1
|
Zhou J, Zhang Z, Huang R, Zhuang X, Ni S. Mechanisms of Picrasma quassioides against hepatocellular carcinoma elucidated by network pharmacology and experimental validation. Am J Transl Res 2025; 17:1402-1415. [PMID: 40092104 PMCID: PMC11909513 DOI: 10.62347/vlgd3371] [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: 09/26/2024] [Accepted: 01/18/2025] [Indexed: 03/19/2025]
Abstract
OBJECTIVE The medicinal plant Picrasma quassioides (P. quassioides) Benn exerts an inhibitory effect on the growth of hepatocellular carcinoma (HCC) cells via an unknown mechanism. This study explored the targets and signaling pathways underlying the mechanism of P. quassioides against HCC. METHODS Targets of P. quassioides active compounds were identified using the HERB database, and the HCC targets were found with the GeneCards database. The optimal serum concentration and intervention time were determined using the CCK-8 assay. Apoptosis, cell cycle, invasion, cloning, and wound-healing abilities were assessed using flow cytometry. Core protein targets and signaling pathway-related metabolic enzymes were evaluated with Western blotting. The anti-HCC effect of P. quassioides medicated serum was verified using arachidonic acid (AA)-related enzyme agonists. RESULTS Network pharmacology identified 19 effective compounds of P. quassioides and 105 HCC-associated targets. It also revealed the AA pathway was the central pathway of P. quassioides against HCC, with AURKA, AURKB, KIF11, and TOP2A identified as core targets that inhibit excessive HCC cell proliferation and promote apoptosis. Flow cytometry findings supported that P. quassioides medicated serum significantly inhibited HCC cell proliferation and promoted apoptosis. By contrast, enzyme agonists related to the AA pathway markedly counteracted the anti-HCC effect of P. quassioides, promoting HCC growth. CONCLUSION P. quassioides medicated serum exerts a prominent anti-HCC effect in vitro. The AA pathway constitutes the mechanism by which P. quassioides medicated serum inhibits excessive proliferation and promotes apoptosis of HCC cells.
Collapse
Affiliation(s)
- Jie Zhou
- School of Pharmacy, Anhui Medical University Hefei 230012, Anhui, China
| | - Zhilan Zhang
- School of Pharmacy, Anhui Medical University Hefei 230012, Anhui, China
| | - Ruiru Huang
- School of Pharmacy, Anhui Medical University Hefei 230012, Anhui, China
| | - Xingxing Zhuang
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University Chaohu 238000, Anhui, China
| | - Shoudong Ni
- School of Pharmacy, Anhui Medical University Hefei 230012, Anhui, China
- Department of Pharmacy, Chaohu Hospital of Anhui Medical University Chaohu 238000, Anhui, China
| |
Collapse
|
2
|
Ding Y, Yang H, Gao J, Tang C, Peng YY, Ma XM, Li S, Wang HY, Lu XM, Wang YT. Synaptic-mitochondrial transport: mechanisms in neural adaptation and degeneration. Mol Cell Biochem 2025:10.1007/s11010-025-05209-y. [PMID: 39841406 DOI: 10.1007/s11010-025-05209-y] [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: 10/16/2024] [Accepted: 01/04/2025] [Indexed: 01/23/2025]
Abstract
Synaptic plasticity is the basis for the proper functioning of the central nervous system. Synapses are the contact points between neurons and are crucial for information transmission, the structure and function of synapses change adaptively based on the different activities of neurons, thus affecting processes such as learning, memory, and neural development and repair. Synaptic activity requires a large amount of energy provided by mitochondria. Mitochondrial transport proteins regulate the positioning and movement of mitochondria to maintain normal energy metabolism. Recent studies have shown a close relationship between mitochondrial transport proteins and synaptic plasticity, providing a new direction for the study of adaptive changes in the central nervous system and new targets for the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yang Ding
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Huan Yang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Jie Gao
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Can Tang
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yu-Yuan Peng
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Xin-Mei Ma
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Sen Li
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Hai-Yan Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Xiu-Min Lu
- College of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing, 400054, China
| | - Yong-Tang Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
3
|
Lu W, Lee BS, Deng HXY, Lakonishok M, Martin-Blanco E, Gelfand VI. "Mitotic" kinesin-5 is a dynamic brake for axonal growth. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.12.612721. [PMID: 39314406 PMCID: PMC11419024 DOI: 10.1101/2024.09.12.612721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
During neuronal development, neurons undergo significant microtubule reorganization to shape axons and dendrites, establishing the framework for efficient wiring of the nervous system. Previous studies from our laboratory demonstrated the key role of kinesin-1 in driving microtubule-microtubule sliding, which provides the mechanical forces necessary for early axon outgrowth and regeneration in Drosophila melanogaster. In this study, we reveal the critical role of kinesin-5, a mitotic motor, in modulating the development of postmitotic neurons. Kinesin-5, a conserved homotetrameric motor, typically functions in mitosis by sliding antiparallel microtubules apart in the spindle. Here, we demonstrate that the Drosophila kinesin-5 homolog, Klp61F, is expressed in larval brain neurons, with high levels in ventral nerve cord (VNC) neurons. Knockdown of Klp61F using a pan-neuronal driver leads to severe locomotion defects and complete lethality in adult flies, mainly due to the absence of kinesin-5 in VNC motor neurons during early larval development. Klp61F depletion results in significant axon growth defects, both in cultured and in vivo neurons. By imaging individual microtubules, we observe a significant increase in microtubule motility, and excessive penetration of microtubules into the axon growth cone in Klp61F-depleted neurons. Adult lethality and axon growth defects are fully rescued by a chimeric human-Drosophila kinesin-5 motor, which accumulates at the axon tips, suggesting a conserved role of kinesin-5 in neuronal development. Altogether, our findings show that at the growth cone, kinesin-5 acts as a brake on kinesin-1-driven microtubule sliding, preventing premature microtubule entry into the growth cone. This regulatory role of kinesin-5 is essential for precise axon pathfinding during nervous system development.
Collapse
Affiliation(s)
- Wen Lu
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Brad S. Lee
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Helen Xue Ying Deng
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Margot Lakonishok
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Enrique Martin-Blanco
- Instituto de Biología Molecular de Barcelona, CSIC, Parc Cientific de Barcelona, Baldiri Reixac 10-12, 08028 Barcelona, Spain
| | - Vladimir I. Gelfand
- Department of Cell and Developmental Biology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
4
|
Liu X, Rao L, Qiu W, Berger F, Gennerich A. Kinesin-14 HSET and KlpA are non-processive microtubule motors with load-dependent power strokes. Nat Commun 2024; 15:6564. [PMID: 39095439 PMCID: PMC11297315 DOI: 10.1038/s41467-024-50990-x] [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: 05/31/2023] [Accepted: 07/25/2024] [Indexed: 08/04/2024] Open
Abstract
Accurate chromosome segregation during cell division relies on coordinated actions of microtubule (MT)-based motor proteins in the mitotic spindle. Kinesin-14 motors play vital roles in spindle assembly and maintenance by crosslinking antiparallel MTs at the spindle midzone and anchoring spindle MTs' minus ends at the poles. In this study, we investigate the force generation and motility of the Kinesin-14 motors HSET and KlpA. Our findings reveal that both motors are non-processive, producing single load-dependent power strokes per MT encounter, with estimated load-free power strokes of ~30 and ~35 nm, respectively. Each homodimeric motor generates forces of ~0.5 pN, but when assembled in teams, they cooperate to generate forces of 1 pN or more. Notably, the cooperative activity among multiple motors leads to increased MT-sliding velocities. These results quantitatively elucidate the structure-function relationship of Kinesin-14 motors and underscore the significance of cooperative behavior in their cellular functions.
Collapse
Affiliation(s)
- Xinglei Liu
- Department of Biochemistry and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Lu Rao
- Department of Biochemistry and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Weihong Qiu
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, CH, Utrecht, The Netherlands
| | - Florian Berger
- Department of Biochemistry & Biophysics and Department of Physics, Oregon State University, Corvallis, OR, USA.
| | - Arne Gennerich
- Department of Biochemistry and Gruss Lipper Biophotonics Center, Albert Einstein College of Medicine, Bronx, NY, USA.
| |
Collapse
|
5
|
Jia N, Zhang B, Huo Z, Qin J, Ji Q, Geng Y. Binding patterns of inhibitors to different pockets of kinesin Eg5. Arch Biochem Biophys 2024; 756:109998. [PMID: 38641233 DOI: 10.1016/j.abb.2024.109998] [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: 12/20/2023] [Revised: 03/19/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024]
Abstract
The kinesin-5 family member, Eg5, plays very important role in the mitosis. As a mitotic protein, Eg5 is the target of various mitotic inhibitors. There are two targeting pockets in the motor domain of Eg5, which locates in the α2/L5/α3 region and the α4/α6 region respectively. We investigated the interactions between the different inhibitors and the two binding pockets of Eg5 by using all-atom molecular dynamics method. Combined the conformational analysis with the free-energy calculation, the binding patterns of inhibitors to the two binding pockets are shown. The α2/L5/α3 pocket can be divided into 4 regions. The structures and binding conformations of inhibitors in region 1 and 2 are highly conserved. The shape of α4/α6 pocket is alterable. The space of this pocket in ADP-binding state of Eg5 is larger than that in ADP·Pi-binding state due to the limitation of a hydrogen bond formed in the ADP·Pi-binding state. The results of this investigation provide the structural basis of the inhibitor-Eg5 interaction and offer a reference for the Eg5-targeted drug design.
Collapse
Affiliation(s)
- Ning Jia
- School of Science, Hebei University of Technology, Tianjin, China; Institute of Biophysics, Hebei University of Technology, Tianjin, China
| | - Bingbing Zhang
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin, China; Institute of Biophysics, Hebei University of Technology, Tianjin, China
| | - Ziling Huo
- School of Health Sciences & Biomedical Engineering, Hebei University of Technology, Tianjin, China; Institute of Biophysics, Hebei University of Technology, Tianjin, China
| | - Jingyu Qin
- College of Electrical and Information Engineering, Quzhou University, Quzhou, China
| | - Qing Ji
- School of Science, Hebei University of Technology, Tianjin, China; Institute of Biophysics, Hebei University of Technology, Tianjin, China
| | - Yizhao Geng
- School of Science, Hebei University of Technology, Tianjin, China; Institute of Biophysics, Hebei University of Technology, Tianjin, China.
| |
Collapse
|
6
|
Dong L, Feng C, Cheng W, Huang A, Ying K. FOXP3 targets KIF5A to increase lactate production and promote docetaxel resistance in lung adenocarcinoma. Acta Biochim Biophys Sin (Shanghai) 2024; 56:1011-1021. [PMID: 38798241 PMCID: PMC11322870 DOI: 10.3724/abbs.2024082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Accepted: 01/31/2024] [Indexed: 05/29/2024] Open
Abstract
A prominent cause of cancer-related fatalities with a poor prognosis is lung adenocarcinoma (LUAD). KIF5A, a crucial member of the kinesin superfamily, is linked to drug resistance in malignancies. This work aims to investigate the mechanism of KIF5A in docetaxel (DTX) resistance in LUAD cells. The results of bioinformatics analysis, qRT-PCR and western blot analysis show that KIF5A, which is involved in the glycolysis pathway, is highly expressed in LUAD and is positively correlated with glycolysis-related genes. We further verify that silencing of KIF5A inhibits DTX resistance, glycolysis, and lactate production in LUAD cells via cell counting kit-8 (CCK-8), flow cytometry, Seahorse XFe 96, lactate, and glucose assays. Mechanistically, KIF5A promotes DTX resistance in LUAD, and this effect is attenuated upon the addition of an LDHA inhibitor. Chromatin immunoprecipitation and dual-luciferase reporter assays reveal that FOXP3 transcriptionally activates KIF5A. Knockdown of FOXP3 reduces lactate production and enhances DTX sensitivity in LUAD, which is restored upon simultaneous overexpression of KIF5A. Our findings reveal that FOXP3 increases DTX resistance in LUAD cells by enhancing lactate production through the upregulation of KIF5A level. In conclusion, our study provides a novel treatment target for improving chemosensitivity in LUAD.
Collapse
Affiliation(s)
- Liangliang Dong
- Department of Respiratory and Critical Care MedicineRegional Medical Center for National Institute of Respiratory DiseasesSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310020China
| | - Chan Feng
- Department of Respiratory and Critical Care MedicineRegional Medical Center for National Institute of Respiratory DiseasesSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310020China
| | - Wenwen Cheng
- Department of Education OfficeSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310020China
| | - Aihua Huang
- Department of PathologySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310020China
| | - Kejing Ying
- Department of Respiratory and Critical Care MedicineRegional Medical Center for National Institute of Respiratory DiseasesSir Run Run Shaw HospitalZhejiang University School of MedicineHangzhou310020China
| |
Collapse
|
7
|
Laporte D, Massoni-Laporte A, Lefranc C, Dompierre J, Mauboules D, Nsamba ET, Royou A, Gal L, Schuldiner M, Gupta ML, Sagot I. A stable microtubule bundle formed through an orchestrated multistep process controls quiescence exit. eLife 2024; 12:RP89958. [PMID: 38527106 PMCID: PMC10963028 DOI: 10.7554/elife.89958] [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] [Indexed: 03/27/2024] Open
Abstract
Cells fine-tune microtubule assembly in both space and time to give rise to distinct edifices with specific cellular functions. In proliferating cells, microtubules are highly dynamics, and proliferation cessation often leads to their stabilization. One of the most stable microtubule structures identified to date is the nuclear bundle assembled in quiescent yeast. In this article, we characterize the original multistep process driving the assembly of this structure. This Aurora B-dependent mechanism follows a precise temporality that relies on the sequential actions of kinesin-14, kinesin-5, and involves both microtubule-kinetochore and kinetochore-kinetochore interactions. Upon quiescence exit, the microtubule bundle is disassembled via a cooperative process involving kinesin-8 and its full disassembly is required prior to cells re-entry into proliferation. Overall, our study provides the first description, at the molecular scale, of the entire life cycle of a stable microtubule structure in vivo and sheds light on its physiological function.
Collapse
Affiliation(s)
| | | | | | | | | | - Emmanuel T Nsamba
- Genetics, Development, and Cell Biology, Iowa State UniversityAmesUnited States
| | - Anne Royou
- Univ. Bordeaux, CNRS, IBGC, UMR 5095BordeauxFrance
| | - Lihi Gal
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Maya Schuldiner
- Department of Molecular Genetics, Weizmann Institute of ScienceRehovotIsrael
| | - Mohan L Gupta
- Genetics, Development, and Cell Biology, Iowa State UniversityAmesUnited States
| | | |
Collapse
|
8
|
Singh SK, Siegler N, Pandey H, Yanir N, Popov M, Goldstein-Levitin A, Sadan M, Debs G, Zarivach R, Frank GA, Kass I, Sindelar CV, Zalk R, Gheber L. Noncanonical interaction with microtubules via the N-terminal nonmotor domain is critical for the functions of a bidirectional kinesin. SCIENCE ADVANCES 2024; 10:eadi1367. [PMID: 38324691 PMCID: PMC10849588 DOI: 10.1126/sciadv.adi1367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 01/08/2024] [Indexed: 02/09/2024]
Abstract
Several kinesin-5 motors (kinesin-5s) exhibit bidirectional motility. The mechanism of such motility remains unknown. Bidirectional kinesin-5s share a long N-terminal nonmotor domain (NTnmd), absent in exclusively plus-end-directed kinesins. Here, we combined in vivo, in vitro, and cryo-electron microscopy (cryo-EM) studies to examine the impact of NTnmd mutations on the motor functions of the bidirectional kinesin-5, Cin8. We found that NTnmd deletion mutants exhibited cell viability and spindle localization defects. Using cryo-EM, we examined the structure of a microtubule (MT)-bound motor domain of Cin8, containing part of its NTnmd. Modeling and molecular dynamic simulations based on the cryo-EM map suggested that the NTnmd of Cin8 interacts with the C-terminal tail of β-tubulin. In vitro experiments on subtilisin-treated MTs confirmed this notion. Last, we showed that NTnmd mutants are defective in plus-end-directed motility in single-molecule and antiparallel MT sliding assays. These findings demonstrate that the NTnmd, common to bidirectional kinesin-5s, is critical for their bidirectional motility and intracellular functions.
Collapse
Affiliation(s)
- Sudhir K. Singh
- 1Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Nurit Siegler
- 1Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Himanshu Pandey
- 1Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Neta Yanir
- 1Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Mary Popov
- 1Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | | | - Mayan Sadan
- 1Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Garrett Debs
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Raz Zarivach
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Gabriel A. Frank
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Itamar Kass
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Charles V. Sindelar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, CT 06510, USA
| | - Ran Zalk
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| | - Larisa Gheber
- 1Department of Chemistry, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
- Ilse Katz Institute for Nanoscale Science & Technology, Ben-Gurion University of the Negev, Beer-Sheva 8410501, Israel
| |
Collapse
|
9
|
Benoit MP, Hunter B, Allingham JS, Sosa H. New insights into the mechanochemical coupling mechanism of kinesin-microtubule complexes from their high-resolution structures. Biochem Soc Trans 2023; 51:1505-1520. [PMID: 37560910 PMCID: PMC10586761 DOI: 10.1042/bst20221238] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/11/2023]
Abstract
Kinesin motor proteins couple mechanical movements in their motor domain to the binding and hydrolysis of ATP in their nucleotide-binding pocket. Forces produced through this 'mechanochemical' coupling are typically used to mobilize kinesin-mediated transport of cargos along microtubules or microtubule cytoskeleton remodeling. This review discusses the recent high-resolution structures (<4 Å) of kinesins bound to microtubules or tubulin complexes that have resolved outstanding questions about the basis of mechanochemical coupling, and how family-specific modifications of the motor domain can enable its use for motility and/or microtubule depolymerization.
Collapse
Affiliation(s)
| | - Byron Hunter
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, ON K7L 3N6, Canada
| | - John S. Allingham
- 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, U.S.A
| |
Collapse
|
10
|
Liu X, Rao L, Qiu W, Gennerich A. Kinesin-14 HSET and KlpA are non-processive microtubule motors with load-dependent power strokes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.09.544415. [PMID: 37333225 PMCID: PMC10274885 DOI: 10.1101/2023.06.09.544415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Accurate chromosome segregation during cell division relies on coordinated actions of microtubule (MT)-based motor proteins in the mitotic spindle. Kinesin-14 motors play vital roles in spindle assembly and maintenance by crosslinking antiparallel MTs at the spindle midzone and anchoring spindle MTs' minus ends at the poles. We investigate the force generation and motility of the Kinesin-14 motors HSET and KlpA, revealing that both motors function as non-processive motors under load, producing single power strokes per MT encounter. Each homodimeric motor generates forces of ∼0.5 pN, but when assembled in teams, they cooperate to generate forces of 1 pN or more. Importantly, cooperative activity among multiple motors leads to increased MT-sliding velocities. Our findings deepen our understanding of the structure-function relationship of Kinesin-14 motors and underscore the significance of cooperative behavior in their cellular functions.
Collapse
|
11
|
Differential Expression of Mitosis and Cell Cycle Regulatory Genes during Recovery from an Acute Respiratory Virus Infection. Pathogens 2021; 10:pathogens10121625. [PMID: 34959580 PMCID: PMC8708581 DOI: 10.3390/pathogens10121625] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/10/2021] [Accepted: 12/11/2021] [Indexed: 12/27/2022] Open
Abstract
Acute respiratory virus infections can have profound and long-term effects on lung function that persist even after the acute responses have fully resolved. In this study, we examined gene expression by RNA sequencing in the lung tissue of wild-type BALB/c mice that were recovering from a sublethal infection with the pneumonia virus of mice (PVM), a natural rodent pathogen of the same virus family and genus as the human respiratory syncytial virus. We compared these responses to gene expression in PVM-infected mice treated with Lactobacillus plantarum, an immunobiotic agent that limits inflammation and averts the negative clinical sequelae typically observed in response to acute infection with this pathogen. Our findings revealed prominent differential expression of inflammation-associated genes as well as numerous genes and gene families implicated in mitosis and cell-cycle regulation, including cyclins, cyclin-dependent kinases, cell division cycle genes, E2F transcription factors, kinesins, centromere proteins, and aurora kinases, among others. Of particular note was the differential expression of the cell division cycle gene Cdc20b, which was previously identified as critical for the ex vivo differentiation of multi-ciliated cells. Collectively, these findings provided us with substantial insight into post-viral repair processes and broadened our understanding of the mechanisms underlying Lactobacillus-mediated protection.
Collapse
|