF18A is a promising target for combinatorial therapies with PLK1 inhibitors.
Collapse
Affiliation(s)
- Karine Normandin
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | | | - Corinne St-Denis
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Alexandre Bernard
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Mohammed Bourouh
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Thierry Bertomeu
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Département de médecine, Université de Montréal, Montréal, Canada
| | - Vincent Archambault
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Canada
- Département de biochimie et médecine moléculaire, Université de Montréal, Montréal, Canada
| |
Collapse
|
33
|
Chen X, Portran D, Widmer LA, Stangier MM, Czub MP, Liakopoulos D, Stelling J, Steinmetz MO, Barral Y. The motor domain of the kinesin Kip2 promotes microtubule polymerization at microtubule tips. J Cell Biol 2023; 222:214052. [PMID: 37093124 PMCID: PMC10130750 DOI: 10.1083/jcb.202110126] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/01/2023] [Accepted: 03/22/2023] [Indexed: 04/25/2023] Open
Abstract
Kinesins are microtubule-dependent motor proteins, some of which moonlight as microtubule polymerases, such as the yeast protein Kip2. Here, we show that the CLIP-170 ortholog Bik1 stabilizes Kip2 at microtubule ends where the motor domain of Kip2 promotes microtubule polymerization. Live-cell imaging and mathematical estimation of Kip2 dynamics reveal that disrupting the Kip2-Bik1 interaction aborts Kip2 dwelling at microtubule ends and abrogates its microtubule polymerization activity. Structural modeling and biochemical experiments identify a patch of positively charged residues that enables the motor domain to bind free tubulin dimers alternatively to the microtubule shaft. Neutralizing this patch abolished the ability of Kip2 to promote microtubule growth both in vivo and in vitro without affecting its ability to walk along microtubules. Our studies suggest that Kip2 utilizes Bik1 as a cofactor to track microtubule tips, where its motor domain then recruits free tubulin and catalyzes microtubule assembly.
Collapse
Affiliation(s)
- Xiuzhen Chen
- Institute of Biochemistry, Eidgenössische Technische Hochschule Zürich , Zurich, Switzerland
| | - Didier Portran
- CRBM, Université de Montpellier , CNRS, Montpellier, France
| | - Lukas A Widmer
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Marcel M Stangier
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Mateusz P Czub
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
| | - Dimitris Liakopoulos
- CRBM, Université de Montpellier , CNRS, Montpellier, France
- Laboratory of Biology, University of Ioannina, Faculty of Medicine, Ioannina, Greece
| | - Jörg Stelling
- Department of Biosystems Science and Engineering, Eidgenössische Technische Hochschule Zürich, and Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Michel O Steinmetz
- Department of Biology and Chemistry, Laboratory of Biomolecular Research, Paul Scherrer Institute, Villigen, Switzerland
- University of Basel, Biozentrum , Basel, Switzerland
| | - Yves Barral
- Institute of Biochemistry, Eidgenössische Technische Hochschule Zürich , Zurich, Switzerland
| |
Collapse
|
34
|
Velasco CD, Santarella-Mellwig R, Schorb M, Gao L, Thorn-Seshold O, Llobet A. Microtubule depolymerization contributes to spontaneous neurotransmitter release in vitro. Commun Biol 2023; 6:488. [PMID: 37147475 PMCID: PMC10163034 DOI: 10.1038/s42003-023-04779-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 03/29/2023] [Indexed: 05/07/2023] Open
Abstract
Microtubules are key to multiple neuronal functions involving the transport of organelles, however, their relationship to neurotransmitter release is still unresolved. Here, we show that microtubules present in the presynaptic compartment of cholinergic autaptic synapses are dynamic. To investigate how the balance between microtubule growth and shrinkage affects neurotransmission we induced synchronous microtubule depolymerization by photoactivation of the chemical inhibitor SBTub3. The consequence was an increase in spontaneous neurotransmitter release. An analogous effect was obtained by dialyzing the cytosol with Kif18A, a plus-end-directed kinesin with microtubule depolymerizing activity. Kif18A also inhibited the refilling of the readily releasable pool of synaptic vesicles during high frequency stimulation. The action of Kif18A was associated to one order of magnitude increases in the numbers of exo-endocytic pits and endosomes present in the presynaptic terminal. An enhancement of spontaneous neurotransmitter release was also observed when neurons were dialyzed with stathmin-1, a protein with a widespread presence in the nervous system that induces microtubule depolymerization. Taken together, these results support that microtubules restrict spontaneous neurotransmitter release as well as promote the replenishment of the readily releasable pool of synaptic vesicles.
Collapse
Affiliation(s)
- Cecilia D Velasco
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain
- Bellvitge Biomedical Research Institute (IDIBELL), 08907, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Rachel Santarella-Mellwig
- Electron Microscopy Core Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Martin Schorb
- Electron Microscopy Core Facility, European Molecular Biology Laboratory (EMBL), Meyerhofstrasse 1, 69117, Heidelberg, Germany
| | - Li Gao
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Munich, 81377, Germany
| | - Oliver Thorn-Seshold
- Department of Pharmacy, Ludwig-Maximilians University of Munich, Munich, 81377, Germany
| | - Artur Llobet
- Laboratory of Neurobiology, Department of Pathology and Experimental Therapy, Institute of Neurosciences, University of Barcelona, 08907, L'Hospitalet de Llobregat, Barcelona, Spain.
- Bellvitge Biomedical Research Institute (IDIBELL), 08907, L'Hospitalet de Llobregat, Barcelona, Spain.
| |
Collapse
|
35
|
Queen KA, Cario A, Berger CL, Stumpff J. Modification of the Neck Linker of KIF18A Alters Microtubule Subpopulation Preference. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.02.539080. [PMID: 37205510 PMCID: PMC10187232 DOI: 10.1101/2023.05.02.539080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Kinesins support many diverse cellular processes, including facilitating cell division through mechanical regulation of the mitotic spindle. However, how kinesin activity is controlled to facilitate this process is not well understood. Interestingly, post-translational modifications have been identified within the enzymatic region of all 45 mammalian kinesins, but the significance of these modifications has gone largely unexplored. Given the critical role of the enzymatic region in facilitating nucleotide and microtubule binding, it may serve as a primary site for kinesin regulation. Consistent with this idea, a phosphomimetic mutation at S357 in the neck-linker of KIF18A alters the localization of KIF18A within the spindle from kinetochore microtubules to peripheral microtubules. Changes in localization of KIF18A-S357D are accompanied by defects in mitotic spindle positioning and the ability to promote mitotic progression. This altered localization pattern is mimicked by a shortened neck-linker mutant, suggesting that KIF18A-S357D may cause the motor to adopt a shortened neck-linker like state that prevents KIF18A from accumulating at the plus-ends of kinetochore microtubules. These findings demonstrate that post-translational modifications in the enzymatic region of kinesins could be important for biasing their localization to particular microtubule subpopulations.
Collapse
Affiliation(s)
- Katelyn A. Queen
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05401
| | - Alisa Cario
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05401
- Current Institution: Department of Cell and Developmental Biology, Vanderbilt School of Medicine, Nashville, TN
| | - Christopher L. Berger
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05401
| | - Jason Stumpff
- Department of Molecular Physiology and Biophysics, University of Vermont, Burlington, VT, 05401
| |
Collapse
|
36
|
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
|
37
|
Liu T, Yang K, Chen J, Qi L, Zhou X, Wang P. Comprehensive Pan-Cancer Analysis of KIF18A as a Marker for Prognosis and Immunity. Biomolecules 2023; 13:biom13020326. [PMID: 36830695 PMCID: PMC9953516 DOI: 10.3390/biom13020326] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/30/2023] [Accepted: 02/06/2023] [Indexed: 02/11/2023] Open
Abstract
KIF18A belongs to the Kinesin family, which participates in the occurrence and progression of tumors. However, few pan-cancer analyses have been performed on KIF18A to date. We used multiple public databases such as TIMER, The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), and Human Protein Atlas (HPA) to explore KIF18A mRNA expression in 33 tumors. We performed immunohistochemistry on liver cancer and pancreatic cancer tissues and corresponding normal tissues to examine the expression of KIF18A protein. Univariate Cox regression and Kaplan-Meier survival analysis were applied to detect the effect of KIF18A on overall survival (OS), disease-specific survival (DSS), and progression-free interval (PFI) of patients with these tumors. Subsequently, we explored KIF18A gene alterations in different tumor tissues using cBioPortal. The relationship between KIF18A and clinical characteristics, tumor microenvironment (TME), immune regulatory genes, immune checkpoints, tumor mutational burden (TMB), microsatellite instability (MSI), mismatch repairs (MMRs), DNA methylation, RNA methylation, and drug sensitivity was applied for further study using the R language. Gene Set Enrichment Analysis (GSEA) was utilized to explore the molecular mechanism of KIF18A. Bioinformatic analysis and immunohistochemical experiments confirmed that KIF18A was up-regulated in 27 tumors and was correlated with the T stage, N stage, pathological stage, histological grade, and Ki-67 index in many cancers. The overexpression of KIF18A had poor OS, DSS, and PFI in adrenocortical carcinoma (ACC), kidney renal clear cell carcinoma (KIRC), kidney renal papillary cell carcinoma (KIRP), brain lower-grade glioma (LGG), liver cancer (LIHC), lung adenocarcinoma (LUAD), and pancreatic cancer (PAAD). Univariate and multivariate regression analysis confirmed KIF18A as an independent prognostic factor for LIHC and PAAD. The mutation frequency of KIF18A is the highest in endometrial cancer. KIF18A expression levels were positively associated with immunocyte infiltration, immune regulatory genes, immune checkpoints, TMB, MSI, MMRs, DNA methylation, RNA methylation, and drug sensitivity in certain cancers. In addition, we discovered that KIF18A participated in the cell cycle at the single-cell level and GSEA analysis for most cancers. These findings suggested that KIF18A could be regarded as a latent prognostic marker and a new target for cancer immunological therapy.
Collapse
|
38
|
Singh A, Busacca S, Gaba A, Sheaff M, Poile C, Nakas A, Dzialo J, Bzura A, Dawson AG, Fennell DA, Fry AM. BAP1 loss induces mitotic defects in mesothelioma cells through BRCA1-dependent and independent mechanisms. Oncogene 2023; 42:572-585. [PMID: 36550359 PMCID: PMC9937923 DOI: 10.1038/s41388-022-02577-3] [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: 07/15/2022] [Revised: 12/06/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022]
Abstract
The tumour suppressor BRCA1-associated protein 1 (BAP1) is the most frequently mutated cancer gene in mesothelioma. Here we report novel functions for BAP1 in mitotic progression highlighting the relationship between BAP1 and control of genome stability in mesothelioma cells with therapeutic implications. Depletion of BAP1 protein induced proteasome-mediated degradation of BRCA1 in mesothelioma cells while loss of BAP1 correlated with BRCA1 loss in mesothelioma patient tumour samples. BAP1 loss also led to mitotic defects that phenocopied the loss of BRCA1 including spindle assembly checkpoint failure, centrosome amplification and chromosome segregation errors. However, loss of BAP1 also led to additional mitotic changes that were not observed upon BRCA1 loss, including an increase in spindle length and enhanced growth of astral microtubules. Intriguingly, these consequences could be explained by loss of expression of the KIF18A and KIF18B kinesin motors that occurred upon depletion of BAP1 but not BRCA1, as spindle and astral microtubule defects were rescued by re-expression of KIF18A and KIF18B, respectively. We therefore propose that BAP1 inactivation causes mitotic defects through BRCA1-dependent and independent mechanisms revealing novel routes by which mesothelioma cells lacking BAP1 may acquire genome instability and exhibit altered responses to microtubule-targeted agents.
Collapse
Affiliation(s)
- Anita Singh
- grid.9918.90000 0004 1936 8411Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester, LE1 9HN UK ,grid.9918.90000 0004 1936 8411Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX UK
| | - Sara Busacca
- grid.9918.90000 0004 1936 8411Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX UK
| | - Aarti Gaba
- grid.9918.90000 0004 1936 8411Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX UK
| | - Michael Sheaff
- Department of Histopathology, Barts Health NHS Trust, Queen Mary University of London, The Royal London Hospital, London, E1 2ES UK
| | - Charlotte Poile
- grid.9918.90000 0004 1936 8411Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX UK
| | - Apostolos Nakas
- grid.412925.90000 0004 0400 6581University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, LE3 9QP UK
| | - Joanna Dzialo
- grid.9918.90000 0004 1936 8411Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX UK
| | - Aleksandra Bzura
- grid.9918.90000 0004 1936 8411Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX UK
| | - Alan G. Dawson
- grid.9918.90000 0004 1936 8411Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX UK ,grid.412925.90000 0004 0400 6581University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, LE3 9QP UK
| | - Dean A. Fennell
- grid.9918.90000 0004 1936 8411Leicester Cancer Research Centre, Department of Genetics and Genome Biology, University of Leicester, Robert Kilpatrick Clinical Sciences Building, Leicester, LE2 7LX UK ,grid.412925.90000 0004 0400 6581University Hospitals of Leicester NHS Trust, Glenfield Hospital, Leicester, LE3 9QP UK
| | - Andrew M. Fry
- grid.9918.90000 0004 1936 8411Department of Molecular and Cell Biology, University of Leicester, Lancaster Road, Leicester, LE1 9HN UK
| |
Collapse
|
39
|
Matković J, Ghosh S, Ćosić M, Eibes S, Barišić M, Pavin N, Tolić IM. Kinetochore- and chromosome-driven transition of microtubules into bundles promotes spindle assembly. Nat Commun 2022; 13:7307. [PMID: 36435852 PMCID: PMC9701229 DOI: 10.1038/s41467-022-34957-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 11/11/2022] [Indexed: 11/28/2022] Open
Abstract
Mitotic spindle assembly is crucial for chromosome segregation and relies on bundles of microtubules that extend from the poles and overlap in the middle. However, how these structures form remains poorly understood. Here we show that overlap bundles arise through a network-to-bundles transition driven by kinetochores and chromosomes. STED super-resolution microscopy reveals that PRC1-crosslinked microtubules initially form loose arrays, which become rearranged into bundles. Kinetochores promote microtubule bundling by lateral binding via CENP-E/kinesin-7 in an Aurora B-regulated manner. Steric interactions between the bundle-associated chromosomes at the spindle midplane drive bundle separation and spindle widening. In agreement with experiments, theoretical modeling suggests that bundles arise through competing attractive and repulsive mechanisms. Finally, perturbation of overlap bundles leads to inefficient correction of erroneous kinetochore-microtubule attachments. Thus, kinetochores and chromosomes drive coarsening of a uniform microtubule array into overlap bundles, which promote not only spindle formation but also chromosome segregation fidelity.
Collapse
Affiliation(s)
- Jurica Matković
- grid.4905.80000 0004 0635 7705Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Subhadip Ghosh
- grid.4808.40000 0001 0657 4636Department of Physics, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Mateja Ćosić
- grid.4905.80000 0004 0635 7705Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| | - Susana Eibes
- grid.417390.80000 0001 2175 6024Cell Division and Cytoskeleton, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Marin Barišić
- grid.417390.80000 0001 2175 6024Cell Division and Cytoskeleton, Danish Cancer Society Research Center, Copenhagen, Denmark ,grid.5254.60000 0001 0674 042XDepartment of Cellular and Molecular Medicine, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Nenad Pavin
- grid.4808.40000 0001 0657 4636Department of Physics, Faculty of Science, University of Zagreb, Zagreb, Croatia
| | - Iva M. Tolić
- grid.4905.80000 0004 0635 7705Division of Molecular Biology, Ruđer Bošković Institute, Zagreb, Croatia
| |
Collapse
|
40
|
Liu T, Shilliday F, Cook AD, Zeeshan M, Brady D, Tewari R, Sutherland CJ, Roberts AJ, Moores CA. Mechanochemical tuning of a kinesin motor essential for malaria parasite transmission. Nat Commun 2022; 13:6988. [PMID: 36384964 PMCID: PMC9669022 DOI: 10.1038/s41467-022-34710-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 11/02/2022] [Indexed: 11/17/2022] Open
Abstract
Plasmodium species cause malaria and kill hundreds of thousands annually. The microtubule-based motor kinesin-8B is required for development of the flagellated Plasmodium male gamete, and its absence completely blocks parasite transmission. To understand the molecular basis of kinesin-8B's essential role, we characterised the in vitro properties of kinesin-8B motor domains from P. berghei and P. falciparum. Both motors drive ATP-dependent microtubule gliding, but also catalyse ATP-dependent microtubule depolymerisation. We determined these motors' microtubule-bound structures using cryo-electron microscopy, which showed very similar modes of microtubule interaction in which Plasmodium-distinct sequences at the microtubule-kinesin interface influence motor function. Intriguingly however, P. berghei kinesin-8B exhibits a non-canonical structural response to ATP analogue binding such that neck linker docking is not induced. Nevertheless, the neck linker region is required for motility and depolymerisation activities of these motors. These data suggest that the mechanochemistry of Plasmodium kinesin-8Bs is functionally tuned to support flagella formation.
Collapse
Affiliation(s)
- Tianyang Liu
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, UK
| | - Fiona Shilliday
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, UK
| | - Alexander D Cook
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, UK
- Department of Biochemistry, University of Oxford, Oxford, OX1 3QU, UK
| | - Mohammad Zeeshan
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Declan Brady
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Rita Tewari
- School of Life Sciences, University of Nottingham, Nottingham, NG7 2UH, UK
| | - Colin J Sutherland
- Department of Infection Biology, Faculty of Infectious & Tropical Diseases, London School of Hygiene & Tropical Medicine, Keppel Street, London, WC1E 7HT, UK
| | - Anthony J Roberts
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, UK
| | - Carolyn A Moores
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX, UK.
| |
Collapse
|
41
|
Štimac V, Koprivec I, Manenica M, Simunić J, Tolić IM. Augmin prevents merotelic attachments by promoting proper arrangement of bridging and kinetochore fibers. eLife 2022; 11:e83287. [PMID: 36269126 PMCID: PMC9640188 DOI: 10.7554/elife.83287] [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: 09/07/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022] Open
Abstract
The human mitotic spindle is made of microtubules nucleated at centrosomes, at kinetochores, and from pre-existing microtubules by the augmin complex. However, it is unknown how the augmin-mediated nucleation affects distinct microtubule classes and thereby mitotic fidelity. Here, we use superresolution microscopy to analyze the previously indistinguishable microtubule arrangements within the crowded metaphase plate area and demonstrate that augmin is vital for the formation of uniformly arranged parallel units consisting of sister kinetochore fibers connected by a bridging fiber. This ordered geometry helps both prevent and resolve merotelic attachments. Whereas augmin-nucleated bridging fibers prevent merotelic attachments by creating a nearly parallel and highly bundled microtubule arrangement unfavorable for creating additional attachments, augmin-nucleated k-fibers produce robust force required to resolve errors during anaphase. STED microscopy revealed that bridging fibers were impaired twice as much as k-fibers following augmin depletion. The complete absence of bridging fibers from a significant portion of kinetochore pairs, especially in the inner part of the spindle, resulted in the specific reduction of the interkinetochore distance. Taken together, we propose a model where augmin promotes mitotic fidelity by generating assemblies consisting of bridging and kinetochore fibers that align sister kinetochores to face opposite poles, thereby preventing erroneous attachments.
Collapse
Affiliation(s)
- Valentina Štimac
- Division of Molecular Biology, Ruđer Bošković InstituteZagrebCroatia
| | - Isabella Koprivec
- Division of Molecular Biology, Ruđer Bošković InstituteZagrebCroatia
| | - Martina Manenica
- Division of Molecular Biology, Ruđer Bošković InstituteZagrebCroatia
| | - Juraj Simunić
- Division of Molecular Biology, Ruđer Bošković InstituteZagrebCroatia
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković InstituteZagrebCroatia
| |
Collapse
|
42
|
Risteski P, Božan D, Jagrić M, Bosilj A, Pavin N, Tolić IM. Length-dependent poleward flux of sister kinetochore fibers promotes chromosome alignment. Cell Rep 2022; 40:111169. [PMID: 35926461 PMCID: PMC9364240 DOI: 10.1016/j.celrep.2022.111169] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 04/20/2022] [Accepted: 07/14/2022] [Indexed: 11/25/2022] Open
Abstract
Chromosome alignment at the spindle equator promotes proper chromosome segregation and depends on pulling forces exerted at kinetochore fiber tips together with polar ejection forces. However, kinetochore fibers are also subjected to forces driving their poleward flux. Here we introduce a flux-driven centering model that relies on flux generated by forces within the overlaps of bridging and kinetochore fibers. This centering mechanism works so that the longer kinetochore fiber fluxes faster than the shorter one, moving the kinetochores toward the center. We develop speckle microscopy in human spindles and confirm the key prediction that kinetochore fiber flux is length dependent. Kinetochores are better centered when overlaps are shorter and the kinetochore fiber flux slower than the bridging fiber flux. We identify Kif18A and Kif4A as overlap and flux regulators and NuMA as a fiber coupler. Thus, length-dependent sliding forces exerted by the bridging fiber onto kinetochore fibers support chromosome alignment. A flux-driven centering model explains chromosome alignment at the spindle equator Kinetochore fiber poleward flux is driven by overlap microtubules Kinetochore centering requires faster flux of the longer sister kinetochore fiber Chromosome alignment depends on the overlap length of bridging microtubules
Collapse
Affiliation(s)
- Patrik Risteski
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Domagoj Božan
- Department of Physics, Faculty of Science, University of Zagreb, Bijenička cesta 32, 10000 Zagreb, Croatia
| | - Mihaela Jagrić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia
| | - Agneza Bosilj
- Department of Physics, Faculty of Science, University of Zagreb, Bijenička cesta 32, 10000 Zagreb, Croatia
| | - Nenad Pavin
- Department of Physics, Faculty of Science, University of Zagreb, Bijenička cesta 32, 10000 Zagreb, Croatia.
| | - Iva M Tolić
- Division of Molecular Biology, Ruđer Bošković Institute, Bijenička cesta 54, 10000 Zagreb, Croatia.
| |
Collapse
|
43
|
Mora-Bermúdez F, Kanis P, Macak D, Peters J, Naumann R, Xing L, Sarov M, Winkler S, Oegema CE, Haffner C, Wimberger P, Riesenberg S, Maricic T, Huttner WB, Pääbo S. Longer metaphase and fewer chromosome segregation errors in modern human than Neanderthal brain development. SCIENCE ADVANCES 2022; 8:eabn7702. [PMID: 35905187 PMCID: PMC9337762 DOI: 10.1126/sciadv.abn7702] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Accepted: 06/15/2022] [Indexed: 06/15/2023]
Abstract
Since the ancestors of modern humans separated from those of Neanderthals, around 100 amino acid substitutions spread to essentially all modern humans. The biological significance of these changes is largely unknown. Here, we examine all six such amino acid substitutions in three proteins known to have key roles in kinetochore function and chromosome segregation and to be highly expressed in the stem cells of the developing neocortex. When we introduce these modern human-specific substitutions in mice, three substitutions in two of these proteins, KIF18a and KNL1, cause metaphase prolongation and fewer chromosome segregation errors in apical progenitors of the developing neocortex. Conversely, the ancestral substitutions cause shorter metaphase length and more chromosome segregation errors in human brain organoids, similar to what we find in chimpanzee organoids. These results imply that the fidelity of chromosome segregation during neocortex development improved in modern humans after their divergence from Neanderthals.
Collapse
Affiliation(s)
- Felipe Mora-Bermúdez
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Philipp Kanis
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Dominik Macak
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Jula Peters
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Ronald Naumann
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Lei Xing
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Mihail Sarov
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Sylke Winkler
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | | | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Pauline Wimberger
- Department of Gynecology and Obstetrics, Technische Universität Dresden, Dresden, Germany
| | | | - Tomislav Maricic
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Wieland B. Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Svante Pääbo
- Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Okinawa Institute of Science and Technology, Onna-son 904-0495, Japan
| |
Collapse
|
44
|
Hunter B, Benoit MPMH, Asenjo AB, Doubleday C, Trofimova D, Frazer C, Shoukat I, Sosa H, Allingham JS. Kinesin-8-specific loop-2 controls the dual activities of the motor domain according to tubulin protofilament shape. Nat Commun 2022; 13:4198. [PMID: 35859148 PMCID: PMC9300613 DOI: 10.1038/s41467-022-31794-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/04/2022] [Indexed: 12/29/2022] Open
Abstract
Kinesin-8s are dual-activity motor proteins that can move processively on microtubules and depolymerize microtubule plus-ends, but their mechanism of combining these distinct activities remains unclear. We addressed this by obtaining cryo-EM structures (2.6-3.9 Å) of Candida albicans Kip3 in different catalytic states on the microtubule lattice and on a curved microtubule end mimic. We also determined a crystal structure of microtubule-unbound CaKip3-ADP (2.0 Å) and analyzed the biochemical activity of CaKip3 and kinesin-1 mutants. These data reveal that the microtubule depolymerization activity of kinesin-8 originates from conformational changes of its motor core that are amplified by dynamic contacts between its extended loop-2 and tubulin. On curved microtubule ends, loop-1 inserts into preceding motor domains, forming head-to-tail arrays of kinesin-8s that complement loop-2 contacts with curved tubulin and assist depolymerization. On straight tubulin protofilaments in the microtubule lattice, loop-2-tubulin contacts inhibit conformational changes in the motor core, but in the ADP-Pi state these contacts are relaxed, allowing neck-linker docking for motility. We propose that these tubulin shape-induced alternations between pro-microtubule-depolymerization and pro-motility kinesin states, regulated by loop-2, are the key to the dual activity of kinesin-8 motors.
Collapse
Affiliation(s)
- Byron Hunter
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Matthieu P M H Benoit
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Ana B Asenjo
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Caitlin Doubleday
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Daria Trofimova
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Corey Frazer
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Irsa Shoukat
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada
| | - Hernando Sosa
- Department of Biochemistry, Albert Einstein College of Medicine, Bronx, NY, 10461, USA.
| | - John S Allingham
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON, K7L 3N6, Canada.
| |
Collapse
|
45
|
Peyrégne S, Kelso J, Peter BM, Pääbo S. The evolutionary history of human spindle genes includes back-and-forth gene flow with Neandertals. eLife 2022; 11:e75464. [PMID: 35816093 PMCID: PMC9273211 DOI: 10.7554/elife.75464] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 06/14/2022] [Indexed: 12/13/2022] Open
Abstract
Proteins associated with the spindle apparatus, a cytoskeletal structure that ensures the proper segregation of chromosomes during cell division, experienced an unusual number of amino acid substitutions in modern humans after the split from the ancestors of Neandertals and Denisovans. Here, we analyze the history of these substitutions and show that some of the genes in which they occur may have been targets of positive selection. We also find that the two changes in the kinetochore scaffold 1 (KNL1) protein, previously believed to be specific to modern humans, were present in some Neandertals. We show that the KNL1 gene of these Neandertals shared a common ancestor with present-day Africans about 200,000 years ago due to gene flow from the ancestors (or relatives) of modern humans into Neandertals. Subsequently, some non-Africans inherited this modern human-like gene variant from Neandertals, but none inherited the ancestral gene variants. These results add to the growing evidence of early contacts between modern humans and archaic groups in Eurasia and illustrate the intricate relationships among these groups.
Collapse
Affiliation(s)
- Stéphane Peyrégne
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
| | - Janet Kelso
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
| | - Benjamin M Peter
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
| | - Svante Pääbo
- Department of Evolutionary Genetics, Max Planck Institute for Evolutionary AnthropologyLeipzigGermany
| |
Collapse
|
46
|
Teng C, Zhu Y, Li Y, Dai L, Pan Z, Wanggou S, Li X. Recurrence- and Malignant Progression-Associated Biomarkers in Low-Grade Gliomas and Their Roles in Immunotherapy. Front Immunol 2022; 13:899710. [PMID: 35677036 PMCID: PMC9168984 DOI: 10.3389/fimmu.2022.899710] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 04/12/2022] [Indexed: 12/15/2022] Open
Abstract
Despite a generally better prognosis than high-grade glioma (HGG), recurrence and malignant progression are the main causes for the poor prognosis and difficulties in the treatment of low-grade glioma (LGG). It is of great importance to learn about the risk factors and underlying mechanisms of LGG recurrence and progression. In this study, the transcriptome characteristics of four groups, namely, normal brain tissue and recurrent LGG (rLGG), normal brain tissue and secondary glioblastoma (sGBM), primary LGG (pLGG) and rLGG, and pLGG and sGBM, were compared using Chinese Glioma Genome Atlas (CGGA) and Genotype-Tissue Expression Project (GTEx) databases. In this study, 296 downregulated and 396 upregulated differentially expressed genes (DEGs) with high consensus were screened out. Univariate Cox regression analysis of data from The Cancer Genome Atlas (TCGA) yielded 86 prognostically relevant DEGs; a prognostic prediction model based on five key genes (HOXA1, KIF18A, FAM133A, HGF, and MN1) was established using the least absolute shrinkage and selection operator (LASSO) regression dimensionality reduction and multivariate Cox regression analysis. LGG was divided into high- and low-risk groups using this prediction model. Gene Set Enrichment Analysis (GSEA) revealed that signaling pathway differences in the high- and low-risk groups were mainly seen in tumor immune regulation and DNA damage-related cell cycle checkpoints. Furthermore, the infiltration of immune cells in the high- and low-risk groups was analyzed, which indicated a stronger infiltration of immune cells in the high-risk group than that in the low-risk group, suggesting that an immune microenvironment more conducive to tumor growth emerged due to the interaction between tumor and immune cells. The tumor mutational burden and tumor methylation burden in the high- and low-risk groups were also analyzed, which indicated higher gene mutation burden and lower DNA methylation level in the high-risk group, suggesting that with the accumulation of genomic mutations and epigenetic changes, tumor cells continued to evolve and led to the progression of LGG to HGG. Finally, the value of potential therapeutic targets for the five key genes was analyzed, and findings demonstrated that KIF18A was the gene most likely to be a potential therapeutic target. In conclusion, the prediction model based on these five key genes can better identify the high- and low-risk groups of LGG and lay a solid foundation for evaluating the risk of LGG recurrence and malignant progression.
Collapse
Affiliation(s)
- Chubei Teng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China.,Department of Neurosurgery, The First Affiliated Hospital, University of South China, Hengyang, China
| | - Yongwei Zhu
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Yueshuo Li
- Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Luohuan Dai
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Zhouyang Pan
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Siyi Wanggou
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| | - Xuejun Li
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China.,Hunan International Scientific and Technological Cooperation Base of Brain Tumor Research, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
47
|
Feng M, Swevers L, Sun J. Hemocyte Clusters Defined by scRNA-Seq in Bombyx mori: In Silico Analysis of Predicted Marker Genes and Implications for Potential Functional Roles. Front Immunol 2022; 13:852702. [PMID: 35281044 PMCID: PMC8914287 DOI: 10.3389/fimmu.2022.852702] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 02/07/2022] [Indexed: 12/16/2022] Open
Abstract
Within the hemolymph, insect hemocytes constitute a heterogeneous population of macrophage-like cells that play important roles in innate immunity, homeostasis and development. Classification of hemocytes in different subtypes by size, morphology and biochemical or immunological markers has been difficult and only in Drosophila extensive genetic analysis allowed the construction of a coherent picture of hemocyte differentiation from pro-hemocytes to granulocytes, crystal cells and plasmatocytes. However, the advent of high-throughput single cell technologies, such as single cell RNA sequencing (scRNA-seq), is bound to have a high impact on the study of hemocytes subtypes and their phenotypes in other insects for which a sophisticated genetic toolbox is not available. Instead of averaging gene expression across all cells as occurs in bulk-RNA-seq, scRNA-seq allows high-throughput and specific visualization of the differentiation status of individual cells. With scRNA-seq, interesting cell types can be identified in heterogeneous populations and direct analysis of rare cell types is possible. Next to its ability to profile the transcriptomes of individual cells in tissue samples, scRNA-seq can be used to propose marker genes that are characteristic of different hemocyte subtypes and predict their functions. In this perspective, the identities of the different marker genes that were identified by scRNA-seq analysis to define 13 distinct cell clusters of hemocytes in larvae of the silkworm, Bombyx mori, are discussed in detail. The analysis confirms the broad division of hemocytes in granulocytes, plasmatocytes, oenocytoids and perhaps spherulocytes but also reveals considerable complexity at the molecular level and highly specialized functions. In addition, predicted hemocyte marker genes in Bombyx generally show only limited convergence with the genes that are considered characteristic for hemocyte subtypes in Drosophila.
Collapse
Affiliation(s)
- Min Feng
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| | - Luc Swevers
- Insect Molecular Genetics and Biotechnology, Institute of Biosciences & Applications, National Centre for Scientific Research "Demokritos", Aghia Paraskevi, Athens, Greece
| | - Jingchen Sun
- Guangdong Provincial Key Laboratory of Agro-animal Genomics and Molecular Breeding, College of Animal Science, South China Agricultural University, Guangzhou, China
| |
Collapse
|
48
|
Wang LB, Zhang XB, Liu J, Liu QJ. The Proliferation of Glioblastoma Is Contributed to Kinesin Family Member 18A and Medical Data Analysis of GBM. Front Genet 2022; 13:858882. [PMID: 35464837 PMCID: PMC9033168 DOI: 10.3389/fgene.2022.858882] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 01/26/2022] [Indexed: 11/23/2022] Open
Abstract
Background: Glioblastoma (GBM) is widely known as a classical kind of malignant tumor originating in the brain with high morbidity and mortality. Targeted therapy has shown great promise in treating glioblastoma, but more promising targets, including effective therapeutic targets, remain to be identified. 18A (KIF18A) is a microtubule-based motor protein that is dysregulated and involved in the progression of multiple human cancers. However, the possible effects of KIF18A on GBM progression are still unclear. Methods: We performed DEG analysis, medical data analysis, and network analysis to identify critical genes affecting glioma progression. We also performed immunohistochemical analysis of the KIF18A levels in 94 patients with glioblastoma and the associated surrounding tissues. Patients were divided into two groups according to the high and low expression. Using a clinical analysis, we showed the potential associations between KIF18A expression and clinical characteristics of 94 GBM patients. We then investigated the effects of KIF18A on GBM cell proliferation by colony establishment, MTT, and immune blogging. The possible effect of KIF18A on GBM tumor growth was determined in mice. Results: We identified KIF18A as a potential gene affecting GBM progression. We further demonstrated that GBM tissues expressed KIF18A much higher, and its presentation was associated with recurrence in glioblastoma patients. We believe KIF18A promotes GBM cell proliferation. Conclusion: We demonstrated that KIF18A could be a promising target in treating GBM.
Collapse
Affiliation(s)
- Lei-Bo Wang
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Xue-Bin Zhang
- Department of Pathology, Tianjin Huanhu Hospital, Tianjin, China
| | - Jun Liu
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
| | - Qing-Jun Liu
- Department of Neurosurgery, Tianjin Huanhu Hospital, Tianjin, China
- *Correspondence: Qing-Jun Liu,
| |
Collapse
|
49
|
Cohen-Armon M. Exclusive modifications of NuMA in malignant epithelial cells: A potential therapeutic mechanism. Drug Discov Today 2022; 27:1205-1209. [PMID: 35143964 DOI: 10.1016/j.drudis.2022.02.002] [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: 09/02/2021] [Revised: 12/13/2021] [Accepted: 02/03/2022] [Indexed: 11/03/2022]
Abstract
NuMA (nuclear mitotic apparatus) protein is indispensable in the mitosis of human proliferating cells, both malignant and benign. The progression of mitosis requires stable spindles, which depend on the bipolar clustering of NuMA within the spindles. The phenanthridine PJ34 kills malignant epithelial cells during mitosis and targets NuMA. PJ34 exclusively blocks the post-translational modification of NuMA in a variety of malignant epithelial cells, but not in benign cells. This blockage of the post-translational modification of NuMA affects its protein-binding capacity and causes construction faults in the mitotic spindle poles of PJ34-treated cancer cells, leading to mitotic catastrophe cell death. PJ34 is a potent PARP1 inhibitor, so its cytotoxicity in human malignant cells is exclusively independent of PARP, challenging the currently accepted notion that inhibition of PARP1 halts cancer by preventing DNA repair. Certain molecules that act as PARP1 inhibitors target other proteins and vital mechanisms in human cancer cells.
Collapse
Affiliation(s)
- Malka Cohen-Armon
- The Sackler School of Medicine and Sagol School of Neuroscience, Tel Aviv University, Tel-Aviv 69978, Israel.
| |
Collapse
|
50
|
Begley MA, Solon AL, Davis EM, Sherrill MG, Ohi R, Elting MW. K-fiber bundles in the mitotic spindle are mechanically reinforced by Kif15. Mol Biol Cell 2021; 32:br11. [PMID: 34668719 PMCID: PMC8694074 DOI: 10.1091/mbc.e20-06-0426] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The mitotic spindle, a self-constructed microtubule-based machine, segregates chromosomes during cell division. In mammalian cells, microtubule bundles called kinetochore fibers (k-fibers) connect chromosomes to the spindle poles. Chromosome segregation thus depends on the mechanical integrity of k-fibers. Here we investigate the physical and molecular basis of k-fiber bundle cohesion. We detach k-fibers from poles by laser ablation-based cutting, thus revealing the contribution of pole-localized forces to k-fiber cohesion. We then measure the physical response of the remaining kinetochore-bound segments of the k-fibers. We observe that microtubules within ablated k-fibers often splay apart from their minus-ends. Furthermore, we find that minus-end clustering forces induced by ablation seem at least partially responsible for k-fiber splaying. We also investigate the role of the k-fiber-binding kinesin-12 Kif15. We find that pharmacological inhibition of Kif15-microtubule binding reduces the mechanical integrity of k-fibers. In contrast, inhibition of its motor activity but not its microtubule binding ability, i.e., locking Kif15 into a rigor state, does not greatly affect splaying. Altogether, the data suggest that forces holding k-fibers together are of similar magnitude to other spindle forces, and that Kif15, acting as a microtubule cross-linker, helps fortify and repair k-fibers. This feature of Kif15 may help support robust k-fiber function and prevent chromosome segregation errors.
Collapse
Affiliation(s)
- Marcus A Begley
- Department of Physics, North Carolina State University, Raleigh, NC 27607
| | - April L Solon
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | | | | | - Ryoma Ohi
- Department of Cell & Developmental Biology, University of Michigan, Ann Arbor, MI 48109
| | - Mary Williard Elting
- Department of Physics, North Carolina State University, Raleigh, NC 27607.,Quantitative and Computational Developmental Biology Cluster, North Carolina State University, Raleigh, NC 27695
| |
Collapse
|