1
|
Birkedal R, Branovets J, Vendelin M. Compartmentalization in cardiomyocytes modulates creatine kinase and adenylate kinase activities. FEBS Lett 2024; 598:2623-2640. [PMID: 39112921 DOI: 10.1002/1873-3468.14994] [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: 03/14/2024] [Revised: 06/03/2024] [Accepted: 07/21/2024] [Indexed: 11/12/2024]
Abstract
Intracellular molecules are transported by motor proteins or move by diffusion resulting from random molecular motion. Cardiomyocytes are packed with structures that are crucial for function, but also confine the diffusional spaces, providing cells with a means to control diffusion. They form compartments in which local concentrations are different from the overall, average concentrations. For example, calcium and cyclic AMP are highly compartmentalized, allowing these versatile second messengers to send different signals depending on their location. In energetic compartmentalization, the ratios of AMP and ADP to ATP are different from the average ratios. This is important for the performance of ATPases fuelling cardiac excitation-contraction coupling and mechanical work. A recent study suggested that compartmentalization modulates the activity of creatine kinase and adenylate kinase in situ. This could have implications for energetic signaling through, for example, AMP-activated kinase. It highlights the importance of taking compartmentalization into account in our interpretation of cellular physiology and developing methods to assess local concentrations of AMP and ADP to enhance our understanding of compartmentalization in different cell types.
Collapse
Affiliation(s)
- Rikke Birkedal
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| | - Jelena Branovets
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| | - Marko Vendelin
- Laboratory of Systems Biology, Department of Cybernetics, Tallinn University of Technology, Estonia
| |
Collapse
|
2
|
Halder R, Chu ZT, Ti R, Zhu L, Warshel A. On the Control of Directionality of Myosin. J Am Chem Soc 2024. [PMID: 39367841 DOI: 10.1021/jacs.4c09528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/07/2024]
Abstract
The origin of the unique directionality of myosin has been a problem of fundamental and practical importance. This work establishes in a conclusive way that the directionality is controlled by tuning the barrier for the rate-determining step, namely, the ADP release step. This conclusion is based on exploring the molecular origin behind the reverse directionality of myosins V and VI and the determination of the origin of the change in the barriers of the ADP release for the forward and backward motions. Our investigation is performed by combining different simulation methods such as steer molecular dynamics (SMD), umbrella sampling, renormalization method, and automated path searching method. It is found that in the case of myosin V, the ADP release from the postrigor (trailing head) state overcomes a lower barrier than the prepowerstroke (leading head) state, which is also evident from experimental observation. In the case of myosin VI, we noticed a different trend when compared to myosin V. Since the directionality of myosins V and VI follows a reverse trend, we conclude that such differences in the directionality are controlled by the free energy barrier for the ADP release. Overall, the proof that the directionality of myosin is determined by the activation barrier of the rate-determining step in the cycle, rather than by some unspecified dynamical effects, has general importance.
Collapse
Affiliation(s)
- Ritaban Halder
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| | - Zhen Tao Chu
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| | - Rujuan Ti
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Lizhe Zhu
- Warshel Institute for Computational Biology, School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong 518172, China
| | - Arieh Warshel
- Department of Chemistry, University of Southern California, Los Angeles, California 90089-1062, United States
| |
Collapse
|
3
|
Liu B, Liu C, Li Z, Liu W, Cui H, Yuan J. A subpellicular microtubule dynein transport machinery regulates ookinete morphogenesis for mosquito transmission of Plasmodium yoelii. Nat Commun 2024; 15:8590. [PMID: 39366980 PMCID: PMC11452633 DOI: 10.1038/s41467-024-52970-7] [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: 06/28/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024] Open
Abstract
The cortical cytoskeleton of subpellicular microtubules (SPMTs) supports the Plasmodium ookinete morphogenesis during mosquito transmission of malaria. SPMTs are hypothesized to function as the cytoskeletal tracks in motor-driven cargo transport for apical organelle and structure assembly in ookinetes. However, the SPMT-based transport motor has not been identified in the Plasmodium. The cytoplasmic dynein is the motor moving towards the minus end of microtubules (MTs) and likely be responsible for cargo transport to the apical part in ookinetes. Here we screen 7 putative dynein heavy chain (DHC) proteins in the P. yoelii and identify DHC3 showing peripheral localization in ookinetes. DHC3 is localized at SPMTs throughout ookinete morphogenesis. We also identify five other dynein subunits localizing at SPMTs. DHC3 disruption impairs ookinete development, shape, and gliding, leading to failure in mosquito infection of Plasmodium. The DHC3-deficient ookinetes display defective formation or localization of apical organelles and structures. Rab11A and Rab11B interact with DHC3 at SPMTs in a DHC3-dependent manner, likely functioning as the receptors for the cargoes driven by SPMT-dynein. Disturbing Rab11A or Rab11B phenocopies DHC3 deficiency in ookinete morphogenesis. Our study reveals an SPMT-based dynein motor driving the transport of Rab11A- and Rab11B-labeled cargoes in the ookinete morphogenesis of Plasmodium.
Collapse
Affiliation(s)
- Bing Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Cong Liu
- Department of Health Inspection and Quarantine, School of Public Health, Hengyang Medical School, University of South China, Hengyang, China
| | - Zhenkui Li
- Institute of Pathogenic Biology and Key Laboratory of Special Pathogen Prevention and Control of Hunan Province, School of Basic Medical Sciences, Hengyang Medical School, University of South China, Hengyang, China
| | - Wenjia Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China
| | - Huiting Cui
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| | - Jing Yuan
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, China.
| |
Collapse
|
4
|
Peng AYT, Li J, Freeman BC. Nuclear Type I Myosins are Essential for Life and Genome Organization. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.26.615191. [PMID: 39386516 PMCID: PMC11463430 DOI: 10.1101/2024.09.26.615191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The active transport of large biomolecules within a cell is critical for homeostasis. While the cytoplasmic process is well-studied, how the spacing of nucleoplasmic cargo is coordinated is poorly understood. We investigated the impact of myosin motors in the nucleus of budding yeast. We found that life requires a nuclear type I myosin whereas the essential type II or V myosins were not requisite in the nucleus. Nuclear depletion of type I myosins triggered 3D genome disorganization, nucleolar disruption, broad gene expression changes, and nuclear membrane morphology collapse. Genome disorganization occurred first supporting a model where type I myosins actively maintain genome architecture that scaffolds nuclear membrane and nucleolar morphologies. Overall, nuclear myosin is critical for the form and function of the nucleus.
Collapse
|
5
|
Huang J, Qiao Z, Yu H, Lu Z, Chen W, Lu J, Wu J, Bao Y, Shahid MQ, Liu X. OsRH52A, a DEAD-box protein, regulates functional megaspore specification and is required for embryo sac development in rice. JOURNAL OF EXPERIMENTAL BOTANY 2024; 75:4802-4821. [PMID: 38642102 PMCID: PMC11350083 DOI: 10.1093/jxb/erae180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/18/2024] [Indexed: 04/22/2024]
Abstract
The development of the embryo sac is an important factor that affects seed setting in rice. Numerous genes associated with embryo sac (ES) development have been identified in plants; however, the function of the DEAD-box RNA helicase family genes is poorly known in rice. Here, we characterized a rice DEAD-box protein, RH52A, which is localized in the nucleus and cytoplasm and highly expressed in the floral organs. The knockout mutant rh52a displayed partial ES sterility, including degeneration of the ES (21%) and the presence of a double-female-gametophyte (DFG) structure (11.8%). The DFG developed from two functional megaspores near the chalazal end in one ovule, and 3.4% of DFGs were able to fertilize via the sac near the micropylar pole in rh52a. RH52A was found to interact with MFS1 and ZIP4, both of which play a role in homologous recombination in rice meiosis. RNA-sequencing identified 234 down-regulated differentially expressed genes associated with reproductive development, including two, MSP1 and HSA1b, required for female germline cell specification. Taken together, our study demonstrates that RH52A is essential for the development of the rice embryo sac and provides cytological details regarding the formation of DFGs.
Collapse
Affiliation(s)
- Jinghua Huang
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Zhengping Qiao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Hang Yu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Zijun Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Weibin Chen
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Junming Lu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Jinwen Wu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Yueming Bao
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Muhammad Qasim Shahid
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Xiangdong Liu
- State Key Laboratory for Conservation and Utilization of Subtropical Agro-Bioresources, Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
- Guangdong Provincial Key Laboratory of Plant Molecular Breeding, South China Agricultural University, Guangzhou 510642, China
- Guangdong Base Bank for Lingnan Rice Germplasm Resources, College of Agriculture, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
6
|
Guo X, Huang CH, Akagi T, Niwa S, McKenney RJ, Wang JR, Lee YRJ, Liu B. An Arabidopsis Kinesin-14D motor is associated with midzone microtubules for spindle morphogenesis. Curr Biol 2024; 34:3747-3762.e6. [PMID: 39163829 PMCID: PMC11361718 DOI: 10.1016/j.cub.2024.07.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 05/19/2024] [Accepted: 07/03/2024] [Indexed: 08/22/2024]
Abstract
The acentrosomal spindle apparatus has kinetochore fibers organized and converged toward opposite poles; however, mechanisms underlying the organization of these microtubule fibers into an orchestrated bipolar array were largely unknown. Kinesin-14D is one of the four classes of Kinesin-14 motors that are conserved from green algae to flowering plants. In Arabidopsis thaliana, three Kinesin-14D members displayed distinct cell cycle-dependent localization patterns on spindle microtubules in mitosis. Notably, Kinesin-14D1 was enriched on the midzone microtubules of prophase and mitotic spindles and later persisted in the spindle and phragmoplast midzones. The kinesin-14d1 mutant had kinetochore fibers disengaged from each other during mitosis and exhibited hypersensitivity to the microtubule-depolymerizing herbicide oryzalin. Oryzalin-treated kinesin-14d1 mutant cells had kinetochore fibers tangled together in collapsed spindle microtubule arrays. Kinesin-14D1, unlike other Kinesin-14 motors, showed slow microtubule plus end-directed motility, and its localization and function were dependent on its motor activity and the novel malectin-like domain. Our findings revealed a Kinesin-14D1-dependent mechanism that employs interpolar microtubules to regulate the organization of kinetochore fibers for acentrosomal spindle morphogenesis.
Collapse
Affiliation(s)
- Xiaojiang Guo
- Department of Plant Biology, College of Biological Sciences, University of California, Davis, CA 95616, USA; State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Triticeae Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Calvin H Huang
- Department of Plant Biology, College of Biological Sciences, University of California, Davis, CA 95616, USA
| | - Takashi Akagi
- Graduate School of Environmental and Life Sciences, Okayama University, Okayama, Japan
| | - Shinsuke Niwa
- Frontier Research Institute for Interdisciplinary Sciences (FRIS), Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi 980-0845, Japan
| | - Richard J McKenney
- Department of Molecular and Cellular Biology, College of Biological Sciences, University of California, Davis, CA 95616, USA
| | - Ji-Rui Wang
- State Key Laboratory of Crop Gene Exploration and Utilization in Southwest China, Sichuan Agricultural University, Chengdu, Sichuan 611130, China; Triticeae Research Institute, Sichuan Agricultural University, Chengdu 611130, China
| | - Yuh-Ru Julie Lee
- Department of Plant Biology, College of Biological Sciences, University of California, Davis, CA 95616, USA
| | - Bo Liu
- Department of Plant Biology, College of Biological Sciences, University of California, Davis, CA 95616, USA.
| |
Collapse
|
7
|
Master K, El Khalki L, Bayachou M, Sossey-Alaoui K. Role of WAVE3 as an of actin binding protein in the pathology of triple negative breast cancer. Cytoskeleton (Hoboken) 2024. [PMID: 39021344 DOI: 10.1002/cm.21898] [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: 02/20/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Breast cancer, a prevalent global health concern, has sparked extensive research efforts, particularly focusing on triple negative breast cancer (TNBC), a subtype lacking estrogen receptor (ER), progesterone receptor, and epidermal growth factor receptor. TNBC's aggressive nature and resistance to hormone-based therapies heightens the risk of tumor progression and recurrence. Actin-binding proteins, specifically WAVE3 from the Wiskott-Aldrich syndrome protein (WASP) family, have emerged as major drivers in understanding TNBC biology. This review delves into the intricate molecular makeup of TNBC, shedding light on actin's fundamental role in cellular processes. Actin, a structural element in the cytoskeleton, regulates various cellular pathways essential for homeostasis. Its dynamic nature enables functions such as cell migration, motility, intracellular transport, cell division, and signal transduction. Actin-binding proteins, including WAVE3, play pivotal roles in these processes. WAVE3, a member of the WASP family, remains the focus of this review due to its potential involvement in TNBC progression. While actin-binding proteins are studied for their roles in healthy cellular cycles, their significance in TNBC remains underexplored. This review aims to discuss WAVE3's impact on TNBC, exploring its molecular makeup, functions, and significance in tumor progression. The intricate structure of WAVE3, featuring elements like the verprolin-cofilin-acidic domain and regulatory elements, plays a crucial role in regulating actin dynamics. Dysregulation of WAVE3 in TNBC has been linked to enhanced cell migration, invasion, extracellular matrix remodeling, epithelial-mesenchymal transition, tumor proliferation, and therapeutic resistance. Understanding the role of actin-binding proteins in cancer biology has potential clinical implications, making them potential prognostic biomarkers and promising therapeutic targets. The review emphasizes the need for further research into actin-binding proteins' clinical applications, diagnostic value, and therapeutic interventions. In conclusion, this comprehensive review explores the complex interplay between actin and actin-binding proteins, with special emphasis on WAVE3, in the context of TNBC. By unraveling the molecular intricacies, structural characteristics, and functional significance, the review paves the way for future research directions, clinical applications, and potential therapeutic strategies in the challenging landscape of TNBC.
Collapse
Affiliation(s)
- Kruyanshi Master
- Department of Chemistry, Cleveland State University, Cleveland, Ohio, USA
| | - Lamyae El Khalki
- MetroHealth System, Cleveland, Ohio, USA
- Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| | - Mekki Bayachou
- Department of Chemistry, Cleveland State University, Cleveland, Ohio, USA
| | - Khalid Sossey-Alaoui
- MetroHealth System, Cleveland, Ohio, USA
- Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| |
Collapse
|
8
|
Seo D, Yue Y, Yamazaki S, Verhey KJ, Gammon DB. Poxvirus A51R Proteins Negatively Regulate Microtubule-Dependent Transport by Kinesin-1. Int J Mol Sci 2024; 25:7825. [PMID: 39063067 PMCID: PMC11277487 DOI: 10.3390/ijms25147825] [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: 06/05/2024] [Revised: 07/09/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Microtubule (MT)-dependent transport is a critical means of intracellular movement of cellular cargo by kinesin and dynein motors. MT-dependent transport is tightly regulated by cellular MT-associated proteins (MAPs) that directly bind to MTs and either promote or impede motor protein function. Viruses have been widely shown to usurp MT-dependent transport to facilitate their virion movement to sites of replication and/or for exit from the cell. However, it is unclear if viruses also negatively regulate MT-dependent transport. Using single-molecule motility and cellular transport assays, we show that the vaccinia virus (VV)-encoded MAP, A51R, inhibits kinesin-1-dependent transport along MTs in vitro and in cells. This inhibition is selective as the function of kinesin-3 is largely unaffected by VV A51R. Interestingly, we show that A51R promotes the perinuclear accumulation of cellular cargo transported by kinesin-1 such as lysosomes and mitochondria during infection. Moreover, A51R also regulates the release of specialized VV virions that exit the cell using kinesin-1-dependent movement. Using a fluorescently tagged rigor mutant of kinesin-1, we show that these motors accumulate on A51R-stabilized MTs, suggesting these stabilized MTs may form a "kinesin-1 sink" to regulate MT-dependent transport in the cell. Collectively, our findings uncover a new mechanism by which viruses regulate host cytoskeletal processes.
Collapse
Affiliation(s)
- Dahee Seo
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yang Yue
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Shin Yamazaki
- Department of Neuroscience and Peter O’Donnell Jr. Brain Institute, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kristen J. Verhey
- Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Don B. Gammon
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
9
|
Wang L, Sheth V, Liu K, Panja P, Frickenstein AN, He Y, Yang W, Thomas AG, Jamei MH, Park J, Lyu S, Donahue ND, Chen WR, Bhattacharya R, Mukherjee P, Wilhelm S. Primary Human Breast Cancer-Associated Endothelial Cells Favor Interactions with Nanomedicines. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403986. [PMID: 38663008 PMCID: PMC11239290 DOI: 10.1002/adma.202403986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 04/19/2024] [Indexed: 05/04/2024]
Abstract
Cancer nanomedicines predominately rely on transport processes controlled by tumor-associated endothelial cells to deliver therapeutic and diagnostic payloads into solid tumors. While the dominant role of this class of endothelial cells for nanoparticle transport and tumor delivery is established in animal models, the translational potential in human cells needs exploration. Using primary human breast cancer as a model, the differential interactions of normal and tumor-associated endothelial cells with clinically relevant nanomedicine formulations are explored and quantified. Primary human breast cancer-associated endothelial cells exhibit up to ≈2 times higher nanoparticle uptake than normal human mammary microvascular endothelial cells. Super-resolution imaging studies reveal a significantly higher intracellular vesicle number for tumor-associated endothelial cells, indicating a substantial increase in cellular transport activities. RNA sequencing and gene expression analysis indicate the upregulation of transport-related genes, especially motor protein genes, in tumor-associated endothelial cells. Collectively, the results demonstrate that primary human breast cancer-associated endothelial cells exhibit enhanced interactions with nanomedicines, suggesting a potentially significant role for these cells in nanoparticle tumor delivery in human patients. Engineering nanoparticles that leverage the translational potential of tumor-associated endothelial cell-mediated transport into human solid tumors may lead to the development of safer and more effective clinical cancer nanomedicines.
Collapse
Affiliation(s)
- Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Vinit Sheth
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Prasanta Panja
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Alex N Frickenstein
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Yuxin He
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Wen Yang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Abigail G Thomas
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Mohammad Hasan Jamei
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Jeesoo Park
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Shanxin Lyu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Nathan D Donahue
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Priyabrata Mukherjee
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Stefan Wilhelm
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, 73019, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
- Institute for Biomedical Engineering, Science and Technology (IBEST), Norman, OK, 73019, USA
| |
Collapse
|
10
|
Xie C, Chen G, Li M, Huang P, Chen Z, Lei K, Li D, Wang Y, Cleetus A, Mohamed MA, Sonar P, Feng W, Ökten Z, Ou G. Neurons dispose of hyperactive kinesin into glial cells for clearance. EMBO J 2024; 43:2606-2635. [PMID: 38806659 PMCID: PMC11217292 DOI: 10.1038/s44318-024-00118-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 04/06/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Microtubule-based kinesin motor proteins are crucial for intracellular transport, but their hyperactivation can be detrimental for cellular functions. This study investigated the impact of a constitutively active ciliary kinesin mutant, OSM-3CA, on sensory cilia in C. elegans. Surprisingly, we found that OSM-3CA was absent from cilia but underwent disposal through membrane abscission at the tips of aberrant neurites. Neighboring glial cells engulf and eliminate the released OSM-3CA, a process that depends on the engulfment receptor CED-1. Through genetic suppressor screens, we identified intragenic mutations in the OSM-3CA motor domain and mutations inhibiting the ciliary kinase DYF-5, both of which restored normal cilia in OSM-3CA-expressing animals. We showed that conformational changes in OSM-3CA prevent its entry into cilia, and OSM-3CA disposal requires its hyperactivity. Finally, we provide evidence that neurons also dispose of hyperactive kinesin-1 resulting from a clinic variant associated with amyotrophic lateral sclerosis, suggesting a widespread mechanism for regulating hyperactive kinesins.
Collapse
Affiliation(s)
- Chao Xie
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing, China
- State Key Laboratory for Membrane Biology, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Guanghan Chen
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing, China
- State Key Laboratory for Membrane Biology, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Ming Li
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing, China
- State Key Laboratory for Membrane Biology, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Peng Huang
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing, China
- State Key Laboratory for Membrane Biology, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Zhe Chen
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing, China
- State Key Laboratory for Membrane Biology, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Kexin Lei
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing, China
- State Key Laboratory for Membrane Biology, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Dong Li
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, 100101, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Yuhe Wang
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China
- McGovern Institute for Brain Research, Tsinghua University, Beijing, China
- State Key Laboratory for Membrane Biology, Beijing, China
- School of Life Sciences, Tsinghua University, Beijing, China
| | - Augustine Cleetus
- Physik Department E22, Technische Universitat Munchen, James-Franck-Strasse, Garching, 85748, Germany
| | - Mohamed Aa Mohamed
- Physik Department E22, Technische Universitat Munchen, James-Franck-Strasse, Garching, 85748, Germany
| | - Punam Sonar
- Physik Department E22, Technische Universitat Munchen, James-Franck-Strasse, Garching, 85748, Germany
| | - Wei Feng
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, 15 Datun Road, 100101, Beijing, China
- College of Life Sciences, University of Chinese Academy of Sciences, 100049, Beijing, China
| | - Zeynep Ökten
- Physik Department E22, Technische Universitat Munchen, James-Franck-Strasse, Garching, 85748, Germany
| | - Guangshuo Ou
- Tsinghua-Peking Center for Life Sciences, Tsinghua University, Beijing, China.
- Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing, China.
- McGovern Institute for Brain Research, Tsinghua University, Beijing, China.
- State Key Laboratory for Membrane Biology, Beijing, China.
- School of Life Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
11
|
Gao H, Tian Y, Zhang H, Li Y, Li C, Li B. Species-specific duplicated FMRFaR-like gene A62 regulates spontaneous locomotion in Apolygus lucorum. PEST MANAGEMENT SCIENCE 2024; 80:3358-3368. [PMID: 38385791 DOI: 10.1002/ps.8039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/05/2024] [Accepted: 02/22/2024] [Indexed: 02/23/2024]
Abstract
BACKGROUND Apolygus lucorum, a major cotton pest, has undergone a significant expansion of the FMRFaR gene within the GPCR superfamily, resulting in two classes of GPCR, namely FMRFaR (A54-55) and newly duplicated FMRFaR-like (A56-62). Notably, FMRFaR-like genes, particularly A62, show enhanced expression in the legs and wings of adults, indicating their potential role in locomotion. Employing A62 as a representative of FMRFaR-like, our study investigates the influence of FMRFa, FMRFaR, and FMRFaR-like on locomotion and development of A. lucorum. RESULTS FMRFaR and FMRFa exhibit comparable temporal and tissue expression patterns, whereas the FMRFaR-like genes within A. lucorum exhibit completely distinct evolutionary and expression patterns compared to classical FMRFaR. RNA interference (RNAi) experiments revealed that suppressing FMRFa expression results in complete lethality in A. lucorum, but neither FMRFaR nor A62 exhibit the same effect after RNAi. Suppressing the expression of FMRFa only decreases the expression of the A54 gene simultaneously, suggesting that A54 may function as a classical FMRFaR activated by FMRFa. RNAi of A62 leads to wing malformation and a significant reduction in spontaneous movement behavior in A. lucorum. Further transcriptomic analysis revealed that A62 affects the A. lucorum's movement behavior through energy metabolism pathways and motor protein pathways. CONCLUSION Our study unveils the unique and complex roles of FMRFa and its receptor in A. lucorum. These findings provide valuable insights into potential targets for pest control strategies aimed at managing A. lucorum populations in cotton fields. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Han Gao
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Ying Tian
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Hui Zhang
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Yanxiao Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Chenjun Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Bin Li
- Jiangsu Key Laboratory for Biodiversity and Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
12
|
Sun Y, Tao X, Han Y, Lin X, Tian R, Wang H, Chang P, Sun Q, Ge L, Zhang M. A dual role of ERGIC-localized Rabs in TMED10-mediated unconventional protein secretion. Nat Cell Biol 2024; 26:1077-1092. [PMID: 38926505 DOI: 10.1038/s41556-024-01445-4] [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: 11/22/2023] [Accepted: 05/23/2024] [Indexed: 06/28/2024]
Abstract
Cargo translocation across membranes is a crucial aspect of secretion. In conventional secretion signal peptide-equipped proteins enter the endoplasmic reticulum (ER), whereas a subset of cargo lacking signal peptides translocate into the ER-Golgi intermediate compartment (ERGIC) in a process called unconventional protein secretion (UcPS). The regulatory events at the ERGIC in UcPS are unclear. Here we reveal the involvement of ERGIC-localized small GTPases, Rab1 (Rab1A and Rab1B) and Rab2A, in regulating UcPS cargo transport via TMED10 on the ERGIC. Rab1 enhances TMED10 translocator activity, promoting cargo translocation into the ERGIC, whereas Rab2A, in collaboration with KIF5B, regulates ERGIC compartmentalization, establishing a UcPS-specific compartment. This study highlights the pivotal role of ERGIC-localized Rabs in governing cargo translocation and specifying the ERGIC's function in UcPS.
Collapse
Affiliation(s)
- Yuxin Sun
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xuan Tao
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Yaping Han
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Xubo Lin
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
| | - Rui Tian
- Department of Biochemistry and Department of Cardiology of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Haodong Wang
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Pei Chang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China
| | - Qiming Sun
- Department of Biochemistry and Department of Cardiology of Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Ge
- State Key Laboratory of Membrane Biology, Tsinghua University-Peking University Joint Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China.
| | - Min Zhang
- State Key Laboratory of Membrane Biology, School of Pharmaceutical Sciences, Tsinghua University, Beijing, China.
| |
Collapse
|
13
|
Chakraborti A, Tardiff JC, Schwartz SD. Myosin-Catalyzed ATP Hydrolysis in the Presence of Disease-Causing Mutations: Mavacamten as a Way to Repair Mechanism. J Phys Chem B 2024; 128:4716-4727. [PMID: 38708944 PMCID: PMC11103257 DOI: 10.1021/acs.jpcb.4c01601] [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] [Indexed: 05/07/2024]
Abstract
Hypertrophic cardiomyopathy is one of the most common forms of genetic cardiomyopathy. Mavacamten is a first-in-class myosin modulator that was identified via activity screening on the wild type, and it is FDA-approved for the treatment of obstructive hypertrophic cardiomyopathy (HCM). The drug selectively binds to the cardiac β-myosin, inhibiting myosin function to decrease cardiac contractility. Though the drug is thought to affect multiple steps of the myosin cross-bridge cycle, its detailed mechanism of action is still under investigation. Individual steps in the overall cross-bridge cycle must be queried to elucidate the full mechanism of action. In this study, we utilize the rare-event method of transition path sampling to generate reactive trajectories to gain insights into the action of the drug on the dynamics and rate of the ATP hydrolysis step for human cardiac β-myosin. We study three known HCM causative myosin mutations: R453C, P710R, and R712L to observe the effect of the drug on the alterations caused by these mutations in the chemical step. Since the crystal structure of the drug-bound myosin was not available at the time of this work, we created a model of the drug-bound system utilizing a molecular docking approach. We find a significant effect of the drug in one case, where the actual mechanism of the reaction is altered from the wild type by mutation. The drug restores both the rate of hydrolysis to the wildtype level and the mechanism of the reaction. This is a way to check the effect of the drug on untested mutations.
Collapse
Affiliation(s)
- Ananya Chakraborti
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| | - Jil C Tardiff
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona 85724, United States
| | - Steven D Schwartz
- Department of Chemistry and Biochemistry, University of Arizona, Tucson, Arizona 85721, United States
| |
Collapse
|
14
|
Tell I Puig A, Soldati-Favre D. Roles of the tubulin-based cytoskeleton in the Toxoplasma gondii apical complex. Trends Parasitol 2024; 40:401-415. [PMID: 38531711 DOI: 10.1016/j.pt.2024.02.010] [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: 01/08/2024] [Revised: 02/26/2024] [Accepted: 02/28/2024] [Indexed: 03/28/2024]
Abstract
Microtubules (MTs) play a vital role as key components of the eukaryotic cytoskeleton. The phylum Apicomplexa comprises eukaryotic unicellular parasitic organisms defined by the presence of an apical complex which consists of specialized secretory organelles and tubulin-based cytoskeletal elements. One apicomplexan parasite, Toxoplasma gondii, is an omnipresent opportunistic pathogen with significant medical and veterinary implications. To ensure successful infection and widespread dissemination, T. gondii heavily relies on the tubulin structures present in the apical complex. Recent advances in high-resolution imaging, coupled with reverse genetics, have offered deeper insights into the composition, functionality, and dynamics of these tubulin-based structures. The apicomplexan tubulins differ from those of their mammalian hosts, endowing them with unique attributes and susceptibility to specific classes of inhibitory compounds.
Collapse
Affiliation(s)
- Albert Tell I Puig
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva, Switzerland.
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, CMU, University of Geneva, Geneva, Switzerland
| |
Collapse
|
15
|
Golyshev SA, Lyupina YV, Kravchuk OI, Mikhailov KV, Gornostaev NG, Burakov AV. Transient Interphase Microtubules Appear in Differentiating Sponge Cells. Cells 2024; 13:736. [PMID: 38727272 PMCID: PMC11082956 DOI: 10.3390/cells13090736] [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: 02/11/2024] [Revised: 04/09/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
Microtubules are an indispensable component of all eukaryotic cells due to their role in mitotic spindle formation, yet their organization and number can vary greatly in the interphase. The last common ancestor of all eukaryotes already had microtubules and microtubule motor proteins moving along them. Sponges are traditionally regarded as the oldest animal phylum. Their body does not have a clear differentiation into tissues, but it contains several distinguishable cell types. The choanocytes stand out among them and are responsible for creating a flow of water with their flagella and increasing the filtering and feeding efficiency of the sponge. Choanocyte flagella contain microtubules, but thus far, observing a developed system of cytoplasmic microtubules in non-flagellated interphase sponge cells has been mostly unsuccessful. In this work, we combine transcriptomic analysis, immunofluorescence, and electron microscopy with time-lapse recording to demonstrate that microtubules appear in the cytoplasm of sponge cells only when transdifferentiation processes are activated. We conclude that dynamic cytoplasmic microtubules in the cells of sponges are not a persistent but rather a transient structure, associated with cellular plasticity.
Collapse
Affiliation(s)
- Sergei A. Golyshev
- A.N. Belozersky Institute of Physical and Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (S.A.G.); (K.V.M.)
| | - Yulia V. Lyupina
- N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia; (Y.V.L.); (O.I.K.); (N.G.G.)
| | - Oksana I. Kravchuk
- N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia; (Y.V.L.); (O.I.K.); (N.G.G.)
| | - Kirill V. Mikhailov
- A.N. Belozersky Institute of Physical and Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (S.A.G.); (K.V.M.)
- Kharkevich Institute for Information Transmission Problems, Russian Academy of Sciences, Moscow 127051, Russia
| | - Nicolay G. Gornostaev
- N.K. Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow 119334, Russia; (Y.V.L.); (O.I.K.); (N.G.G.)
| | - Anton V. Burakov
- A.N. Belozersky Institute of Physical and Chemical Biology, Lomonosov Moscow State University, Moscow 119992, Russia; (S.A.G.); (K.V.M.)
| |
Collapse
|
16
|
Li Y, Yang Z, Zhang S, Li J. Miro-mediated mitochondrial transport: A new dimension for disease-related abnormal cell metabolism? Biochem Biophys Res Commun 2024; 705:149737. [PMID: 38430606 DOI: 10.1016/j.bbrc.2024.149737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 02/15/2024] [Accepted: 02/27/2024] [Indexed: 03/05/2024]
Abstract
Mitochondria are versatile and highly dynamic organelles found in eukaryotic cells that play important roles in a variety of cellular processes. The importance of mitochondrial transport in cell metabolism, including variations in mitochondrial distribution within cells and intercellular transfer, has grown in recent years. Several studies have demonstrated that abnormal mitochondrial transport represents an early pathogenic alteration in a variety of illnesses, emphasizing its significance in disease development and progression. Mitochondrial Rho GTPase (Miro) is a protein found on the outer mitochondrial membrane that is required for cytoskeleton-dependent mitochondrial transport, mitochondrial dynamics (fusion and fission), and mitochondrial Ca2+ homeostasis. Miro, as a critical regulator of mitochondrial transport, has yet to be thoroughly investigated in illness. This review focuses on recent developments in recognizing Miro as a crucial molecule in controlling mitochondrial transport and investigates its roles in diverse illnesses. It also intends to shed light on the possibilities of targeting Miro as a therapeutic method for a variety of diseases.
Collapse
Affiliation(s)
- Yanxing Li
- Xi'an Jiaotong University Health Science Center, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Zhen Yang
- Xi'an Jiaotong University Health Science Center, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Shumei Zhang
- Xi'an Jiaotong University Health Science Center, Xi'an, 710000, Shaanxi, People's Republic of China
| | - Jianjun Li
- Department of Cardiology, Jincheng People's Hospital Affiliated to Changzhi Medical College, Jincheng, Shanxi, People's Republic of China.
| |
Collapse
|
17
|
Wang L, Bu T, Wu X, Li L, Sun F, Cheng CY. Motor proteins, spermatogenesis and testis function. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 141:381-445. [PMID: 38960481 DOI: 10.1016/bs.apcsb.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/05/2024]
Abstract
The role of motor proteins in supporting intracellular transports of vesicles and organelles in mammalian cells has been known for decades. On the other hand, the function of motor proteins that support spermatogenesis is also well established since the deletion of motor protein genes leads to subfertility and/or infertility. Furthermore, mutations and genetic variations of motor protein genes affect fertility in men, but also a wide range of developmental defects in humans including multiple organs besides the testis. In this review, we seek to provide a summary of microtubule and actin-dependent motor proteins based on earlier and recent findings in the field. Since these two cytoskeletons are polarized structures, different motor proteins are being used to transport cargoes to different ends of these cytoskeletons. However, their involvement in germ cell transport across the blood-testis barrier (BTB) and the epithelium of the seminiferous tubules remains relatively unknown. It is based on recent findings in the field, we have provided a hypothetical model by which motor proteins are being used to support germ cell transport across the BTB and the seminiferous epithelium during the epithelial cycle of spermatogenesis. In our discussion, we have highlighted the areas of research that deserve attention to bridge the gap of research in relating the function of motor proteins to spermatogenesis.
Collapse
Affiliation(s)
- Lingling Wang
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Tiao Bu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Xiaolong Wu
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - Linxi Li
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China
| | - Fei Sun
- Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China
| | - C Yan Cheng
- The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, P.R. China; Department of Urology and Andrology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, P.R. China.
| |
Collapse
|
18
|
Morales PN, Coons AN, Koopman AJ, Patel S, Chase PB, Parvatiyar MS, Pinto JR. Post-translational modifications of vertebrate striated muscle myosin heavy chains. Cytoskeleton (Hoboken) 2024:10.1002/cm.21857. [PMID: 38587113 PMCID: PMC11458826 DOI: 10.1002/cm.21857] [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: 12/11/2023] [Revised: 03/06/2024] [Accepted: 03/25/2024] [Indexed: 04/09/2024]
Abstract
Post-translational modifications (PTMs) play a crucial role in regulating the function of many sarcomeric proteins, including myosin. Myosins comprise a family of motor proteins that play fundamental roles in cell motility in general and muscle contraction in particular. A myosin molecule consists of two myosin heavy chains (MyHCs) and two pairs of myosin light chains (MLCs); two MLCs are associated with the neck region of each MyHC's N-terminal head domain, while the two MyHC C-terminal tails form a coiled-coil that polymerizes with other MyHCs to form the thick filament backbone. Myosin undergoes extensive PTMs, and dysregulation of these PTMs may lead to abnormal muscle function and contribute to the development of myopathies and cardiovascular disorders. Recent studies have uncovered the significance of PTMs in regulating MyHC function and showed how these PTMs may provide additional modulation of contractile processes. Here, we discuss MyHC PTMs that have been biochemically and/or functionally studied in mammals' and rodents' striated muscle. We have identified hotspots or specific regions in three isoforms of myosin (MYH2, MYH6, and MYH7) where the prevalence of PTMs is more frequent and could potentially play a significant role in fine-tuning the activity of these proteins.
Collapse
Affiliation(s)
- Paula Nieto Morales
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306 USA
| | - Arianna N. Coons
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Amelia J. Koopman
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Sonu Patel
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - P. Bryant Chase
- Department of Biological Science, Florida State University, Tallahassee, FL 32306 USA
| | - Michelle S. Parvatiyar
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306 USA
| | - Jose R. Pinto
- Department of Biomedical Sciences, Florida State University College of Medicine, Tallahassee, FL 32306 USA
| |
Collapse
|
19
|
Shadab M, Abbasi AA, Ejaz A, Ben-Mahmoud A, Gupta V, Kim HG, Vona B. Autosomal recessive non-syndromic hearing loss genes in Pakistan during the previous three decades. J Cell Mol Med 2024; 28:e18119. [PMID: 38534090 DOI: 10.1111/jcmm.18119] [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: 06/25/2023] [Revised: 11/29/2023] [Accepted: 01/02/2024] [Indexed: 03/28/2024] Open
Abstract
Hearing loss is a clinically and genetically heterogeneous disorder, with over 148 genes and 170 loci associated with its pathogenesis. The spectrum and frequency of causal variants vary across different genetic ancestries and are more prevalent in populations that practice consanguineous marriages. Pakistan has a rich history of autosomal recessive gene discovery related to non-syndromic hearing loss. Since the first linkage analysis with a Pakistani family that led to the mapping of the DFNB1 locus on chromosome 13, 51 genes associated with this disorder have been identified in this population. Among these, 13 of the most prevalent genes, namely CDH23, CIB2, CLDN14, GJB2, HGF, MARVELD2, MYO7A, MYO15A, MSRB3, OTOF, SLC26A4, TMC1 and TMPRSS3, account for more than half of all cases of profound hearing loss, while the prevalence of other genes is less than 2% individually. In this review, we discuss the most common autosomal recessive non-syndromic hearing loss genes in Pakistani individuals as well as the genetic mapping and sequencing approaches used to discover them. Furthermore, we identified enriched gene ontology terms and common pathways involved in these 51 autosomal recessive non-syndromic hearing loss genes to gain a better understanding of the underlying mechanisms. Establishing a molecular understanding of the disorder may aid in reducing its future prevalence by enabling timely diagnostics and genetic counselling, leading to more effective clinical management and treatments of hearing loss.
Collapse
Affiliation(s)
- Madiha Shadab
- Department of Zoology, Mirpur University of Science and Technology, Mirpur, Pakistan
| | - Ansar Ahmed Abbasi
- Department of Zoology, Mirpur University of Science and Technology, Mirpur, Pakistan
| | - Ahsan Ejaz
- Department of Physics, University of Kotli Azad Jammu and Kashmir, Kotli, Pakistan
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou, China
| | - Afif Ben-Mahmoud
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Vijay Gupta
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
| | - Hyung-Goo Kim
- Neurological Disorders Research Center, Qatar Biomedical Research Institute, Hamad Bin Khalifa University, Doha, Qatar
- College of Health & Life Sciences, Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Barbara Vona
- Institute of Human Genetics, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and Inner Ear Lab, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
20
|
Miyoshi T, Belyantseva IA, Sajeevadathan M, Friedman TB. Pathophysiology of human hearing loss associated with variants in myosins. Front Physiol 2024; 15:1374901. [PMID: 38562617 PMCID: PMC10982375 DOI: 10.3389/fphys.2024.1374901] [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: 01/24/2024] [Accepted: 02/21/2024] [Indexed: 04/04/2024] Open
Abstract
Deleterious variants of more than one hundred genes are associated with hearing loss including MYO3A, MYO6, MYO7A and MYO15A and two conventional myosins MYH9 and MYH14. Variants of MYO7A also manifest as Usher syndrome associated with dysfunction of the retina and vestibule as well as hearing loss. While the functions of MYH9 and MYH14 in the inner ear are debated, MYO3A, MYO6, MYO7A and MYO15A are expressed in inner ear hair cells along with class-I myosin MYO1C and are essential for developing and maintaining functional stereocilia on the apical surface of hair cells. Stereocilia are large, cylindrical, actin-rich protrusions functioning as biological mechanosensors to detect sound, acceleration and posture. The rigidity of stereocilia is sustained by highly crosslinked unidirectionally-oriented F-actin, which also provides a scaffold for various proteins including unconventional myosins and their cargo. Typical myosin molecules consist of an ATPase head motor domain to transmit forces to F-actin, a neck containing IQ-motifs that bind regulatory light chains and a tail region with motifs recognizing partners. Instead of long coiled-coil domains characterizing conventional myosins, the tails of unconventional myosins have various motifs to anchor or transport proteins and phospholipids along the F-actin core of a stereocilium. For these myosins, decades of studies have elucidated their biochemical properties, interacting partners in hair cells and variants associated with hearing loss. However, less is known about how myosins traffic in a stereocilium using their motor function, and how each variant correlates with a clinical condition including the severity and onset of hearing loss, mode of inheritance and presence of symptoms other than hearing loss. Here, we cover the domain structures and functions of myosins associated with hearing loss together with advances, open questions about trafficking of myosins in stereocilia and correlations between hundreds of variants in myosins annotated in ClinVar and the corresponding deafness phenotypes.
Collapse
Affiliation(s)
- Takushi Miyoshi
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
- Division of Molecular and Integrative Physiology, Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Inna A. Belyantseva
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - Mrudhula Sajeevadathan
- Division of Molecular and Integrative Physiology, Department of Biomedical Sciences, Southern Illinois University School of Medicine, Carbondale, IL, United States
| | - Thomas B. Friedman
- Laboratory of Molecular Genetics, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
21
|
Chae SJ, Kim DW, Igoshin OA, Lee S, Kim JK. Beyond microtubules: The cellular environment at the endoplasmic reticulum attracts proteins to the nucleus, enabling nuclear transport. iScience 2024; 27:109235. [PMID: 38439967 PMCID: PMC10909898 DOI: 10.1016/j.isci.2024.109235] [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: 08/17/2023] [Revised: 01/03/2024] [Accepted: 02/09/2024] [Indexed: 03/06/2024] Open
Abstract
All proteins are translated in the cytoplasm, yet many, including transcription factors, play vital roles in the nucleus. While previous research has concentrated on molecular motors for the transport of these proteins to the nucleus, recent observations reveal perinuclear accumulation even in the absence of an energy source, hinting at alternative mechanisms. Here, we propose that structural properties of the cellular environment, specifically the endoplasmic reticulum (ER), can promote molecular transport to the perinucleus without requiring additional energy expenditure. Specifically, physical interaction between proteins and the ER impedes their diffusion and leads to their accumulation near the nucleus. This result explains why larger proteins, more frequently interacting with the ER membrane, tend to accumulate at the perinucleus. Interestingly, such diffusion in a heterogeneous environment follows Chapman's law rather than the popular Fick's law. Our findings suggest a novel protein transport mechanism arising solely from characteristics of the intracellular environment.
Collapse
Affiliation(s)
- Seok Joo Chae
- Department of Mathematical Sciences, KAIST, Daejeon 34141, Republic of Korea
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon 34126, Republic of Korea
| | - Dae Wook Kim
- Department of Mathematics, University of Michigan, Ann Arbor, MI 48109, USA
| | - Oleg A. Igoshin
- Department of Bioengineering, Rice University, Houston, TX 77005, USA
- Center for Theoretical Biological Physics, Rice University, Houston, TX 77005, USA
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Department of Biosciences, Rice University, Houston, TX 77005, USA
| | - Seunggyu Lee
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon 34126, Republic of Korea
- Division of Applied Mathematical Sciences, Korea University, Sejong 30019, Republic of Korea
| | - Jae Kyoung Kim
- Department of Mathematical Sciences, KAIST, Daejeon 34141, Republic of Korea
- Biomedical Mathematics Group, Pioneer Research Center for Mathematical and Computational Sciences, Institute for Basic Science, Daejeon 34126, Republic of Korea
| |
Collapse
|
22
|
Danziger M, Noble H, Roque DM, Xu F, Rao GG, Santin AD. Microtubule-Targeting Agents: Disruption of the Cellular Cytoskeleton as a Backbone of Ovarian Cancer Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1452:1-19. [PMID: 38805122 DOI: 10.1007/978-3-031-58311-7_1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Microtubules are dynamic polymers composed of α- and β-tubulin heterodimers. Microtubules are universally conserved among eukaryotes and participate in nearly every cellular process, including intracellular trafficking, replication, polarity, cytoskeletal shape, and motility. Due to their fundamental role in mitosis, they represent a classic target of anti-cancer therapy. Microtubule-stabilizing agents currently constitute a component of the most effective regimens for ovarian cancer therapy in both primary and recurrent settings. Unfortunately, the development of resistance continues to present a therapeutic challenge. An understanding of the underlying mechanisms of resistance to microtubule-active agents may facilitate the development of novel and improved approaches to this disease.
Collapse
Affiliation(s)
- Michael Danziger
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Helen Noble
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Dana M Roque
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Fuhua Xu
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Gautam G Rao
- Division of Gynecologic Oncology, Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD, USA
| | | |
Collapse
|
23
|
Jang S, Engelman AN. Capsid-host interactions for HIV-1 ingress. Microbiol Mol Biol Rev 2023; 87:e0004822. [PMID: 37750702 PMCID: PMC10732038 DOI: 10.1128/mmbr.00048-22] [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: 09/27/2023] Open
Abstract
The HIV-1 capsid, composed of approximately 1,200 copies of the capsid protein, encases genomic RNA alongside viral nucleocapsid, reverse transcriptase, and integrase proteins. After cell entry, the capsid interacts with a myriad of host factors to traverse the cell cytoplasm, pass through the nuclear pore complex (NPC), and then traffic to chromosomal sites for viral DNA integration. Integration may very well require the dissolution of the capsid, but where and when this uncoating event occurs remains hotly debated. Based on size constraints, a long-prevailing view was that uncoating preceded nuclear transport, but recent research has indicated that the capsid may remain largely intact during nuclear import, with perhaps some structural remodeling required for NPC traversal. Completion of reverse transcription in the nucleus may further aid capsid uncoating. One canonical type of host factor, typified by CPSF6, leverages a Phe-Gly (FG) motif to bind capsid. Recent research has shown these peptides reside amid prion-like domains (PrLDs), which are stretches of protein sequence devoid of charged residues. Intermolecular PrLD interactions along the exterior of the capsid shell impart avid host factor binding for productive HIV-1 infection. Herein we overview capsid-host interactions implicated in HIV-1 ingress and discuss important research questions moving forward. Highlighting clinical relevance, the long-acting ultrapotent inhibitor lenacapavir, which engages the same capsid binding pocket as FG host factors, was recently approved to treat people living with HIV.
Collapse
Affiliation(s)
- Sooin Jang
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Alan N. Engelman
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Medler S. Graham Hoyle (1923-1985): exploring the depths of muscle diversity. ADVANCES IN PHYSIOLOGY EDUCATION 2023; 47:893-903. [PMID: 37823190 DOI: 10.1152/advan.00098.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/25/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
Graham Hoyle was an important neuroscientist, muscle biologist, and zoologist throughout much of the second half of the twentieth century. A native of England, Hoyle studied under Bernard Katz in London before earning his D.Sc. in neurophysiology from the University of Glasgow. He immigrated to the United States in the mid-1950s and worked with C.A.G. Wiersma at Caltech, with whom he shared a love for crustacean neuromuscular physiology. Hoyle accepted a position at the University of Oregon in 1961 and remained there as a professor until his death in 1985 at the age of 61. Hoyle was active scientifically at a time when the basics of muscle biology were still being discovered. He made many important contributions to the field of neuromuscular physiology, particularly in the realm of comparative physiology. Hoyle was passionate about the importance of a comparative approach in physiology and emphasized that "as a comparative physiologist, I value knowledge of the diverse forms not only for its own sake, but also because it embodies the general truth." Perhaps Hoyle's most lasting legacy is embodied in the many students and postdocs who trained with him early in their careers. Many of these young scientists went on to build prominent careers and trained numerous students of their own. In addition to offering an overview of Hoyle's career, this article revisits some of Hoyle's central contributions to muscle biology and assesses them in light of our current understanding of muscle structure and function.NEW & NOTEWORTHY Graham Hoyle was an important neuroscientist, muscle biologist, and zoologist throughout much of the second half of the twentieth century. He was trained by Bernard Katz at University College London and later worked with C.A.G. Wiersma at Caltech. As a professor at the University of Oregon, Hoyle helped found the Institute of Neuroscience and trained many prominent scientists in the fields of neuromuscular biology and neuroethology.
Collapse
Affiliation(s)
- Scott Medler
- Dennis R. DePerro School of Health Professions, St. Bonaventure University, St. Bonaventure, New York, United States
| |
Collapse
|
25
|
Hall D. HSAFM-MIREBA - Methodology for Inferring REsolution in biological applications. Anal Biochem 2023; 681:115320. [PMID: 37717838 DOI: 10.1016/j.ab.2023.115320] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 08/22/2023] [Accepted: 09/10/2023] [Indexed: 09/19/2023]
Abstract
Due to a lack of requirement for any direct labelling of the target molecule, high speed atomic force microscopy (HS-AFM) is a potentially powerful procedure for the assessment of biological processes involving macromolecules. When the sample is static the AFM device can be purposefully setup to recover high-resolution information about the feature in question. However, when the feature to be studied moves an appreciable amount during the course of the measurement, the obtained image will be blurred. Encountering such blurred observations prompts the experimenter to sacrifice higher resolution images for higher scanning speeds by tuning available experimental parameters (such as the scanned image area, the image pixel size, the resonance frequency of the cantilever and/or the diameter of the AFM tip). The present work describes a software tool, HSAFM-MIREBA (High Speed Atomic Force Microscopy - Methodology for Inferring REsolution in Biological Applications) that allows for pre-experimental optimization of such parameters through iterative rounds of simulation of both the dynamic surface process and the HS-AFM measurement (based on the particular set of governing parameters). A representative set of five dynamic biological processes that describe a range of diffusive and directed motions (which can themselves be tuned by altering characteristic governing parameter sets) are provided.
Collapse
Affiliation(s)
- Damien Hall
- WPI Nano Life Science Institute. Kanazawa University, Kakumamachi, Kanazawa, Ishikawa, 920-1164, Japan.
| |
Collapse
|
26
|
Jiang X, Ogawa T, Yonezawa K, Shimizu N, Ichinose S, Uchihashi T, Nagaike W, Moriya T, Adachi N, Kawasaki M, Dohmae N, Senda T, Hirokawa N. The two-step cargo recognition mechanism of heterotrimeric kinesin. EMBO Rep 2023; 24:e56864. [PMID: 37575008 PMCID: PMC10626431 DOI: 10.15252/embr.202356864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/21/2023] [Accepted: 08/03/2023] [Indexed: 08/15/2023] Open
Abstract
Kinesin-driven intracellular transport is essential for various cell biological events and thus plays a crucial role in many pathological processes. However, little is known about the molecular basis of the specific and dynamic cargo-binding mechanism of kinesins. Here, an integrated structural analysis of the KIF3/KAP3 and KIF3/KAP3-APC complexes unveils the mechanism by which KIF3/KAP3 can dynamically grasp APC in a two-step manner, which suggests kinesin-cargo recognition dynamics composed of cargo loading, locking, and release. Our finding is the first demonstration of the two-step cargo recognition and stabilization mechanism of kinesins, which provides novel insights into the intracellular trafficking machinery.
Collapse
Affiliation(s)
- Xuguang Jiang
- Department of Cell Biology and Anatomy, Graduate School of MedicineThe University of TokyoTokyoJapan
- Tsinghua‐Peking Center for Life Sciences, School of Life SciencesTsinghua UniversityBeijingChina
| | - Tadayuki Ogawa
- Department of Cell Biology and Anatomy, Graduate School of MedicineThe University of TokyoTokyoJapan
- Research Center for Advanced Medical ScienceDokkyo Medical UniversityTochigiJapan
- Biomolecular Characterization UnitRIKEN Center for Sustainable Resource ScienceWakoJapan
| | - Kento Yonezawa
- Structural Biology Research Center, Photon FactoryInstitute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)TsukubaJapan
- Center for Digital Green‐InnovationNara Institute of Science and TechnologyNaraJapan
| | - Nobutaka Shimizu
- Structural Biology Research Center, Photon FactoryInstitute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)TsukubaJapan
| | - Sotaro Ichinose
- Department of Cell Biology and Anatomy, Graduate School of MedicineThe University of TokyoTokyoJapan
- Department of Anatomy, Graduate School of MedicineGunma UniversityGunmaJapan
| | - Takayuki Uchihashi
- Department of PhysicsNagoya UniversityNagoyaJapan
- Exploratory Research Center on Life and Living Systems (ExCELLS)National Institutes of Natural SciencesOkazakiJapan
| | | | - Toshio Moriya
- Structural Biology Research Center, Photon FactoryInstitute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)TsukubaJapan
| | - Naruhiko Adachi
- Structural Biology Research Center, Photon FactoryInstitute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)TsukubaJapan
| | - Masato Kawasaki
- Structural Biology Research Center, Photon FactoryInstitute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)TsukubaJapan
| | - Naoshi Dohmae
- Biomolecular Characterization UnitRIKEN Center for Sustainable Resource ScienceWakoJapan
| | - Toshiya Senda
- Structural Biology Research Center, Photon FactoryInstitute of Materials Structure Science, High Energy Accelerator Research Organization (KEK)TsukubaJapan
| | - Nobutaka Hirokawa
- Department of Cell Biology and Anatomy, Graduate School of MedicineThe University of TokyoTokyoJapan
- Juntendo Advanced Research Institute for Health ScienceJuntendo UniversityTokyoJapan
| |
Collapse
|
27
|
Liu ZY, Li YH, Li BW, Xin L. Development and validation of a vesicle-mediated transport-associated gene signature for predicting prognosis and immune therapy response in hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:13211-13230. [PMID: 37479759 DOI: 10.1007/s00432-023-05079-1] [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: 05/28/2023] [Accepted: 06/29/2023] [Indexed: 07/23/2023]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a malignant tumor with a poor prognosis. The progression of numerous malignancies has been linked to abnormal vesicle-mediated transport-related gene (VMTRG) expression. The prognostic importance of VMTRGs in HCC is uncertain nonetheless. METHODS Utilizing HCC data from TCGA and ICGC, we employed univariate cox analysis, unsupervised clustering, and lasso analysis to construct molecular subtypes and prognostic signature of HCC based on the prognostic-associated VMTRGs expression levels. Subsequently, we validated the expression levels of the signature genes. We investigated the probable pathways using gene set variation analysis (GSVA) and gene set enrichment analysis (GSEA). Six methods were utilized to compare immune cell infiltration between two risk groups. Moreover, the "pRRophetic" algorithm was utilized to test the drug sensitivity of both groups. RESULTS We identified two distinct subtypes with divergent biological behaviors and immune functionality through unsupervised clustering. Subtype C1 demonstrated a poorer prognosis. A prognostic signature incorporating two VMTRGs (KIF2C and RAC1) was formulated. Immunohistochemistry and qRT-PCR analyses unveiled a significant upregulation of these pivotal genes within HCC tissues. The prognosis was worse for the high-risk group, which also had a higher clinicopathological grade, higher levels of tumor mutation burden (TMB), a higher immunological infiltration of CD8 + T cells, a higher expression of immune checkpoints, and enhanced immunotherapy efficacy. These two risk groups also have varied chemotherapy drug sensitivities. CONCLUSIONS Based on VMTRGs, we have developed a signature that assists in accurate prognosis prediction and formulating personalized treatment strategies for HCC patients.
Collapse
Affiliation(s)
- Zhi-Yang Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Yi-He Li
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, China
| | - Bo-Wen Li
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China
| | - Lin Xin
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Road, Donghu District, Nanchang, 330006, Jiangxi, China.
| |
Collapse
|
28
|
Chen YZ, Zimyanin V, Redemann S. Loss of the mitochondrial protein SPD-3 elevates PLK-1 levels and dysregulates mitotic events. Life Sci Alliance 2023; 6:e202302011. [PMID: 37684042 PMCID: PMC10488725 DOI: 10.26508/lsa.202302011] [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: 02/23/2023] [Revised: 08/28/2023] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
In metazoans, Polo-like kinase (PLK1) controls several mitotic events including nuclear envelope breakdown, centrosome maturation, spindle assembly and progression through mitosis. Here we show that a mutation in the mitochondria-localized protein SPD-3 affects mitotic events by inducing elevated levels of PLK-1 in early Caenorhabditis elegans embryos. SPD-3 mutant embryos contain abnormally positioned mitotic chromosomes, show a delay in anaphase onset and asymmetrically disassemble the nuclear lamina. We found that more PLK-1 accumulated on centrosomes, nuclear envelope, nucleoplasm, and chromatin before NEBD, suggesting that PLK-1 overexpression is responsible for some of the observed mitotic phenotypes. In agreement with this, the chromosome positioning defects of the spd-3(oj35) mutant could be rescued by reducing PLK-1 levels. Our data suggests that the mitochondrial SPD-3 protein affects chromosome positioning and nuclear envelope integrity by up-regulating the endogenous levels of PLK-1 during early embryogenesis in C. elegans This finding suggests a novel link between mitochondria and nuclear envelope dynamics and chromosome positioning by increasing the amount of a key mitotic regulator, PLK-1, providing a novel link between mitochondria and mitosis.
Collapse
Affiliation(s)
- Yu-Zen Chen
- https://ror.org/0153tk833 Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, USA
- https://ror.org/0153tk833 Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Vitaly Zimyanin
- https://ror.org/0153tk833 Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, USA
- https://ror.org/0153tk833 Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, USA
| | - Stefanie Redemann
- https://ror.org/0153tk833 Center for Membrane and Cell Physiology, School of Medicine, University of Virginia, Charlottesville, VA, USA
- https://ror.org/0153tk833 Department of Molecular Physiology and Biological Physics, School of Medicine, University of Virginia, Charlottesville, VA, USA
- https://ror.org/0153tk833 Department of Cell Biology, School of Medicine, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
29
|
Chan CC, Yen TH, Tseng HC, Mai B, Ho PK, Chou JL, Wu GJ, Huang YC. A Comprehensive Genetic Study of Microtubule-Associated Gene Clusters for Male Infertility in a Taiwanese Cohort. Int J Mol Sci 2023; 24:15363. [PMID: 37895049 PMCID: PMC10607339 DOI: 10.3390/ijms242015363] [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: 08/30/2023] [Revised: 10/14/2023] [Accepted: 10/15/2023] [Indexed: 10/29/2023] Open
Abstract
Advanced reproductive technologies are utilized to identify the genetic mutations that lead to spermatogenic impairment, and allow informed genetic counseling to patients to prevent the transmission of genetic defects to offspring. The purpose of this study was to identify potential single nucleotide polymorphisms (SNPs) associated with male infertility. Genetic variants that may cause infertility are identified by combining the targeted next-generation sequencing (NGS) panel and whole exome sequencing (WES). The validation step of Sanger sequencing adds confidence to the identified variants. Our analysis revealed five distinct affected genes covering seven SNPs based on the targeted NGS panel and WES data: SPATA16 (rs16846616, 1515442, 1515441), CFTR (rs213950), KIF6 (rs2273063), STPG2 (r2903150), and DRC7 (rs3809611). Infertile men have a higher mutation rate than fertile men, especially those with azoospermia. These findings strongly support the hypothesis that the dysfunction of microtubule-related and spermatogenesis-specific genes contributes to idiopathic male infertility. The SPATA16, CFTR, KIF6, STPG2, and DRC7 mutations are associated with male infertility, specifically azoospermia, and a further examination of this genetic function is required.
Collapse
Affiliation(s)
- Chying-Chyuan Chan
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (C.-C.C.)
- Department of Obstetrics and Gynecology, Taipei City Hospital-Renai Branch, Taipei 103212, Taiwan
| | - Te-Hsin Yen
- Department of Obstetrics and Gynecology, Taipei City Hospital-Renai Branch, Taipei 103212, Taiwan
| | - Hao-Chen Tseng
- School of Pharmacy, National Defense Medical Center, Taipei 114201, Taiwan
| | - Brang Mai
- School of Pharmacy, National Defense Medical Center, Taipei 114201, Taiwan
| | - Pin-Kuan Ho
- School of Dentistry, National Defense Medical Center, Taipei 114201, Taiwan
| | - Jian-Liang Chou
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (C.-C.C.)
- Department of Research and Development, National Defense Medical Center, Taipei 114201, Taiwan
| | - Gwo-Jang Wu
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (C.-C.C.)
- Reproductive Medical Center, Department of Obstetrics and Gynecology, Tri-Service General Hospital, Taipei 114202, Taiwan
| | - Yu-Chuan Huang
- Graduate Institute of Medical Sciences, National Defense Medical Center, Taipei 114201, Taiwan; (C.-C.C.)
- School of Pharmacy, National Defense Medical Center, Taipei 114201, Taiwan
- Department of Research and Development, National Defense Medical Center, Taipei 114201, Taiwan
| |
Collapse
|
30
|
Gao K, Gao Z, Xia M, Li H, Di J. Role of plectin and its interacting molecules in cancer. Med Oncol 2023; 40:280. [PMID: 37632650 DOI: 10.1007/s12032-023-02132-4] [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: 04/24/2023] [Accepted: 07/20/2023] [Indexed: 08/28/2023]
Abstract
Plectin, as the cytolinker and scaffolding protein, are widely expressed and abundant in many tissues, and has involved in various cellular activities contributing to tumorigenesis, such as cell adhesion, migration, and signal transduction. Due to the specific expression and differential localization of plectin in cancer, most researchers focus on the role of plectin in cancer, and it has emerged as a potent driver of malignant hallmarks in many human cancers, which provides the possibility for plectin to be widely used as a biomarker and therapeutic target in the early diagnosis and targeted drug delivery of the disease. However, there is still a lack of systematic review on the interaction molecules and mechanism of plectin. Herein, we summarized the structure, expression and function of plectin, and mainly focused on recent studies on the functional and physical interactions between plectin and its interacting molecules, shedding light on the potential of targeting plectin for cancer therapy.
Collapse
Affiliation(s)
- Keyu Gao
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Zhimin Gao
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Mingyi Xia
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China
| | - Hailong Li
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China.
| | - Jiehui Di
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 West Huaihai Road, Xuzhou, 221002, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, 221004, Jiangsu, China.
| |
Collapse
|
31
|
Zhang HF, Delaidelli A, Javed S, Turgu B, Morrison T, Hughes CS, Yang X, Pachva M, Lizardo MM, Singh G, Hoffmann J, Huang YZ, Patel K, Shraim R, Kung SH, Morin GB, Aparicio S, Martinez D, Maris JM, Bosse KR, Williams KC, Sorensen PH. A MYCN-independent mechanism mediating secretome reprogramming and metastasis in MYCN-amplified neuroblastoma. SCIENCE ADVANCES 2023; 9:eadg6693. [PMID: 37611092 PMCID: PMC10446492 DOI: 10.1126/sciadv.adg6693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 07/21/2023] [Indexed: 08/25/2023]
Abstract
MYCN amplification (MNA) is a defining feature of high-risk neuroblastoma (NB) and predicts poor prognosis. However, whether genes within or in close proximity to the MYCN amplicon also contribute to MNA+ NB remains poorly understood. Here, we identify that GREB1, a transcription factor encoding gene neighboring the MYCN locus, is frequently coexpressed with MYCN and promotes cell survival in MNA+ NB. GREB1 controls gene expression independently of MYCN, among which we uncover myosin 1B (MYO1B) as being highly expressed in MNA+ NB and, using a chick chorioallantoic membrane (CAM) model, as a crucial regulator of invasion and metastasis. Global secretome and proteome profiling further delineates MYO1B in regulating secretome reprogramming in MNA+ NB cells, and the cytokine MIF as an important pro-invasive and pro-metastatic mediator of MYO1B activity. Together, we have identified a putative GREB1-MYO1B-MIF axis as an unconventional mechanism promoting aggressive behavior in MNA+ NB and independently of MYCN.
Collapse
Affiliation(s)
- Hai-Feng Zhang
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Alberto Delaidelli
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Sumreen Javed
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Busra Turgu
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Taylor Morrison
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Christopher S. Hughes
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Xiaqiu Yang
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Manideep Pachva
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Michael M. Lizardo
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Gurdeep Singh
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Jennifer Hoffmann
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Yue Zhou Huang
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Khushbu Patel
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Rawan Shraim
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Gregg B. Morin
- Canada’s Michael Smith Genome Sciences Centre, Vancouver, BC V5Z4S6, Canada
- Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T1Z4, Canada
| | - Samuel Aparicio
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| | - Daniel Martinez
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John M. Maris
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kristopher R. Bosse
- Division of Oncology and Center for Childhood Cancer Research, Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pediatrics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Karla C. Williams
- Faculty of Pharmaceutical Sciences, The University of British Columbia, Vancouver, BC, Canada
| | - Poul H. Sorensen
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, BC V6T1Z4, Canada
- Department of Molecular Oncology, BC Cancer Agency, Vancouver, BC V5Z1L3, Canada
| |
Collapse
|
32
|
Monteiro P, Yeon B, Wallis SS, Godinho SA. Centrosome amplification fine tunes tubulin acetylation to differentially control intracellular organization. EMBO J 2023; 42:e112812. [PMID: 37403793 PMCID: PMC10425843 DOI: 10.15252/embj.2022112812] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2022] [Revised: 06/05/2023] [Accepted: 06/14/2023] [Indexed: 07/06/2023] Open
Abstract
Intracellular organelle organization is conserved in eukaryotic cells and is primarily achieved through active transport by motor proteins along the microtubule cytoskeleton. Microtubule post-translational modifications (PTMs) can contribute to microtubule diversity and differentially regulate motor-mediated transport. Here, we show that centrosome amplification, commonly observed in cancer and shown to promote aneuploidy and invasion, induces a global change in organelle positioning towards the cell periphery and facilitates nuclear migration through confined spaces. This reorganization requires kinesin-1 and is analogous to the loss of dynein. Cells with amplified centrosomes display increased levels of acetylated tubulin, a PTM that could enhance kinesin-1-mediated transport. Depletion of α-tubulin acetyltransferase 1 (αTAT1) to block tubulin acetylation rescues the displacement of centrosomes, mitochondria, and vimentin but not Golgi or endosomes. Analyses of the distribution of total and acetylated microtubules indicate that the polarized distribution of modified microtubules, rather than levels alone, plays an important role in the positioning of specific organelles, such as the centrosome. We propose that increased tubulin acetylation differentially impacts kinesin-1-mediated organelle displacement to regulate intracellular organization.
Collapse
Affiliation(s)
- Pedro Monteiro
- Centre for Cancer Cell and Molecular Biology, Barts Cancer InstituteQueen Mary University of LondonLondonUK
- Institut Curie, Paris Sciences and Lettres Research UniversityCentre National de la Recherche Scientifique, UMR144ParisFrance
| | - Bongwhan Yeon
- Centre for Cancer Cell and Molecular Biology, Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Samuel S Wallis
- Centre for Cancer Cell and Molecular Biology, Barts Cancer InstituteQueen Mary University of LondonLondonUK
| | - Susana A Godinho
- Centre for Cancer Cell and Molecular Biology, Barts Cancer InstituteQueen Mary University of LondonLondonUK
| |
Collapse
|
33
|
Yang C, Wei M, Zhao Y, Yang Z, Song M, Mi J, Yang X, Tian G. Regulation of insulin secretion by the post-translational modifications. Front Cell Dev Biol 2023; 11:1217189. [PMID: 37601108 PMCID: PMC10436566 DOI: 10.3389/fcell.2023.1217189] [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: 05/05/2023] [Accepted: 07/24/2023] [Indexed: 08/22/2023] Open
Abstract
Post-translational modification (PTM) has a significant impact on cellular signaling and function regulation. In pancreatic β cells, PTMs are involved in insulin secretion, cell development, and viability. The dysregulation of PTM in β cells is clinically associated with the development of diabetes mellitus. Here, we summarized current findings on major PTMs occurring in β cells and their roles in insulin secretion. Our work provides comprehensive insight into understanding the mechanisms of insulin secretion and potential therapeutic targets for diabetes from the perspective of protein PTMs.
Collapse
Affiliation(s)
- Chunhua Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Mengna Wei
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Yanpu Zhao
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Zhanyi Yang
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Mengyao Song
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Jia Mi
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| | - Xiaoyong Yang
- Yale Center for Molecular and Systems Metabolism, Department of Comparative Medicine, Department of Cellular and Molecular Physiology, Yale University School of Medicine, New Haven, CT, United States
| | - Geng Tian
- Shandong Technology Innovation Center of Molecular Targeting and Intelligent Diagnosis and Treatment, Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
34
|
Pinotsis DA, Fridman G, Miller EK. Cytoelectric Coupling: Electric fields sculpt neural activity and "tune" the brain's infrastructure. Prog Neurobiol 2023; 226:102465. [PMID: 37210066 DOI: 10.1016/j.pneurobio.2023.102465] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/09/2023] [Accepted: 05/17/2023] [Indexed: 05/22/2023]
Abstract
We propose and present converging evidence for the Cytoelectric Coupling Hypothesis: Electric fields generated by neurons are causal down to the level of the cytoskeleton. This could be achieved via electrodiffusion and mechanotransduction and exchanges between electrical, potential and chemical energy. Ephaptic coupling organizes neural activity, forming neural ensembles at the macroscale level. This information propagates to the neuron level, affecting spiking, and down to molecular level to stabilize the cytoskeleton, "tuning" it to process information more efficiently.
Collapse
Affiliation(s)
- Dimitris A Pinotsis
- Centre for Mathematical Neuroscience and Psychology and Department of Psychology, City -University of London, London EC1V 0HB, United Kingdom; The Picower Institute for Learning & Memory and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Gene Fridman
- Departments of Otolaryngology, Biomedical Engineering, and Electrical Engineering, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Earl K Miller
- The Picower Institute for Learning & Memory and Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| |
Collapse
|
35
|
Beckett D, Voth GA. Unveiling the Catalytic Mechanism of GTP Hydrolysis in Microtubules. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.01.538927. [PMID: 37205601 PMCID: PMC10187240 DOI: 10.1101/2023.05.01.538927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/21/2023]
Abstract
Microtubules (MTs) are large cytoskeletal polymers, composed of αβ-tubulin heterodimers, capable of stochastically converting from polymerizing to depolymerizing states and vice-versa. Depolymerization is coupled with hydrolysis of GTP within β-tubulin. Hydrolysis is favored in the MT lattice compared to free heterodimer with an experimentally observed rate increase of 500 to 700 fold, corresponding to an energetic barrier lowering of 3.8 to 4.0 kcal/mol. Mutagenesis studies have implicated α-tubulin residues, α:E254 and α:D251, as catalytic residues completing the β-tubulin active site of the lower heterodimer in the MT lattice. The mechanism for GTP hydrolysis in the free heterodimer, however, is not understood. Additionally, there has been debate concerning whether the GTP-state lattice is expanded or compacted relative to the GDP-state and whether a "compacted" GDP-state lattice is required for hydrolysis. In this work, extensive QM/MM simulations with transition-tempered metadynamics free energy sampling of compacted and expanded inter-dimer complexes, as well as free heterodimer, have been carried out to provide clear insight into the GTP hydrolysis mechanism. α:E254 was found to be the catalytic residue in a compacted lattice, while in the expanded lattice disruption of a key salt bridge interaction renders α:E254 less effective. The simulations reveal a barrier decrease of 3.8 ± 0.5 kcal/mol for the compacted lattice compared to free heterodimer, in good agreement with experimental kinetic measurements. Additionally, the expanded lattice barrier was found to be 6.3 ± 0.5 kcal/mol higher than compacted, demonstrating that GTP hydrolysis is variable with lattice state and slower at the MT tip. Significance Statement Microtubules (MTs) are large and dynamic components of the eukaryotic cytoskeleton with the ability to stochastically convert from a polymerizing to a depolymerizing state and vice-versa. Depolymerization is coupled to the hydrolysis of guanosine-5'-triphosphate (GTP), which is orders of magnitude faster in the MT lattice than in free tubulin heterodimers. Our results computationally ascertain the catalytic residue contacts in the MT lattice that accelerate GTP hydrolysis compared to the free heterodimer as well as confirm that a compacted MT lattice is necessary for hydrolysis while a more expanded lattice is unable to form the necessary contacts and thereby hydrolyze GTP.
Collapse
|
36
|
Yap YT, Li W, Huang Q, Zhou Q, Zhang D, Sheng Y, Mladenovic-Lucas L, Yee SP, Orwig KE, Granneman JG, Williams DC, Hess RA, Toure A, Zhang Z. DNALI1 interacts with the MEIG1/PACRG complex within the manchette and is required for proper sperm flagellum assembly in mice. eLife 2023; 12:e79620. [PMID: 37083624 PMCID: PMC10185345 DOI: 10.7554/elife.79620] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 03/12/2023] [Indexed: 04/22/2023] Open
Abstract
The manchette is a transient and unique structure present in elongating spermatids and required for proper differentiation of the germ cells during spermatogenesis. Previous work indicated that the MEIG1/PACRG complex locates in the manchette and is involved in the transport of cargos, such as SPAG16L, to build the sperm flagellum. Here, using co-immunoprecipitation and pull-down approaches in various cell systems, we established that DNALI1, an axonemal component originally cloned from Chlamydomonas reinhardtii, recruits and stabilizes PACRG and we confirm in vivo, the co-localization of DNALI1 and PACRG in the manchette by immunofluorescence of elongating murine spermatids. We next generated mice with a specific deficiency of DNALI1 in male germ cells, and observed a dramatic reduction of the sperm cells, which results in male infertility. In addition, we observed that the majority of the sperm cells exhibited abnormal morphology including misshapen heads, bent tails, enlarged midpiece, discontinuous accessory structure, emphasizing the importance of DNALI1 in sperm differentiation. Examination of testis histology confirmed impaired spermiogenesis in the mutant mice. Importantly, while testicular levels of MEIG1, PACRG, and SPAG16L proteins were unchanged in the Dnali1 mutant mice, their localization within the manchette was greatly affected, indicating that DNALI1 is required for the formation of the MEIG1/PACRG complex within the manchette. Interestingly, in contrast to MEIG1 and PACRG-deficient mice, the DNALI1-deficient mice also showed impaired sperm spermiation/individualization, suggesting additional functions beyond its involvement in the manchette structure. Overall, our work identifies DNALI1 as a protein required for sperm development.
Collapse
Affiliation(s)
- Yi Tian Yap
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
| | - Wei Li
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
| | - Qian Huang
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
- Department of Occupational and Environmental Medicine, School of Public Health, Wuhan University of Science and TechnologyWuhanChina
| | - Qi Zhou
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
- Department of Occupational and Environmental Medicine, School of Public Health, Wuhan University of Science and TechnologyWuhanChina
| | - David Zhang
- College of William and MaryWilliamsburgUnited States
| | - Yi Sheng
- Molecular Genetics and Developmental Biology Graduate Program, Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - Ljljiana Mladenovic-Lucas
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
| | - Siu-Pok Yee
- Department of Cell Biology, University of Connecticut Health CenterFarmingtonUnited States
| | - Kyle E Orwig
- Molecular Genetics and Developmental Biology Graduate Program, Department of Obstetrics, Gynecology and Reproductive Sciences, Magee-Womens Research Institute, University of Pittsburgh School of MedicinePittsburghUnited States
| | - James G Granneman
- Center for Molecular Medicine and Genetics, Wayne State University School of MedicineDetroitUnited States
| | - David C Williams
- Department of Pathology and Laboratory Medicine, University of North CarolinaChapel HillUnited States
| | - Rex A Hess
- Department of Comparative Biosciences, College of Veterinary Medicine, University of IllinoisUrbanaUnited States
| | - Aminata Toure
- University Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Team Physiology and Pathophysiology of Sperm cells, Institute for Advanced BiosciencesGrenobleFrance
| | - Zhibing Zhang
- Department of Physiology, Wayne State University School of MedicineDetroitUnited States
- Department of Obstetrics & Gynecology, Wayne State UniversityDetroitUnited States
| |
Collapse
|
37
|
Kalra A, Benny A, Travis SM, Zizzi EA, Morales-Sanchez A, Oblinsky DG, Craddock TJA, Hameroff SR, MacIver MB, Tuszyński JA, Petry S, Penrose R, Scholes GD. Electronic Energy Migration in Microtubules. ACS CENTRAL SCIENCE 2023; 9:352-361. [PMID: 36968538 PMCID: PMC10037452 DOI: 10.1021/acscentsci.2c01114] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Indexed: 05/29/2023]
Abstract
The repeating arrangement of tubulin dimers confers great mechanical strength to microtubules, which are used as scaffolds for intracellular macromolecular transport in cells and exploited in biohybrid devices. The crystalline order in a microtubule, with lattice constants short enough to allow energy transfer between amino acid chromophores, is similar to synthetic structures designed for light harvesting. After photoexcitation, can these amino acid chromophores transfer excitation energy along the microtubule like a natural or artificial light-harvesting system? Here, we use tryptophan autofluorescence lifetimes to probe energy hopping between aromatic residues in tubulin and microtubules. By studying how the quencher concentration alters tryptophan autofluorescence lifetimes, we demonstrate that electronic energy can diffuse over 6.6 nm in microtubules. We discover that while diffusion lengths are influenced by tubulin polymerization state (free tubulin versus tubulin in the microtubule lattice), they are not significantly altered by the average number of protofilaments (13 versus 14). We also demonstrate that the presence of the anesthetics etomidate and isoflurane reduce exciton diffusion. Energy transport as explained by conventional Förster theory (accommodating for interactions between tryptophan and tyrosine residues) does not sufficiently explain our observations. Our studies indicate that microtubules are, unexpectedly, effective light harvesters.
Collapse
Affiliation(s)
- Aarat
P. Kalra
- Department
of Chemistry, New Frick Chemistry Building, Princeton University, Princeton, New Jersey08544, United States
| | - Alfy Benny
- Department
of Chemistry, New Frick Chemistry Building, Princeton University, Princeton, New Jersey08544, United States
| | - Sophie M. Travis
- Department
of Molecular Biology, Schultz Laboratory, Princeton University, Princeton, New Jersey08544, United States
| | - Eric A. Zizzi
- Department
of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino, Torino10129, Italy
| | - Austin Morales-Sanchez
- Department
of Chemistry, New Frick Chemistry Building, Princeton University, Princeton, New Jersey08544, United States
| | - Daniel G. Oblinsky
- Department
of Chemistry, New Frick Chemistry Building, Princeton University, Princeton, New Jersey08544, United States
| | - Travis J. A. Craddock
- Departments
of Psychology & Neuroscience, Computer Science, and Clinical Immunology, Nova Southeastern University, Ft. Lauderdale, Florida33314, United States
| | - Stuart R. Hameroff
- Department
of Anesthesiology, Center for Consciousness Studies, University of Arizona, Tucson, Arizona85721, United States
| | - M. Bruce MacIver
- Department
of Anesthesiology, Stanford University School
of Medicine, Stanford, California94305, United States
| | - Jack A. Tuszyński
- Department
of Mechanical and Aerospace Engineering (DIMEAS), Politecnico di Torino, Torino10129, Italy
- Department
of Physics, University of Alberta, Edmonton, AlbertaT6G 2E1, Canada
- Department
of Oncology, University of Alberta, Edmonton, AlbertaT6G 1Z2, Canada
| | - Sabine Petry
- Department
of Molecular Biology, Schultz Laboratory, Princeton University, Princeton, New Jersey08544, United States
| | - Roger Penrose
- Mathematical
Institute, Andrew Wiles Building, University
of Oxford, Radcliffe Observatory Quarter, Woodstock Road, Oxford, OX2 6GG, United
Kingdom
| | - Gregory D. Scholes
- Department
of Chemistry, New Frick Chemistry Building, Princeton University, Princeton, New Jersey08544, United States
| |
Collapse
|
38
|
Ikeda R, Sakagami T, Hamada M, Sakamoto T, Hatabu T, Saito N, Ando M. De novo transcriptome analysis of the centrohelid Raphidocystis contractilis to identify genes involved in microtubule-based motility. J Eukaryot Microbiol 2023; 70:e12955. [PMID: 36409155 DOI: 10.1111/jeu.12955] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/17/2022] [Accepted: 11/14/2022] [Indexed: 11/23/2022]
Abstract
The centrohelid heliozoan Raphidocystis contractilis has many radiating axopodia, each containing axopodial microtubules. The axopodia show rapid contraction at nearly a video rate (30 frames per second) in response to mechanical stimuli. The axopodial contraction is accompanied by cytoskeletal microtubule depolymerization, but the molecular mechanism of this phenomenon has not been elucidated. In this study, we performed de novo transcriptome sequencing of R. contractilis to identify genes involved in microtubule dynamics such as the rapid axopodial contraction. The transcriptome sequencing generated 7.15-Gbp clean reads in total, which were assembled as 31,771 unigenes. Using the obtained gene sets, we identified several microtubule-severing proteins which might be involved in the rapid axopodial contraction, and kinesin-like genes that occur in gene duplication. On the other hand, some genes for microtubule motor proteins involved in the formation and motility of flagella were not found in R. contractilis, suggesting that the gene repertoire of R. contractilis reflected the morphological features of nonflagellated protists. Our transcriptome analysis provides basic information for the analysis of the molecular mechanism underlying microtubule dynamics in R. contractilis.
Collapse
Affiliation(s)
- Risa Ikeda
- Laboratory of Cell Physiology, Department of Science Education, Faculty of Education, Okayama University, Okayama, Japan.,Laboratory of Animal Physiology and Pharmacology, Department of Animal Science, Faculty of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Tosuke Sakagami
- Laboratory of Cell Physiology, Department of Science Education, Faculty of Education, Okayama University, Okayama, Japan.,Laboratory of Animal Physiology and Pharmacology, Department of Animal Science, Faculty of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Mayuko Hamada
- Ushimado Marine Institute, Faculty of Science and Technology, Okayama University, Okayama, Japan
| | - Tatsuya Sakamoto
- Ushimado Marine Institute, Faculty of Science and Technology, Okayama University, Okayama, Japan
| | - Toshimitsu Hatabu
- Laboratory of Animal Physiology and Pharmacology, Department of Animal Science, Faculty of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Noboru Saito
- Laboratory of Animal Physiology and Pharmacology, Department of Animal Science, Faculty of Environmental and Life Science, Okayama University, Okayama, Japan
| | - Motonori Ando
- Laboratory of Cell Physiology, Department of Science Education, Faculty of Education, Okayama University, Okayama, Japan.,Laboratory of Animal Physiology and Pharmacology, Department of Animal Science, Faculty of Environmental and Life Science, Okayama University, Okayama, Japan
| |
Collapse
|
39
|
Ma W, You S, Regnier M, McCammon JA. Integrating comparative modeling and accelerated simulations reveals conformational and energetic basis of actomyosin force generation. Proc Natl Acad Sci U S A 2023; 120:e2215836120. [PMID: 36802417 PMCID: PMC9992861 DOI: 10.1073/pnas.2215836120] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 01/15/2023] [Indexed: 02/23/2023] Open
Abstract
Muscle contraction is performed by arrays of contractile proteins in the sarcomere. Serious heart diseases, such as cardiomyopathy, can often be results of mutations in myosin and actin. Direct characterization of how small changes in the myosin-actin complex impact its force production remains challenging. Molecular dynamics (MD) simulations, although capable of studying protein structure-function relationships, are limited owing to the slow timescale of the myosin cycle as well as a lack of various intermediate structures for the actomyosin complex. Here, employing comparative modeling and enhanced sampling MD simulations, we show how the human cardiac myosin generates force during the mechanochemical cycle. Initial conformational ensembles for different myosin-actin states are learned from multiple structural templates with Rosetta. This enables us to efficiently sample the energy landscape of the system using Gaussian accelerated MD. Key myosin loop residues, whose substitutions are related to cardiomyopathy, are identified to form stable or metastable interactions with the actin surface. We find that the actin-binding cleft closure is allosterically coupled to the myosin motor core transitions and ATP-hydrolysis product release from the active site. Furthermore, a gate between switch I and switch II is suggested to control phosphate release at the prepowerstroke state. Our approach demonstrates the ability to link sequence and structural information to motor functions.
Collapse
Affiliation(s)
- Wen Ma
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA92093
| | - Shengjun You
- Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA98109
| | - J. Andrew McCammon
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA92093
- Department of Pharmacology, University of California, San Diego, La Jolla, CA92093
| |
Collapse
|
40
|
Chen YZ, Zimyanin V, Redemann S. Mitotic events depend on regulation of PLK-1 levels by the mitochondrial protein SPD-3. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.11.523633. [PMID: 36711457 PMCID: PMC9882028 DOI: 10.1101/2023.01.11.523633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
In metazoans, Polo Kinase (Plk1) controls several mitotic events including nuclear envelope breakdown, centrosome maturation and kinetochore assembly. Here we show that mitotic events regulated by Polo Like Kinase (PLK-1) in early C. elegans embryos depend on the mitochondrial-localized protein SPD-3. spd-3 mutant one-cell embryos contain abnormally positioned mitotic chromosomes and prematurely and asymmetrically disassemble the nuclear lamina. Nuclear envelope breakdown (NEBD) in C. elegans requires direct dephosphorylation of lamin by PLK-1. In spd-3 mutants PLK-1 levels are ~6X higher in comparison to control embryos and PLK-1::GFP was highly accumulated at centrosomes, the nuclear envelope, nucleoplasm, and chromosomes prior to NEBD. Partial depletion of plk-1 in spd-3 mutant embryos rescued mitotic chromosome and spindle positioning defects indicating that these phenotypes result from higher PLK-1 levels and thus activity. Our data suggests that the mitochondrial SPD-3 protein controls NEBD and chromosome positioning by regulating the endogenous levels of PLK-1 during early embryogenesis in C. elegans . This finding suggests a novel link between mitochondria and mitotic events by controlling the amount of a key mitotic regulator, PLK-1 and thus may have further implications in the context of cancers or age-related diseases and infertility as it provides a novel link between mitochondria and mitosis.
Collapse
Affiliation(s)
- Yu-Zen Chen
- Center for Membrane and Cell Physiology, University of Virginia, School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, USA
| | - Vitaly Zimyanin
- Center for Membrane and Cell Physiology, University of Virginia, School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, USA
| | - Stefanie Redemann
- Center for Membrane and Cell Physiology, University of Virginia, School of Medicine, Charlottesville, VA, USA
- Department of Molecular Physiology and Biological Physics, University of Virginia, School of Medicine, Charlottesville, VA, USA
- Department of Cell Biology, University of Virginia, School of Medicine, Charlottesville, VA, USA
| |
Collapse
|
41
|
Peng S, Wu Y, Zheng Y. High glucose causes developmental abnormalities in neuroepithelial cysts with actin and HK1 distribution changes. Front Cell Dev Biol 2023; 10:1021284. [PMID: 36684439 PMCID: PMC9852901 DOI: 10.3389/fcell.2022.1021284] [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: 08/17/2022] [Accepted: 12/20/2022] [Indexed: 01/09/2023] Open
Abstract
It has been reported that the offspring of diabetic pregnant women have an increased risk for neural tube defects. Previous studies in animal models suggested that high glucose induces cell apoptosis and epigenetic changes in the developing neural tube. However, effects on other cellular aspects such as the cell shape changes were not fully investigated. Actin dynamics plays essential roles in cell shape change. Disruption on actin dynamics is known to cause neural tube defects. In the present study, we used a 3D neuroepithelial cyst model and a rosette model, both cultured from human embryonic stem cells, to study the cellular effects caused by high glucose. By using these models, we observed couple of new changes besides increased apoptosis. First, we observed that high glucose disturbed the distribution of pH3 positive cells in the neuroepithelial cysts. Secondly, we found that high glucose exposure caused a relatively smaller actin inner boundary enclosed area, which was unlikely due to osmolarity changes. We further investigated key glucose metabolic enzymes in our models and the results showed that the distribution of hexokinase1 (HK1) was affected by high glucose. We observed that hexokinase1 has an apical-basal polarized distribution and is highest next to actin at the boundaries. hexokinase1 was more diffused and distributed less polarized under high glucose condition. Together, our observations broadened the cellular effects that may be caused by high glucose in the developing neural tube, especially in the secondary neurulation process.
Collapse
Affiliation(s)
- Sisi Peng
- Department of Cellular and Developmental Biology, School of Life Sciences, Fudan University, Shanghai, China,Obstetrics and Gynecology Hospital, The Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China,State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yu Wu
- Department of Cellular and Developmental Biology, School of Life Sciences, Fudan University, Shanghai, China,Obstetrics and Gynecology Hospital, The Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China,State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
| | - Yufang Zheng
- Department of Cellular and Developmental Biology, School of Life Sciences, Fudan University, Shanghai, China,Obstetrics and Gynecology Hospital, The Institute of Obstetrics and Gynecology, Fudan University, Shanghai, China,State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China,*Correspondence: Yufang Zheng,
| |
Collapse
|
42
|
Niu F, Liu Y, Sun K, Xu S, Dong J, Yu C, Yan K, Wei Z. Autoinhibition and activation mechanisms revealed by the triangular-shaped structure of myosin Va. SCIENCE ADVANCES 2022; 8:eadd4187. [PMID: 36490350 PMCID: PMC9733927 DOI: 10.1126/sciadv.add4187] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 11/03/2022] [Indexed: 06/17/2023]
Abstract
As the prototype of unconventional myosin motor family, myosin Va (MyoVa) transport cellular cargos along actin filaments in diverse cellular processes. The off-duty MyoVa adopts a closed and autoinhibited state, which can be relieved by cargo binding. The molecular mechanisms governing the autoinhibition and activation of MyoVa remain unclear. Here, we report the cryo-electron microscopy structure of the two full-length, closed MyoVa heavy chains in complex with 12 calmodulin light chains at 4.78-Å resolution. The MyoVa adopts a triangular structure with multiple intra- and interpolypeptide chain interactions in establishing the closed state with cargo binding and adenosine triphosphatase activity inhibited. Structural, biochemical, and cellular analyses uncover an asymmetric autoinhibition mechanism, in which the cargo-binding sites in the two MyoVa heavy chains are differently protected. Thus, specific and efficient MyoVa activation requires coincident binding of multiple cargo adaptors, revealing an intricate and elegant activity regulation of the motor in response to cargos.
Collapse
Affiliation(s)
- Fengfeng Niu
- Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Yong Liu
- Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- SUSTech-HIT Joint PhD Program, Harbin Institute of Technology, Harbin, Heilongjiang, China
| | - Kang Sun
- Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shun Xu
- Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Jiayuan Dong
- Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Cong Yu
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research and Shenzhen Key Laboratory of Cell Microenvironment, Shenzhen, Guangdong, China
| | - Kaige Yan
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Zhiyi Wei
- Brain Research Center, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
- Department of Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen, Guangdong, China
| |
Collapse
|
43
|
Basu A, Paul MK, Weiss S. The actin cytoskeleton: Morphological changes in pre- and fully developed lung cancer. BIOPHYSICS REVIEWS 2022; 3:041304. [PMID: 38505516 PMCID: PMC10903407 DOI: 10.1063/5.0096188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 12/09/2022] [Indexed: 03/21/2024]
Abstract
Actin, a primary component of the cell cytoskeleton can have multiple isoforms, each of which can have specific properties uniquely suited for their purpose. These monomers are then bound together to form polymeric filaments utilizing adenosine triphosphate hydrolysis as a source of energy. Proteins, such as Arp2/3, VASP, formin, profilin, and cofilin, serve important roles in the polymerization process. These filaments can further be linked to form stress fibers by proteins called actin-binding proteins, such as α-actinin, myosin, fascin, filamin, zyxin, and epsin. These stress fibers are responsible for mechanotransduction, maintaining cell shape, cell motility, and intracellular cargo transport. Cancer metastasis, specifically epithelial mesenchymal transition (EMT), which is one of the key steps of the process, is accompanied by the formation of thick stress fibers through the Rho-associated protein kinase, MAPK/ERK, and Wnt pathways. Recently, with the advent of "field cancerization," pre-malignant cells have also been demonstrated to possess stress fibers and related cytoskeletal features. Analytical methods ranging from western blot and RNA-sequencing to cryo-EM and fluorescent imaging have been employed to understand the structure and dynamics of actin and related proteins including polymerization/depolymerization. More recent methods involve quantifying properties of the actin cytoskeleton from fluorescent images and utilizing them to study biological processes, such as EMT. These image analysis approaches exploit the fact that filaments have a unique structure (curvilinear) compared to the noise or other artifacts to separate them. Line segments are extracted from these filament images that have assigned lengths and orientations. Coupling such methods with statistical analysis has resulted in development of a new reporter for EMT in lung cancer cells as well as their drug responses.
Collapse
Affiliation(s)
| | | | - Shimon Weiss
- Author to whom correspondence should be addressed:
| |
Collapse
|
44
|
Diaz-Valencia JD, Estrada-Abreo LA, Rodríguez-Cruz L, Salgado-Aguayo AR, Patiño-López G. Class I Myosins, molecular motors involved in cell migration and cancer. Cell Adh Migr 2022; 16:1-12. [PMID: 34974807 PMCID: PMC8741282 DOI: 10.1080/19336918.2021.2020705] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 12/02/2021] [Accepted: 12/16/2021] [Indexed: 01/13/2023] Open
Abstract
Class I Myosins are a subfamily of motor proteins with ATPase activity and a characteristic structure conserved in all myosins: A N-Terminal Motor Domain, a central Neck and a C terminal Tail domain. Humans have eight genes for these myosins. Class I Myosins have different functions: regulate membrane tension, participate in endocytosis, exocytosis, intracellular trafficking and cell migration. Cell migration is influenced by many cellular components including motor proteins, like myosins. Recently has been reported that changes in myosin expression have an impact on the migration of cancer cells, the formation of infiltrates and metastasis. We propose that class I myosins might be potential markers for future diagnostic, prognostic or even as therapeutic targets in leukemia and other cancers.Abbreviations: Myo1g: Myosin 1g; ALL: Acute Lymphoblastic Leukemia, TH1: Tail Homology 1; TH2: Tail Homology 2; TH3: Tail Homology 3.
Collapse
Affiliation(s)
- Juan D. Diaz-Valencia
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| | - Laura A. Estrada-Abreo
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Leonor Rodríguez-Cruz
- Cell Biology and Flow Cytometry Laboratory, Metropolitan Autonomous University, México City, Mexico
| | - Alfonso R. Salgado-Aguayo
- Rheumatic Diseases Laboratory, National Institute of Respiratory Diseases “Ismael Cosío Villegas”, Mexico City, Mexico
| | - Genaro Patiño-López
- Immunology and Proteomics Laboratory, Children’s Hospital of Mexico, Mexico City, Mexico
| |
Collapse
|
45
|
Hassan Ibrahim I, Balah A, Gomaa Abd Elfattah Hassan A, Gamal Abd El-Aziz H. Role of motor proteins in human cancers. Saudi J Biol Sci 2022; 29:103436. [PMID: 36131778 PMCID: PMC9483653 DOI: 10.1016/j.sjbs.2022.103436] [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: 03/29/2022] [Revised: 08/04/2022] [Accepted: 09/01/2022] [Indexed: 11/30/2022] Open
Abstract
Motor proteins include several protein families (Kinesin, Dynein and Myosin) responsible for intracellular transport, intercellular communication, among other functions. In cancer cells, motor proteins along with microtubules (MT) and other tubulin and actin structures, are crucial for cell proliferation and invasion. The cBioPortal platform for Cancer Genomics database was queried for solid cancers in a combined cohort of 9204 patients with complete cancer genomics data. To assess the importance of motor proteins in cancer, copy number alterations (CNAs) and survival rates were analyzed in the combined dataset. Kinesin, Dynein, and Myosin families showed CNAs in 47%, 49%, and 57 % of patients, respectively, in at least one of their members. Survival analysis showed that CNAs in Kinesin and Dynein, families' genes in the same patients were significantly correlated to decreased overall survival. These results added more evidence to previous literature highlighting the importance of motor proteins as a target in cancer therapy. Kinesin inhibitors could act by several mechanisms such as inhibiting spindle assembly or centrosome separation during mitosis, leading to cell cycle arrest and eventually apoptosis. Dynein inhibitors modulate Dynein's activity and MT binding, inhibiting cell proliferation and invasion. Myosin inhibitors act by stabilizing MT, inducing cell cycle arrest and inhibiting invasiveness. Increasing the specificity of motor proteins targeting drugs could improve cancer therapy and patient survival.
Collapse
Affiliation(s)
- Iman Hassan Ibrahim
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Postal code 11765, Egypt
| | - Amany Balah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy (Girls), Al- Azhar University, Postal code 11765, Egypt
| | - Abrar Gomaa Abd Elfattah Hassan
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Postal code 11765, Egypt
| | - Heba Gamal Abd El-Aziz
- Department of Biochemistry and Molecular Biology, Faculty of Pharmacy (Girls), Al-Azhar University, Postal code 11765, Egypt
| |
Collapse
|
46
|
Lyssavirus P Protein Isoforms Diverge Significantly in Subcellular Interactions Underlying Mechanisms of Interferon Antagonism. J Virol 2022; 96:e0139622. [PMID: 36222519 PMCID: PMC9599249 DOI: 10.1128/jvi.01396-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Viral hijacking of microtubule (MT)-dependent transport is well understood, but several viruses also express discrete MT-associated proteins (vMAPs), potentially to modulate MT-dependent processes in the host cell. Specific roles for vMAP-MT interactions include subversion of antiviral responses by P3, an isoform of the P protein of rabies virus (RABV; genus Lyssavirus), which mediates MT-dependent antagonism of interferon (IFN)-dependent signal transducers and activators of transcription 1 (STAT1) signaling. P3 also undergoes nucleocytoplasmic trafficking and inhibits STAT1-DNA binding, indicative of intranuclear roles in a multipronged antagonistic strategy. MT association/STAT1 antagonist functions of P3 correlate with pathogenesis, indicating potential as therapeutic targets. However, key questions remain, including whether other P protein isoforms interact with MTs, the relationship of these interactions with pathogenesis, and the extent of conservation of P3-MT interactions between diverse pathogenic lyssaviruses. Using super-resolution microscopy, live-cell imaging, and immune signaling analyses, we find that multiple P protein isoforms associate with MTs and that association correlates with pathogenesis. Furthermore, P3 proteins from different lyssaviruses exhibit variation in intracellular localization phenotypes that are associated with STAT1 antagonist function, whereby P3-MT association is conserved among lyssaviruses of phylogroup I but not phylogroup II, while nucleocytoplasmic localization varies between P3 proteins of the same phylogroup within both phylogroup I and II. Nevertheless, the divergent P3 proteins retain significant IFN antagonist function, indicative of adaptation to favor different inhibitory mechanisms, with MT interaction important to phylogroup I viruses. IMPORTANCE Lyssaviruses, including rabies virus, cause rabies, a progressive encephalomyelitis that is almost invariably fatal. There are no effective antivirals for symptomatic infection, and effective application of current vaccines is limited in areas of endemicity, such that rabies causes ~59,000 deaths per year. Viral subversion of host cell functions, including antiviral immunity, is critical to disease, and isoforms of the lyssavirus P protein are central to the virus-host interface underpinning immune evasion. Here, we show that specific cellular interactions of P protein isoforms involved in immune evasion vary significantly between different lyssaviruses, indicative of distinct strategies to evade immune responses. These findings highlight the diversity of the virus-host interface, an important consideration in the development of pan-lyssavirus therapeutic approaches.
Collapse
|
47
|
Mitra A, Peterman EJG. Intraflagellar transport: Derailing causes turnarounds. Curr Biol 2022; 32:R967-R969. [PMID: 36167049 DOI: 10.1016/j.cub.2022.07.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
In intraflagellar transport, protein complexes travel along cilia from base to tip without stopping and change direction only at the tip. A new study shows that this directional change does not depend on any tip-associated machinery.
Collapse
Affiliation(s)
- Aniruddha Mitra
- Department of Physics and Astronomy, and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Erwin J G Peterman
- Department of Physics and Astronomy, and LaserLaB Amsterdam, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
48
|
Lafanechère L. The microtubule cytoskeleton: An old validated target for novel therapeutic drugs. Front Pharmacol 2022; 13:969183. [PMID: 36188585 PMCID: PMC9521402 DOI: 10.3389/fphar.2022.969183] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/30/2022] [Indexed: 12/02/2022] Open
Abstract
Compounds targeting microtubules are widely used in cancer therapy with a proven efficacy. However, because they also target non-cancerous cells, their administration leads to numerous adverse effects. With the advancement of knowledge on the structure of tubulin, the regulation of microtubule dynamics and their deregulation in pathological processes, new therapeutic strategies are emerging, both for the treatment of cancer and for other diseases, such as neuronal or even heart diseases and parasite infections. In addition, a better understanding of the mechanism of action of well-known drugs such as colchicine or certain kinase inhibitors contributes to the development of these new therapeutic approaches. Nowadays, chemists and biologists are working jointly to select drugs which target the microtubule cytoskeleton and have improved properties. On the basis of a few examples this review attempts to depict the panorama of these recent advances.
Collapse
|
49
|
Shaw NM, Rios-Monterrosa JL, Fedorchak GR, Ketterer MR, Coombs GS, Lammerding J, Wallrath LL. Effects of mutant lamins on nucleo-cytoskeletal coupling in Drosophila models of LMNA muscular dystrophy. Front Cell Dev Biol 2022; 10:934586. [PMID: 36120560 PMCID: PMC9471154 DOI: 10.3389/fcell.2022.934586] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
The nuclei of multinucleated skeletal muscles experience substantial external force during development and muscle contraction. Protection from such forces is partly provided by lamins, intermediate filaments that form a scaffold lining the inner nuclear membrane. Lamins play a myriad of roles, including maintenance of nuclear shape and stability, mediation of nuclear mechanoresponses, and nucleo-cytoskeletal coupling. Herein, we investigate how disease-causing mutant lamins alter myonuclear properties in response to mechanical force. This was accomplished via a novel application of a micropipette harpooning assay applied to larval body wall muscles of Drosophila models of lamin-associated muscular dystrophy. The assay enables the measurement of both nuclear deformability and intracellular force transmission between the cytoskeleton and nuclear interior in intact muscle fibers. Our studies revealed that specific mutant lamins increase nuclear deformability while other mutant lamins cause nucleo-cytoskeletal coupling defects, which were associated with loss of microtubular nuclear caging. We found that microtubule caging of the nucleus depended on Msp300, a KASH domain protein that is a component of the linker of nucleoskeleton and cytoskeleton (LINC) complex. Taken together, these findings identified residues in lamins required for connecting the nucleus to the cytoskeleton and suggest that not all muscle disease-causing mutant lamins produce similar defects in subcellular mechanics.
Collapse
Affiliation(s)
- Nicholas M. Shaw
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Jose L. Rios-Monterrosa
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Gregory R. Fedorchak
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States
| | - Margaret R. Ketterer
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - Gary S. Coombs
- Biology Department, Waldorf University, Forest City, IA, United States
| | - Jan Lammerding
- The Nancy E. and Peter C. Meinig School of Biomedical Engineering, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, United States
| | - Lori L. Wallrath
- Department of Biochemistry, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
50
|
McMillan SN, Scarff CA. Cryo-electron microscopy analysis of myosin at work and at rest. Curr Opin Struct Biol 2022; 75:102391. [DOI: 10.1016/j.sbi.2022.102391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2022] [Revised: 04/18/2022] [Accepted: 04/22/2022] [Indexed: 01/01/2023]
|