1
|
Sultana T, Zheng C, Morton G, Megraw TL. Zika virus NS3 drives the assembly of a viroplasm-like structure. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613201. [PMID: 39345390 PMCID: PMC11429906 DOI: 10.1101/2024.09.16.613201] [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: 10/01/2024]
Abstract
Zika virus (ZIKV) is a mosquito-transmitted flavivirus that caused an epidemic in 2015-2016 in the Americas and raised serious global health concerns due to its association with congenital brain developmental defects in infected pregnancies. Upon infection, ZIKV assembles virus particles in a virus-generated toroidal compartment next to the nucleus called the replication factory, or viroplasm, which forms by remodeling the host cell endoplasmic reticulum (ER). How the viral proteins control viroplasm assembly remains unknown. Here we show that the ZIKV non-structural protein 3 (NS3) is sufficient to drive the assembly of a viroplasm-like structure (VLS) in human cells. NS3 encodes a dual-function protease and RNA helicase. The VLS is similar to the ZIKV viroplasm in its assembly near centrosomes at the nuclear periphery, its deformation of the nuclear membrane, its recruitment of ER, Golgi, and dsRNA, and its association with microtubules at its surface. While sufficient to generate a VLS, NS3 is less efficient in several aspects compared to viroplasm formation upon ZIKV infection. We further show that the helicase domain and not the protease domain is required for optimal VLS assembly and dsRNA recruitment. Overall, this work advances our understanding of the mechanism of viroplasm assembly by ZIKV and likely will extend to other flaviviruses.
Collapse
Affiliation(s)
- Tania Sultana
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Chunfeng Zheng
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Garret Morton
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, USA
| | - Timothy L. Megraw
- Department of Biomedical Sciences, College of Medicine, Florida State University, Tallahassee, Florida, USA
| |
Collapse
|
2
|
White S, Roller R. Herpes simplex virus type-1 cVAC formation in neuronal cells is mediated by dynein motor function and glycoprotein retrieval from the plasma membrane. J Virol 2024; 98:e0071324. [PMID: 38899931 PMCID: PMC11265375 DOI: 10.1128/jvi.00713-24] [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: 04/22/2024] [Accepted: 06/05/2024] [Indexed: 06/21/2024] Open
Abstract
Herpesvirus assembly requires the cytoplasmic association of large macromolecular and membrane structures that derive from both the nucleus and cytoplasmic membrane systems. Results from the study of human cytomegalovirus (HCMV) in cells where it organizes a perinuclear cytoplasmic virus assembly compartment (cVAC) show a clear requirement for the minus-end-directed microtubule motor, dynein, for virus assembly. In contrast, the assembly of herpes simplex virus -1 (HSV-1) in epithelial cells where it forms multiple dispersed, peripheral assembly sites is only mildly inhibited by the microtubule-depolymerizing agent, nocodazole. Here, we make use of a neuronal cell line system in which HSV-1 forms a single cVAC and show that dynein and its co-factor dynactin localize to the cVAC, and dynactin is associated with membranes that contain the virion tegument protein pUL11. We also show that the virus membrane-associated structural proteins pUL51 and the viral envelope glycoprotein gE arrive at the cVAC by different routes. Specifically, gE arrives at the cVAC after retrieval from the plasma membrane, suggesting the need for an intact retrograde transport system. Finally, we demonstrate that inhibition of dynactin function profoundly inhibits cVAC formation and virus production during the cytoplasmic assembly phase of infection.IMPORTANCEMany viruses reorganize cytoplasmic membrane systems and macromolecular transport systems to promote the production of progeny virions. Clarifying the mechanisms by which they accomplish this may reveal novel therapeutic strategies and illustrate mechanisms that are critical for normal cellular organization. Here, we explore the mechanism by which HSV-1 moves macromolecular and membrane cargo to generate a virus assembly compartment in the infected cell. We find that the virus makes use of a well-characterized, microtubule-based transport system that is stabilized against drugs that disrupt microtubules.
Collapse
Affiliation(s)
- Shaowen White
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Richard Roller
- Department of Microbiology and Immunology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
3
|
Polenghi M, Taverna E. Intracellular traffic and polarity in brain development. Front Neurosci 2023; 17:1172016. [PMID: 37859764 PMCID: PMC10583573 DOI: 10.3389/fnins.2023.1172016] [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: 02/22/2023] [Accepted: 07/31/2023] [Indexed: 10/21/2023] Open
Abstract
Neurons forming the human brain are generated during embryonic development by neural stem and progenitor cells via a process called neurogenesis. A crucial feature contributing to neural stem cell morphological and functional heterogeneity is cell polarity, defined as asymmetric distribution of cellular components. Cell polarity is built and maintained thanks to the interplay between polarity proteins and polarity-generating organelles, such as the endoplasmic reticulum (ER) and the Golgi apparatus (GA). ER and GA affect the distribution of membrane components and work as a hub where glycans are added to nascent proteins and lipids. In the last decades our knowledge on the role of polarity in neural stem and progenitor cells have increased tremendously. However, the role of traffic and associated glycosylation in neural stem and progenitor cells is still relatively underexplored. In this review, we discuss the link between cell polarity, architecture, identity and intracellular traffic, and highlight how studies on neurons have shaped our knowledge and conceptual framework on traffic and polarity. We will then conclude by discussing how a group of rare diseases, called congenital disorders of glycosylation (CDG) offers the unique opportunity to study the contribution of traffic and glycosylation in the context of neurodevelopment.
Collapse
|
4
|
Wildonger J, Than H. Intracellular transport: Finding the motor that will take you where you need to go. Curr Biol 2023; 33:R950-R953. [PMID: 37751706 DOI: 10.1016/j.cub.2023.08.014] [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: 09/28/2023]
Abstract
The Golgi complex is a busy production hub. A new study reveals that a microtubule end-binding (EB) protein enriched at the trans-Golgi network in neurons is needed to pair dense core vesicles with a kinesin motor for transport to axons.
Collapse
Affiliation(s)
- Jill Wildonger
- Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA; Cell and Developmental Biology, School of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA.
| | - Helen Than
- Pediatrics, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
5
|
Valdivia A, Duran C, Lee M, Williams HC, Lee MY, San Martin A. Nox1-based NADPH oxidase regulates the Par protein complex activity to control cell polarization. Front Cell Dev Biol 2023; 11:1231489. [PMID: 37635877 PMCID: PMC10457011 DOI: 10.3389/fcell.2023.1231489] [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: 05/30/2023] [Accepted: 07/25/2023] [Indexed: 08/29/2023] Open
Abstract
Cell migration is essential for many biological and pathological processes. Establishing cell polarity with a trailing edge and forming a single lamellipodium at the leading edge of the cell is crucial for efficient directional cell migration and is a hallmark of mesenchymal cell motility. Lamellipodia formation is regulated by spatial-temporal activation of the small GTPases Rac and Cdc42 at the front edge, and RhoA at the rear end. At a molecular level, partitioning-defective (Par) protein complex comprising Par3, Par6, and atypical Protein Kinase (aPKC isoforms ζ and λ/ι) regulates front-rear axis polarization. At the front edge, integrin clustering activates Cdc42, prompting the formation of Par3/Par6/aPKC complexes to modulate MTOC positioning and microtubule stabilization. Consequently, the Par3/Par6/aPKC complex recruits Rac1-GEF Tiam to activate Rac1, leading to lamellipodium formation. At the rear end, RhoA-ROCK phosphorylates Par3 disrupting its interaction with Tiam and inactivating Rac1. RhoA activity at the rear end allows the formation of focal adhesions and stress fibers necessary to generate the traction forces that allow cell movement. Nox1-based NADPH oxidase is necessary for PDGF-induced migration in vitro and in vivo for many cell types, including fibroblasts and smooth muscle cells. Here, we report that Nox1-deficient cells failed to acquire a normal front-to-rear polarity, polarize MTOC, and form a single lamellipodium. Instead, these cells form multiple protrusions that accumulate Par3 and active Tiam. The exogenous addition of H2O2 rescues this phenotype and is associated with the hyperactivation of Par3, Tiam, and Rac1. Mechanistically, Nox1 deficiency induces the inactivation of PP2A phosphatase, leading to increased activation of aPKC. These results were validated in Nox1y/- primary mouse aortic smooth muscle cells (MASMCs), which also showed PP2A inactivation after PDGF-BB stimulation consistent with exacerbated activation of aPKC. Moreover, we evaluated the physiological relevance of this signaling pathway using a femoral artery wire injury model to generate neointimal hyperplasia. Nox1y/- mice showed increased staining for the inactive form of PP2A and increased signal for active aPKC, suggesting that PP2A and aPKC activities might contribute to reducing neointima formation observed in the arteries of Nox1y/- mice.
Collapse
Affiliation(s)
- Alejandra Valdivia
- Division of Cardiology, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Charity Duran
- Division of Cardiology, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Mingyoung Lee
- Division of Cardiology, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Holly C. Williams
- Division of Cardiology, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
| | - Moo-Yeol Lee
- Division of Cardiology, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University, Goyang, Republic of Korea
| | - Alejandra San Martin
- Division of Cardiology, Department of Medicine, School of Medicine, Emory University, Atlanta, GA, United States
- Institute of Biomedical Sciences, Faculty of Medicine and Faculty of Life Science, Universidad Andres Bello, Santiago, Chile
| |
Collapse
|
6
|
Kim WK, Choi W, Deshar B, Kang S, Kim J. Golgi Stress Response: New Insights into the Pathogenesis and Therapeutic Targets of Human Diseases. Mol Cells 2023; 46:191-199. [PMID: 36574967 PMCID: PMC10086555 DOI: 10.14348/molcells.2023.2152] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 10/24/2022] [Accepted: 10/30/2022] [Indexed: 12/29/2022] Open
Abstract
The Golgi apparatus modifies and transports secretory and membrane proteins. In some instances, the production of secretory and membrane proteins exceeds the capacity of the Golgi apparatus, including vesicle trafficking and the post-translational modification of macromolecules. These proteins are not modified or delivered appropriately due to insufficiency in the Golgi function. These conditions disturb Golgi homeostasis and induce a cellular condition known as Golgi stress, causing cells to activate the 'Golgi stress response,' which is a homeostatic process to increase the capacity of the Golgi based on cellular requirements. Since the Golgi functions are diverse, several response pathways involving TFE3, HSP47, CREB3, proteoglycan, mucin, MAPK/ETS, and PERK regulate the capacity of each Golgi function separately. Understanding the Golgi stress response is crucial for revealing the mechanisms underlying Golgi dynamics and its effect on human health because many signaling molecules are related to diseases, ranging from viral infections to fatal neurodegenerative diseases. Therefore, it is valuable to summarize and investigate the mechanisms underlying Golgi stress response in disease pathogenesis, as they may contribute to developing novel therapeutic strategies. In this review, we investigate the perturbations and stress signaling of the Golgi, as well as the therapeutic potentials of new strategies for treating Golgi stress-associated diseases.
Collapse
Affiliation(s)
- Won Kyu Kim
- Natural Product Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Korea
- Division of Bio-Medical Science & Technology, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Wooseon Choi
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Barsha Deshar
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| | - Shinwon Kang
- Department of Physiology, University of Toronto, Toronto, ON M5S, Canada
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Sinai Health System, Toronto, ON M5G, Canada
| | - Jiyoon Kim
- Department of Pharmacology, Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul 06591, Korea
| |
Collapse
|
7
|
Vijayan K, Arang N, Wei L, Morrison R, Geiger R, Parks KR, Lewis AJ, Mast FD, Douglass AN, Kain HS, Aitchison JD, Johnson JS, Aderem A, Kaushansky A. A genome-wide CRISPR-Cas9 screen identifies CENPJ as a host regulator of altered microtubule organization during Plasmodium liver infection. Cell Chem Biol 2022; 29:1419-1433.e5. [PMID: 35738280 PMCID: PMC9481707 DOI: 10.1016/j.chembiol.2022.06.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Revised: 02/03/2022] [Accepted: 06/01/2022] [Indexed: 11/30/2022]
Abstract
Prior to initiating symptomatic malaria, a single Plasmodium sporozoite infects a hepatocyte and develops into thousands of merozoites, in part by scavenging host resources, likely delivered by vesicles. Here, we demonstrate that host microtubules (MTs) dynamically reorganize around the developing liver stage (LS) parasite to facilitate vesicular transport to the parasite. Using a genome-wide CRISPR-Cas9 screen, we identified host regulators of cytoskeleton organization, vesicle trafficking, and ER/Golgi stress that regulate LS development. Foci of γ-tubulin localized to the parasite periphery; depletion of centromere protein J (CENPJ), a novel regulator identified in the screen, exacerbated this re-localization and increased infection. We demonstrate that the Golgi acts as a non-centrosomal MT organizing center (ncMTOC) by positioning γ-tubulin and stimulating MT nucleation at parasite periphery. Together, these data support a model where the Plasmodium LS recruits host Golgi to form MT-mediated conduits along which host organelles are recruited to PVM and support parasite development.
Collapse
Affiliation(s)
- Kamalakannan Vijayan
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Nadia Arang
- Center for Infectious Disease Research, Seattle, WA, USA
| | - Ling Wei
- Seattle Children's Research Institute, Seattle, WA, USA
| | - Robert Morrison
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Rechel Geiger
- MSTP Program, University of Washington, Seattle, WA, USA
| | - K Rachael Parks
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Adam J Lewis
- Center for Infectious Disease Research, Seattle, WA, USA
| | - Fred D Mast
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA
| | - Alyse N Douglass
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Heather S Kain
- Center for Infectious Disease Research, Seattle, WA, USA
| | - John D Aitchison
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Biochemistry, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | | | - Alan Aderem
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA
| | - Alexis Kaushansky
- Center for Infectious Disease Research, Seattle, WA, USA; Seattle Children's Research Institute, Seattle, WA, USA; Department of Global Health, University of Washington, Seattle, WA, USA; Department of Pediatrics, University of Washington, Seattle, WA, USA; Brotman Baty Institute for Precision Medicine, Seattle, WA, USA.
| |
Collapse
|
8
|
Wang C, Qu K, Wang J, Qin R, Li B, Qiu J, Wang G. Biomechanical regulation of planar cell polarity in endothelial cells. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166495. [PMID: 35850177 DOI: 10.1016/j.bbadis.2022.166495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 07/09/2022] [Accepted: 07/11/2022] [Indexed: 01/03/2023]
Abstract
Cell polarity refers to the uneven distribution of certain cytoplasmic components in a cell with a spatial order. The planar cell polarity (PCP), the cell aligns perpendicular to the polar plane, in endothelial cells (ECs) has become a research hot spot. The planar polarity of ECs has a positive significance on the regulation of cardiovascular dysfunction, pathological angiogenesis, and ischemic stroke. The endothelial polarity is stimulated and regulated by biomechanical force. Mechanical stimuli promote endothelial polarization and make ECs produce PCP to maintain the normal physiological and biochemical functions. Here, we overview recent advances in understanding the interplay and mechanism between PCP and ECs function involved in mechanical forces, with a focus on PCP signaling pathways and organelles in regulating the polarity of ECs. And then showed the related diseases caused by ECs polarity dysfunction. This study provides new ideas and therapeutic targets for the treatment of endothelial PCP-related diseases.
Collapse
Affiliation(s)
- Caihong Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Jing Wang
- Institute of Food and Nutrition Development, Ministry of Agriculture and Rural Affairs, Beijing, China
| | - Rui Qin
- College of Life Sciences, South-Central University for Nationalities, Wuhan, China
| | - Bingyi Li
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China
| | - Juhui Qiu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing, China.
| |
Collapse
|
9
|
Bracey KM, Gu G, Kaverina I. Microtubules in Pancreatic β Cells: Convoluted Roadways Toward Precision. Front Cell Dev Biol 2022; 10:915206. [PMID: 35874834 PMCID: PMC9305484 DOI: 10.3389/fcell.2022.915206] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/14/2022] [Indexed: 11/18/2022] Open
Abstract
Pancreatic islet β cells regulate glucose homeostasis via glucose-stimulated insulin secretion (GSIS). Cytoskeletal polymers microtubules (MTs) serve as tracks for the transport and positioning of secretory insulin granules. MT network in β cells has unique morphology with several distinct features, which support granule biogenesis (via Golgi-derived MT array), net non-directional transport (via interlocked MT mesh), and control availability of granules at secretion sites (via submembrane MT bundle). The submembrane MT array, which is parallel to the plasma membrane and serves to withdraw excessive granules from the secretion hot spots, is destabilized and fragmented downstream of high glucose stimulation, allowing for regulated secretion. The origin of such an unusual MT network, the features that define its functionality, and metabolic pathways that regulate it are still to a large extent elusive and are a matter of active investigation and debate. Besides the MT network itself, it is important to consider the interplay of molecular motors that drive and fine-tune insulin granule transport. Importantly, activity of kinesin-1, which is the major MT-dependent motor in β cells, transports insulin granules, and has a capacity to remodel MT network, is also regulated by glucose. We discuss yet unknown potential avenues toward understanding how MT network and motor proteins provide control for secretion in coordination with other GSIS-regulating mechanisms.
Collapse
|
10
|
Chen F, Wu J, Iwanski MK, Jurriens D, Sandron A, Pasolli M, Puma G, Kromhout JZ, Yang C, Nijenhuis W, Kapitein LC, Berger F, Akhmanova A. Self-assembly of pericentriolar material in interphase cells lacking centrioles. eLife 2022; 11:77892. [PMID: 35787744 PMCID: PMC9307276 DOI: 10.7554/elife.77892] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 07/04/2022] [Indexed: 11/18/2022] Open
Abstract
The major microtubule-organizing center (MTOC) in animal cells, the centrosome, comprises a pair of centrioles surrounded by pericentriolar material (PCM), which nucleates and anchors microtubules. Centrosome assembly depends on PCM binding to centrioles, PCM self-association and dynein-mediated PCM transport, but the self-assembly properties of PCM components in interphase cells are poorly understood. Here, we used experiments and modeling to study centriole-independent features of interphase PCM assembly. We showed that when centrioles are lost due to PLK4 depletion or inhibition, dynein-based transport and self-clustering of PCM proteins are sufficient to form a single compact MTOC, which generates a dense radial microtubule array. Interphase self-assembly of PCM components depends on γ-tubulin, pericentrin, CDK5RAP2 and ninein, but not NEDD1, CEP152, or CEP192. Formation of a compact acentriolar MTOC is inhibited by AKAP450-dependent PCM recruitment to the Golgi or by randomly organized CAMSAP2-stabilized microtubules, which keep PCM mobile and prevent its coalescence. Linking of CAMSAP2 to a minus-end-directed motor leads to the formation of an MTOC, but MTOC compaction requires cooperation with pericentrin-containing self-clustering PCM. Our data reveal that interphase PCM contains a set of components that can self-assemble into a compact structure and organize microtubules, but PCM self-organization is sensitive to motor- and microtubule-based rearrangement.
Collapse
Affiliation(s)
- Fangrui Chen
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Jingchao Wu
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | | | - Daphne Jurriens
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Arianna Sandron
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Milena Pasolli
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Gianmarco Puma
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | | | - Chao Yang
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Wilco Nijenhuis
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | | | - Florian Berger
- Department of Biology, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Department of Biology, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
11
|
Mohammadi S, Rafii-Tabar H, Sasanpour P. A modeling study of the effect of an alternating magnetic field on magnetite nanoparticles in proximity of the neuronal microtubules: A proposed mechanism for detachment of tau proteins. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 222:106913. [PMID: 35738092 DOI: 10.1016/j.cmpb.2022.106913] [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: 01/26/2022] [Revised: 05/17/2022] [Accepted: 05/24/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND AND OBJECTIVE It is known that the disintegration of microtubules in neurons occurs in response to the phosphorylation of the tau proteins that promotes the structural instability of the microtubules, as one of the factors underlying the onset of Alzheimer's disease (AD). METHODS In this study, the mechanical variations undergone by the tau protein's and microtubule's structures due to the action of intrinsic magnetite nanoparticles inside the brain tissue have been computationally modeled using the finite element (FEM) method. RESULTS The von Mises stress induced by magnetite nanoparticles, subject to an applied alternating magnetic field, leads to local heating and mechanical forces, prompting a corresponding deformation in, and displacement of, the microtubule and the tau protein. CONCLUSIONS The induction of these deformations would increase the probability of the microtubules' depolymerization, and hence their eventual structural disintegration.
Collapse
Affiliation(s)
- Simah Mohammadi
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hashem Rafii-Tabar
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran; The Physics Branch of Iran Academy of Sciences, Tehran, Iran.
| | - Pezhman Sasanpour
- Department of Medical Physics & Biomedical Engineering, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
12
|
Schroeder EA, Chirgwin ME, Derbyshire ER. Plasmodium's fight for survival: escaping elimination while acquiring nutrients. Trends Parasitol 2022; 38:544-557. [PMID: 35534377 PMCID: PMC9187605 DOI: 10.1016/j.pt.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/10/2022] [Accepted: 04/10/2022] [Indexed: 01/08/2023]
Abstract
Plasmodium parasites extensively alter their host hepatocyte to evade host detection and support an unprecedented replication rate. Host cell manipulation includes association with the host early and late endomembrane systems, where Plasmodium accesses nutrients while suppressing cellular immune processes. Early endomembrane organelles provide an opportunity to sequester an abundance of lipids and proteins, but the association with late endomembrane organelles also risks autophagy-mediated elimination. While not all parasites survive, those that do benefit from a plethora of nutrients provided through this pathway. In this review, we discuss recent advances in our understanding of how Plasmodium parasites balance the need for host nutrients while avoiding elimination during the liver stage.
Collapse
Affiliation(s)
- Erin A Schroeder
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA
| | | | - Emily R Derbyshire
- Department of Molecular Genetics and Microbiology, Duke University, Durham, NC, USA; Department of Chemistry, Duke University, Durham, NC, USA.
| |
Collapse
|
13
|
Akhmanova A, Kapitein LC. Mechanisms of microtubule organization in differentiated animal cells. Nat Rev Mol Cell Biol 2022; 23:541-558. [PMID: 35383336 DOI: 10.1038/s41580-022-00473-y] [Citation(s) in RCA: 68] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/07/2022] [Indexed: 02/08/2023]
Abstract
Microtubules are polarized cytoskeletal filaments that serve as tracks for intracellular transport and form a scaffold that positions organelles and other cellular components and modulates cell shape and mechanics. In animal cells, the geometry, density and directionality of microtubule networks are major determinants of cellular architecture, polarity and proliferation. In dividing cells, microtubules form bipolar spindles that pull chromosomes apart, whereas in interphase cells, microtubules are organized in a cell type-specific fashion, which strongly correlates with cell physiology. In motile cells, such as fibroblasts and immune cells, microtubules are organized as radial asters, whereas in immotile epithelial and neuronal cells and in muscles, microtubules form parallel or antiparallel arrays and cortical meshworks. Here, we review recent work addressing how the formation of such microtubule networks is driven by the plethora of microtubule regulatory proteins. These include proteins that nucleate or anchor microtubule ends at different cellular structures and those that sever or move microtubules, as well as regulators of microtubule elongation, stability, bundling or modifications. The emerging picture, although still very incomplete, shows a remarkable diversity of cell-specific mechanisms that employ conserved building blocks to adjust microtubule organization in order to facilitate different cellular functions.
Collapse
Affiliation(s)
- Anna Akhmanova
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| | - Lukas C Kapitein
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
14
|
Liang XH, Nichols JG, Tejera D, Crooke ST. Perinuclear positioning of endosomes can affect PS-ASO activities. Nucleic Acids Res 2021; 49:12970-12985. [PMID: 34878127 PMCID: PMC8682747 DOI: 10.1093/nar/gkab1198] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 11/16/2021] [Accepted: 11/22/2021] [Indexed: 11/18/2022] Open
Abstract
Phosphorothioate (PS) modified antisense oligonucleotide (ASO) drugs that act on cellular RNAs must enter cells and be released from endocytic organelles to elicit antisense activity. It has been shown that PS-ASOs are mainly released by late endosomes. However, it is unclear how endosome movement in cells contributes to PS-ASO activity. Here, we show that PS-ASOs in early endosomes display Brownian type motion and migrate only short distances, whereas PS-ASOs in late endosomes (LEs) move linearly along microtubules with substantial distances. In cells with normal microtubules and LE movement, PS-ASO-loaded LEs tend to congregate perinuclearly. Disruption of perinuclear positioning of LEs by reduction of dynein 1 decreased PS-ASO activity, without affecting PS-ASO cellular uptake. Similarly, disruption of perinuclear positioning of PS-ASO-LE foci by reduction of ER tethering proteins RNF26, SQSTM1 and UBE2J1, or by overexpression of P50 all decreased PS-ASO activity. However, enhancing perinuclear positioning through reduction of USP15 or over-expression of RNF26 modestly increased PS-ASO activity, indicating that LE perinuclear positioning is required for ensuring efficient PS-ASO release. Together, these observations suggest that LE movement along microtubules and perinuclear positioning affect PS-ASO productive release.
Collapse
Affiliation(s)
- Xue-Hai Liang
- Core Antisense Research, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Joshua G Nichols
- Core Antisense Research, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Dario Tejera
- Neurology, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| | - Stanley T Crooke
- Core Antisense Research, Ionis Pharmaceuticals, Inc., Carlsbad, CA 92010, USA
| |
Collapse
|
15
|
Buchwalter RA, Ogden SC, York SB, Sun L, Zheng C, Hammack C, Cheng Y, Chen JV, Cone AS, Meckes DG, Tang H, Megraw TL. Coordination of Zika Virus Infection and Viroplasm Organization by Microtubules and Microtubule-Organizing Centers. Cells 2021; 10:3335. [PMID: 34943843 PMCID: PMC8699624 DOI: 10.3390/cells10123335] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 12/30/2022] Open
Abstract
Zika virus (ZIKV) became a global health concern in 2016 due to its links to congenital microcephaly and other birth defects. Flaviviruses, including ZIKV, reorganize the endoplasmic reticulum (ER) to form a viroplasm, a compartment where virus particles are assembled. Microtubules (MTs) and microtubule-organizing centers (MTOCs) coordinate structural and trafficking functions in the cell, and MTs also support replication of flaviviruses. Here we investigated the roles of MTs and the cell's MTOCs on ZIKV viroplasm organization and virus production. We show that a toroidal-shaped viroplasm forms upon ZIKV infection, and MTs are organized at the viroplasm core and surrounding the viroplasm. We show that MTs are necessary for viroplasm organization and impact infectious virus production. In addition, the centrosome and the Golgi MTOC are closely associated with the viroplasm, and the centrosome coordinates the organization of the ZIKV viroplasm toroidal structure. Surprisingly, viroplasm formation and virus production are not significantly impaired when infected cells have no centrosomes and impaired Golgi MTOC, and we show that MTs are anchored to the viroplasm surface in these cells. We propose that the viroplasm is a site of MT organization, and the MTs organized at the viroplasm are sufficient for efficient virus production.
Collapse
Affiliation(s)
- Rebecca A. Buchwalter
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA; (R.A.B.); (S.B.Y.); (L.S.); (C.Z.); (J.V.C.); (A.S.C.); (D.G.M.J.)
| | - Sarah C. Ogden
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA; (S.C.O.); (C.H.); (Y.C.); (H.T.)
| | - Sara B. York
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA; (R.A.B.); (S.B.Y.); (L.S.); (C.Z.); (J.V.C.); (A.S.C.); (D.G.M.J.)
| | - Li Sun
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA; (R.A.B.); (S.B.Y.); (L.S.); (C.Z.); (J.V.C.); (A.S.C.); (D.G.M.J.)
| | - Chunfeng Zheng
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA; (R.A.B.); (S.B.Y.); (L.S.); (C.Z.); (J.V.C.); (A.S.C.); (D.G.M.J.)
| | - Christy Hammack
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA; (S.C.O.); (C.H.); (Y.C.); (H.T.)
| | - Yichen Cheng
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA; (S.C.O.); (C.H.); (Y.C.); (H.T.)
| | - Jieyan V. Chen
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA; (R.A.B.); (S.B.Y.); (L.S.); (C.Z.); (J.V.C.); (A.S.C.); (D.G.M.J.)
| | - Allaura S. Cone
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA; (R.A.B.); (S.B.Y.); (L.S.); (C.Z.); (J.V.C.); (A.S.C.); (D.G.M.J.)
| | - David G. Meckes
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA; (R.A.B.); (S.B.Y.); (L.S.); (C.Z.); (J.V.C.); (A.S.C.); (D.G.M.J.)
| | - Hengli Tang
- Department of Biological Science, Florida State University, Tallahassee, FL 32306, USA; (S.C.O.); (C.H.); (Y.C.); (H.T.)
| | - Timothy L. Megraw
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA; (R.A.B.); (S.B.Y.); (L.S.); (C.Z.); (J.V.C.); (A.S.C.); (D.G.M.J.)
| |
Collapse
|
16
|
Revising Endosomal Trafficking under Insulin Receptor Activation. Int J Mol Sci 2021; 22:ijms22136978. [PMID: 34209489 PMCID: PMC8268289 DOI: 10.3390/ijms22136978] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/25/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
The endocytosis of ligand-bound receptors and their eventual recycling to the plasma membrane (PM) are processes that have an influence on signalling activity and therefore on many cell functions, including migration and proliferation. Like other tyrosine kinase receptors (TKR), the insulin receptor (INSR) has been shown to be endocytosed by clathrin-dependent and -independent mechanisms. Once at the early endosome (EE), the sorting of the receptor, either to the late endosome (LE) for degradation or back to the PM through slow or fast recycling pathways, will determine the intensity and duration of insulin effects. Both the endocytic and the endosomic pathways are regulated by many proteins, the Arf and Rab families of small GTPases being some of the most relevant. Here, we argue for a specific role for the slow recycling route, whilst we review the main molecular mechanisms involved in INSR endocytosis, sorting and recycling, as well as their possible role in cell functions.
Collapse
|
17
|
Zhao T, Hao H, Wang Z, Liang Y, Feng K, He M, Yun X, Bianco PR, Sun Y, Yao B, Lei M. Multi-color structured illumination microscopy for live cell imaging based on the enhanced image recombination transform algorithm. BIOMEDICAL OPTICS EXPRESS 2021; 12:3474-3484. [PMID: 34221673 PMCID: PMC8221967 DOI: 10.1364/boe.423171] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 04/09/2021] [Accepted: 05/10/2021] [Indexed: 05/02/2023]
Abstract
Structured illumination microscopy (SIM) has attracted considerable interest in super-resolution, live-cell imaging because of its low light dose and high imaging speed. Obtaining a high-quality reconstruction image in SIM depends on the precise determination of the parameters of the fringe illumination pattern. The image recombination transform (IRT) algorithm is superior to other algorithms in obtaining the precise initial phase without any approximation, which is promising to provide a considerable solution to address the difficulty of initial phase estimation at low-modulation-depth conditions. However, the IRT algorithm only considers a phase shift of π∕2, which limits its applications in general scenarios. In this letter, we present a general form of IRT algorithm suitable for arbitrary phase shifts, providing a powerful tool for parameter estimation in low signal-to-noise cases. To demonstrate the effectiveness of the enhanced IRT algorithm, we constructed a multicolor, structured illumination microscope and studied at super-resolution, the cargo traffic in HRPE cells, and monitored the movement of mitochondrial structures and microtubules in COS-7 cells. The custom SIM system using the enhanced IRT algorithm allows multicolor capability and a low excitation intensity fluorescence imaging less than 1 W/cm2. High-quality super-resolution images are obtained, which demonstrates the utility of this approach in imaging in the life sciences.
Collapse
Affiliation(s)
- Tianyu Zhao
- MOE Key Laboratory for Non-equilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi’an, Shaanxi 710119, China
- University of Chinese Academy of Sciences, Beijing 100049, China
- Authors contributed equally to this work
| | - Huiwen Hao
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing 100871, China
- Authors contributed equally to this work
| | - Zhaojun Wang
- MOE Key Laboratory for Non-equilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Yansheng Liang
- MOE Key Laboratory for Non-equilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Kun Feng
- MOE Key Laboratory for Non-equilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Minru He
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi’an, Shaanxi 710119, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xue Yun
- MOE Key Laboratory for Non-equilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
| | - Piero R. Bianco
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA
| | - Yujie Sun
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing 100871, China
| | - Baoli Yao
- State Key Laboratory of Transient Optics and Photonics, Xi’an Institute of Optics and Precision Mechanics, Chinese Academy of Sciences, Xi’an, Shaanxi 710119, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ming Lei
- MOE Key Laboratory for Non-equilibrium Synthesis and Modulation of Condensed Matter, School of Physics, Xi’an Jiaotong University, Xi’an 710049, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Ducret V, Richards AJ, Videlier M, Scalvenzi T, Moore KA, Paszkiewicz K, Bonneaud C, Pollet N, Herrel A. Transcriptomic analysis of the trade-off between endurance and burst-performance in the frog Xenopus allofraseri. BMC Genomics 2021; 22:204. [PMID: 33757428 PMCID: PMC7986297 DOI: 10.1186/s12864-021-07517-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 03/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Variation in locomotor capacity among animals often reflects adaptations to different environments. Despite evidence that physical performance is heritable, the molecular basis of locomotor performance and performance trade-offs remains poorly understood. In this study we identify the genes, signaling pathways, and regulatory processes possibly responsible for the trade-off between burst performance and endurance observed in Xenopus allofraseri, using a transcriptomic approach. RESULTS We obtained a total of about 121 million paired-end reads from Illumina RNA sequencing and analyzed 218,541 transcripts obtained from a de novo assembly. We identified 109 transcripts with a significant differential expression between endurant and burst performant individuals (FDR ≤ 0.05 and logFC ≥2), and blast searches resulted in 103 protein-coding genes. We found major differences between endurant and burst-performant individuals in the expression of genes involved in the polymerization and ATPase activity of actin filaments, cellular trafficking, proteoglycans and extracellular proteins secreted, lipid metabolism, mitochondrial activity and regulators of signaling cascades. Remarkably, we revealed transcript isoforms of key genes with functions in metabolism, apoptosis, nuclear export and as a transcriptional corepressor, expressed in either burst-performant or endurant individuals. Lastly, we find two up-regulated transcripts in burst-performant individuals that correspond to the expression of myosin-binding protein C fast-type (mybpc2). This suggests the presence of mybpc2 homoeologs and may have been favored by selection to permit fast and powerful locomotion. CONCLUSION These results suggest that the differential expression of genes belonging to the pathways of calcium signaling, endoplasmic reticulum stress responses and striated muscle contraction, in addition to the use of alternative splicing and effectors of cellular activity underlie locomotor performance trade-offs. Ultimately, our transcriptomic analysis offers new perspectives for future analyses of the role of single nucleotide variants, homoeology and alternative splicing in the evolution of locomotor performance trade-offs.
Collapse
Affiliation(s)
- Valérie Ducret
- UMR 7179 MECADEV, C.N.R.S/M.N.H.N., Département Adaptations du Vivant, 55 Rue Buffon, 75005, Paris, France.
| | - Adam J Richards
- Station d'Ecologie Expérimentale du CNRS, USR 2936, 09200, Moulis, France
| | - Mathieu Videlier
- Functional Ecology Lab, Department of Biology, University of Ottawa, 30 Marie Curie, Ottawa, ON, K1N 6N5, Canada
| | - Thibault Scalvenzi
- Evolution, Génomes, Comportement & Ecologie, Université Paris-Saclay, CNRS, IRD, 91198, Gif-sur-Yvette, France
| | - Karen A Moore
- Exeter Sequencing Service, College of Life and Environmental Sciences, University of Exeter, Exeter, EX4 4QD, UK
| | - Konrad Paszkiewicz
- Exeter Sequencing Service, College of Life and Environmental Sciences, University of Exeter, Exeter, EX4 4QD, UK
| | - Camille Bonneaud
- Station d'Ecologie Expérimentale du CNRS, USR 2936, 09200, Moulis, France
- Centre for Ecology & Conservation, College of Life and Environmental Sciences, University of Exeter, Penryn, Cornwall, UK
| | - Nicolas Pollet
- Evolution, Génomes, Comportement & Ecologie, Université Paris-Saclay, CNRS, IRD, 91198, Gif-sur-Yvette, France
| | - Anthony Herrel
- Station d'Ecologie Expérimentale du CNRS, USR 2936, 09200, Moulis, France
- Evolutionary Morphology of Vertebrates, Ghent University, B-9000, Ghent, Belgium
| |
Collapse
|
19
|
Rong Y, Yang W, Hao H, Wang W, Lin S, Shi P, Huang Y, Li B, Sun Y, Liu Z, Wu C. The Golgi microtubules regulate single cell durotaxis. EMBO Rep 2021; 22:e51094. [PMID: 33559938 PMCID: PMC7926246 DOI: 10.15252/embr.202051094] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 11/27/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022] Open
Abstract
Current understandings on cell motility and directionality rely heavily on accumulated investigations of the adhesion-actin cytoskeleton-actomyosin contractility cycles, while microtubules have been understudied in this context. Durotaxis, the ability of cells to migrate up gradients of substrate stiffness, plays a critical part in development and disease. Here, we identify the pivotal role of Golgi microtubules in durotactic migration of single cells. Using high-throughput analysis of microtubule plus ends/focal adhesion interactions, we uncover that these non-centrosomal microtubules actively impart leading edge focal adhesion (FA) dynamics. Furthermore, we designed a new system where islands of higher stiffness were patterned within RGD peptide coated polyacrylamide gels. We revealed that the positioning of the Golgi apparatus is responsive to external mechanical cues and that the Golgi-nucleus axis aligns with the stiffness gradient in durotaxis. Together, our work unveils the cytoskeletal underpinning for single cell durotaxis. We propose a model in which the Golgi-nucleus axis serves both as a compass and as a steering wheel for durotactic migration, dictating cell directionality through the interaction between non-centrosomal microtubules and the FA dynamics.
Collapse
Affiliation(s)
- Yingxue Rong
- Institute of Systems BiomedicineSchool of Basic Medical SciencePeking University Health Science CenterBeijingChina
| | - Wenzhong Yang
- Institute of Systems BiomedicineSchool of Basic Medical SciencePeking University Health Science CenterBeijingChina
| | - Huiwen Hao
- State Key Laboratory of Membrane BiologyBiomedical Pioneer Innovation Center (BIOPIC)School of Life SciencesPeking UniversityBeijingChina
| | - Wenxu Wang
- The Institute for Advanced StudiesWuhan UniversityWuhanChina
| | - Shaozhen Lin
- Applied Mechanics LaboratoryDepartment of Engineering MechanicsInstitute of Biomechanics and Medical EngineeringTsinghua UniversityBeijingChina
| | - Peng Shi
- Institute of Systems BiomedicineSchool of Basic Medical SciencePeking University Health Science CenterBeijingChina
| | - Yuxing Huang
- Institute of Systems BiomedicineSchool of Basic Medical SciencePeking University Health Science CenterBeijingChina
| | - Bo Li
- Applied Mechanics LaboratoryDepartment of Engineering MechanicsInstitute of Biomechanics and Medical EngineeringTsinghua UniversityBeijingChina
| | - Yujie Sun
- State Key Laboratory of Membrane BiologyBiomedical Pioneer Innovation Center (BIOPIC)School of Life SciencesPeking UniversityBeijingChina
| | - Zheng Liu
- The Institute for Advanced StudiesWuhan UniversityWuhanChina
| | - Congying Wu
- Institute of Systems BiomedicineSchool of Basic Medical SciencePeking University Health Science CenterBeijingChina
| |
Collapse
|
20
|
S Mogre S, Brown AI, Koslover EF. Getting around the cell: physical transport in the intracellular world. Phys Biol 2020; 17:061003. [PMID: 32663814 DOI: 10.1088/1478-3975/aba5e5] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Eukaryotic cells face the challenging task of transporting a variety of particles through the complex intracellular milieu in order to deliver, distribute, and mix the many components that support cell function. In this review, we explore the biological objectives and physical mechanisms of intracellular transport. Our focus is on cytoplasmic and intra-organelle transport at the whole-cell scale. We outline several key biological functions that depend on physically transporting components across the cell, including the delivery of secreted proteins, support of cell growth and repair, propagation of intracellular signals, establishment of organelle contacts, and spatial organization of metabolic gradients. We then review the three primary physical modes of transport in eukaryotic cells: diffusive motion, motor-driven transport, and advection by cytoplasmic flow. For each mechanism, we identify the main factors that determine speed and directionality. We also highlight the efficiency of each transport mode in fulfilling various key objectives of transport, such as particle mixing, directed delivery, and rapid target search. Taken together, the interplay of diffusion, molecular motors, and flows supports the intracellular transport needs that underlie a broad variety of biological phenomena.
Collapse
Affiliation(s)
- Saurabh S Mogre
- Department of Physics, University of California, San Diego, San Diego, California 92093, United States of America
| | | | | |
Collapse
|
21
|
Turn RE, East MP, Prekeris R, Kahn RA. The ARF GAP ELMOD2 acts with different GTPases to regulate centrosomal microtubule nucleation and cytokinesis. Mol Biol Cell 2020; 31:2070-2091. [PMID: 32614697 PMCID: PMC7543072 DOI: 10.1091/mbc.e20-01-0012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
ELMOD2 is a ∼32 kDa protein first purified by its GTPase-activating protein (GAP) activity toward ARL2 and later shown to have uniquely broad specificity toward ARF family GTPases in in vitro assays. To begin the task of defining its functions in cells, we deleted ELMOD2 in immortalized mouse embryonic fibroblasts and discovered a number of cellular defects, which are reversed upon expression of ELMOD2-myc. We show that these defects, resulting from the loss of ELMOD2, are linked to two different pathways and two different GTPases: with ARL2 and TBCD to support microtubule nucleation from centrosomes and with ARF6 in cytokinesis. These data highlight key aspects of signaling by ARF family GAPs that contribute to previously underappreciated sources of complexity, including GAPs acting from multiple sites in cells, working with multiple GTPases, and contributing to the spatial and temporal control of regulatory GTPases by serving as both GAPs and effectors.
Collapse
Affiliation(s)
- Rachel E Turn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322.,Biochemistry, Cell & Developmental Biology Graduate Program, Laney Graduate School, Emory University, Atlanta, GA 30307
| | - Michael P East
- Department of Pharmacology, University of North Carolina Chapel Hill, Chapel Hill, NC 27599
| | - Rytis Prekeris
- Department of Cell and Developmental Biology, University of Colorado, Aurora, CO 80045
| | - Richard A Kahn
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA 30322
| |
Collapse
|
22
|
Yang SZ, Wildonger J. Golgi Outposts Locally Regulate Microtubule Orientation in Neurons but Are Not Required for the Overall Polarity of the Dendritic Cytoskeleton. Genetics 2020; 215:435-447. [PMID: 32265236 PMCID: PMC7268992 DOI: 10.1534/genetics.119.302979] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/29/2020] [Indexed: 11/24/2022] Open
Abstract
Microtubule-organizing centers often play a central role in organizing the cellular microtubule networks that underlie cell function. In neurons, microtubules in axons and dendrites have distinct polarities. Dendrite-specific Golgi "outposts," in particular multicompartment outposts, have emerged as regulators of acentrosomal microtubule growth, raising the question of whether outposts contribute to establishing or maintaining the overall polarity of the dendritic microtubule cytoskeleton. Using a combination of genetic approaches and live imaging in a Drosophila model, we found that dendritic microtubule polarity is unaffected by eliminating known regulators of Golgi-dependent microtubule organization including the cis-Golgi matrix protein GM130, the fly AKAP450 ortholog pericentrin-like protein, and centrosomin. This indicates that Golgi outposts are not essential for the formation or maintenance of a dendrite-specific cytoskeleton. However, the overexpression of GM130, which promotes the formation of ectopic multicompartment units, is sufficient to alter dendritic microtubule polarity. Axonal microtubule polarity is similarly disrupted by the presence of ectopic multicompartment Golgi outposts. Notably, multicompartment outposts alter microtubule polarity independently of microtubule nucleation mediated by the γ-tubulin ring complex. Thus, although Golgi outposts are not essential to dendritic microtubule polarity, altering their organization correlates with changes to microtubule polarity. Based on these data, we propose that the organization of Golgi outposts is carefully regulated to ensure proper dendritic microtubule polarity.
Collapse
Affiliation(s)
- Sihui Z Yang
- Cellular and Molecular Biology Graduate Program, University of Wisconsin-Madison, Wisconsin 53706
- Department of Biochemistry, University of Wisconsin-Madison, Wisconsin 53706
| | - Jill Wildonger
- Department of Biochemistry, University of Wisconsin-Madison, Wisconsin 53706
| |
Collapse
|
23
|
The Golgi ribbon: mechanisms of maintenance and disassembly during the cell cycle. Biochem Soc Trans 2020; 48:245-256. [PMID: 32010930 DOI: 10.1042/bst20190646] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 01/01/2020] [Accepted: 01/06/2020] [Indexed: 12/18/2022]
Abstract
The Golgi complex (GC) has an essential role in the processing and sorting of proteins and lipids. The GC of mammalian cells is composed of stacks of cisternae connected by membranous tubules to create a continuous network, the Golgi ribbon, whose maintenance requires several core and accessory proteins. Despite this complex structural organization, the Golgi apparatus is highly dynamic, and this property becomes particularly evident during mitosis, when the ribbon undergoes a multistep disassembly process that allows its correct partitioning and inheritance by the daughter cells. Importantly, alterations of the Golgi structure are associated with a variety of physiological and pathological conditions. Here, we review the core mechanisms and signaling pathways involved in both the maintenance and disassembly of the Golgi ribbon, and we also report on the signaling pathways that connect the disassembly of the Golgi ribbon to mitotic entry and progression.
Collapse
|
24
|
Hao H, Niu J, Xue B, Su QP, Liu M, Yang J, Qin J, Zhao S, Wu C, Sun Y. Golgi-associated microtubules are fast cargo tracks and required for persistent cell migration. EMBO Rep 2020; 21:e48385. [PMID: 31984633 DOI: 10.15252/embr.201948385] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 12/11/2019] [Accepted: 12/19/2019] [Indexed: 11/09/2022] Open
Abstract
Microtubules derived from the Golgi (Golgi MTs) have been implicated to play critical roles in persistent cell migration, but the underlying mechanisms remain elusive, partially due to the lack of direct observation of Golgi MT-dependent vesicular trafficking. Here, using super-resolution stochastic optical reconstruction microscopy (STORM), we discovered that post-Golgi cargos are more enriched on Golgi MTs and also surprisingly move much faster than on non-Golgi MTs. We found that, compared to non-Golgi MTs, Golgi MTs are morphologically more polarized toward the cell leading edge with significantly fewer inter-MT intersections. In addition, Golgi MTs are more stable and contain fewer lattice repair sites than non-Golgi MTs. Our STORM/live-cell imaging demonstrates that cargos frequently pause at the sites of both MT intersections and MT defects. Furthermore, by optogenetic maneuvering of cell direction, we demonstrate that Golgi MTs are essential for persistent cell migration but not for cells to change direction. Together, our study unveils the role of Golgi MTs in serving as a group of "fast tracks" for anterograde trafficking of post-Golgi cargos.
Collapse
Affiliation(s)
- Huiwen Hao
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing, China
| | - Jiahao Niu
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing, China
| | - Boxin Xue
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing, China
| | - Qian Peter Su
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing, China
| | - Menghan Liu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Junsheng Yang
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing, China
| | - Jinshan Qin
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing, China
| | - Shujuan Zhao
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing, China
| | - Congying Wu
- Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Yujie Sun
- State Key Laboratory of Membrane Biology & Biomedical Pioneer Innovation Center (BIOPIC) & School of Life Sciences, Peking University, Beijing, China
| |
Collapse
|
25
|
Ravichandran Y, Goud B, Manneville JB. The Golgi apparatus and cell polarity: Roles of the cytoskeleton, the Golgi matrix, and Golgi membranes. Curr Opin Cell Biol 2019; 62:104-113. [PMID: 31751898 DOI: 10.1016/j.ceb.2019.10.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Revised: 10/02/2019] [Accepted: 10/14/2019] [Indexed: 12/15/2022]
Abstract
Membrane trafficking plays a crucial role in cell polarity by directing lipids and proteins to specific subcellular locations in the cell and sustaining a polarized state. The Golgi apparatus, the master organizer of membrane trafficking, can be subdivided into three layers that play different mechanical roles: a cytoskeletal layer, the so-called Golgi matrix, and the Golgi membranes. First, the outer regions of the Golgi apparatus interact with cytoskeletal elements, mainly actin and microtubules, which shape, position, and orient the organelle. Closer to the Golgi membranes, a matrix of long coiled-coiled proteins not only selectively captures transport intermediates but also participates in signaling events during polarization of membrane trafficking. Finally, the Golgi membranes themselves serve as active signaling platforms during cell polarity events. We review here the recent findings that link the Golgi apparatus to cell polarity, focusing on the roles of the cytoskeleton, the Golgi matrix, and the Golgi membranes.
Collapse
Affiliation(s)
- Yamini Ravichandran
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d'Ulm F-75005, Paris, France; Sorbonne Université, UPMC University Paris 06, CNRS, UMR 144, 26 rue d'Ulm F-75005, Paris, France; Institut Pasteur, CNRS, UMR 3691, 25 rue du Docteur Roux F-75014, Paris, France
| | - Bruno Goud
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d'Ulm F-75005, Paris, France; Sorbonne Université, UPMC University Paris 06, CNRS, UMR 144, 26 rue d'Ulm F-75005, Paris, France
| | - Jean-Baptiste Manneville
- Institut Curie, PSL Research University, CNRS, UMR 144, 26 rue d'Ulm F-75005, Paris, France; Sorbonne Université, UPMC University Paris 06, CNRS, UMR 144, 26 rue d'Ulm F-75005, Paris, France.
| |
Collapse
|
26
|
Generation and regulation of microtubule network asymmetry to drive cell polarity. Curr Opin Cell Biol 2019; 62:86-95. [PMID: 31739264 DOI: 10.1016/j.ceb.2019.10.004] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 01/19/2023]
Abstract
Microtubules control cell architecture by serving as a scaffold for intracellular transport, signaling, and organelle positioning. Microtubules are intrinsically polarized, and their orientation, density, and post-translational modifications both respond and contribute to cell polarity. Animal cells that can rapidly reorient their polarity axis, such as fibroblasts, immune cells, and cancer cells, contain radially organized microtubule arrays anchored at the centrosome and the Golgi apparatus, whereas stably polarized cells often acquire non-centrosomal microtubule networks attached to the cell cortex, nucleus, or other structures. Microtubule density, longevity, and post-translational modifications strongly depend on the dynamics of their plus ends. Factors controlling microtubule plus-end dynamics are often part of cortical assemblies that integrate cytoskeletal organization, cell adhesion, and secretion and are subject to microtubule-dependent feedback regulation. Finally, microtubules can mechanically contribute to cell asymmetry by promoting cell elongation, a property that might be important for cells with dense microtubule arrays growing in soft environments.
Collapse
|
27
|
Liu S, Majeed W, Grigaitis P, Betts MJ, Climer LK, Starkuviene V, Storrie B. Epistatic Analysis of the Contribution of Rabs and Kifs to CATCHR Family Dependent Golgi Organization. Front Cell Dev Biol 2019; 7:126. [PMID: 31428608 PMCID: PMC6687757 DOI: 10.3389/fcell.2019.00126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 06/26/2019] [Indexed: 01/05/2023] Open
Abstract
Multisubunit members of the CATCHR family: COG and NRZ complexes, mediate intra-Golgi and Golgi to ER vesicle tethering, respectively. We systematically addressed the genetic and functional interrelationships between Rabs, Kifs, and the retrograde CATCHR family proteins: COG3 and ZW10, which are necessary to maintain the organization of the Golgi complex. We scored the ability of siRNAs targeting 19 Golgi-associated Rab proteins and all 44 human Kifs, microtubule-dependent motor proteins, to suppress CATCHR-dependent Golgi fragmentation in an epistatic fluorescent microscopy-based assay. We found that co-depletion of Rab6A, Rab6A’, Rab27A, Rab39A and two minus-end Kifs, namely KIFC3 and KIF25, suppressed both COG3- and ZW10-depletion-induced Golgi fragmentation. ZW10-dependent Golgi fragmentation was suppressed selectively by a separate set of Rabs: Rab11A, Rab33B and the little characterized Rab29. 10 Kifs were identified as hits in ZW10-depletion-induced Golgi fragmentation, and, in contrast to the double suppressive Kifs, these were predominantly plus-end motors. No Rabs or Kifs selectively suppressed COG3-depletion-induced Golgi fragmentation. Protein-protein interaction network analysis indicated putative direct and indirect links between suppressive Rabs and tether function. Validation of the suppressive hits by EM confirmed a restored organization of the Golgi cisternal stack. Based on these outcomes, we propose a three-way competitive model of Golgi organization in which Rabs, Kifs and tethers modulate sequentially the balance between Golgi-derived vesicle formation, consumption, and off-Golgi transport.
Collapse
Affiliation(s)
- Shijie Liu
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Waqar Majeed
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Pranas Grigaitis
- Centre for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), Heidelberg University, Heidelberg, Germany
| | - Matthew J Betts
- Centre for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), Heidelberg University, Heidelberg, Germany
| | - Leslie K Climer
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| | - Vytaute Starkuviene
- Centre for Quantitative Analysis of Molecular and Cellular Biosystems (BioQuant), Heidelberg University, Heidelberg, Germany.,Institute of Pharmacology and Molecular Biotechnology (IPMB), Heidelberg University, Heidelberg, Germany.,Institute of Biosciences, Vilnius University Life Sciences Centre, Vilnius, Lithuania
| | - Brian Storrie
- Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, AR, United States
| |
Collapse
|
28
|
Yamase K, Tanigawa Y, Yamamoto Y, Tanaka H, Komiya T. Mouse TMCO5 is localized to the manchette microtubules involved in vesicle transfer in the elongating spermatids. PLoS One 2019; 14:e0220917. [PMID: 31393949 PMCID: PMC6687282 DOI: 10.1371/journal.pone.0220917] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Accepted: 07/25/2019] [Indexed: 12/31/2022] Open
Abstract
As a result of a high-throughput in situ hybridization screening for adult mouse testes, we found that the mRNA for Tmco5 is expressed in round and elongating spermatids. Tmco5 belongs to the Tmco (Transmembrane and coiled-coil domains) gene family and has a coiled-coil domain in the N-terminal and a transmembrane domain in the C-terminal region. A monoclonal antibody raised against recombinant TMCO5 revealed that the protein is expressed exclusively in the elongating spermatids of step 9 to 12 and is localized to the manchette, a transiently emerging construction, which predominantly consists of cytoskeleton microtubules and actin filaments. This structure serves in the transport of Golgi-derived non-acrosomal vesicles. Moreover, induced expression of TMCO5 in CHO cells resulted in the co-localization of TMCO5 with β-tubulin besides the reorganization of the Golgi apparatus. Judging from the results and considering the domain structure of TMCO5, we assume that Tmco5 may have a role in vesicle transport along the manchette.
Collapse
Affiliation(s)
- Kenya Yamase
- Department of Biological Function, Graduate School of Science, Osaka City University, Sugimoto, Sumiyoshi, Osaka, Japan
| | - Yoko Tanigawa
- Department of Biological Function, Graduate School of Science, Osaka City University, Sugimoto, Sumiyoshi, Osaka, Japan
| | - Yasufumi Yamamoto
- Department of Biological Function, Graduate School of Science, Osaka City University, Sugimoto, Sumiyoshi, Osaka, Japan
| | - Hiromitsu Tanaka
- Faculty of Pharmaceutical Sciences, Nagasaki International University, Sasebo, Nagasaki, Japan
| | - Tohru Komiya
- Department of Biological Function, Graduate School of Science, Osaka City University, Sugimoto, Sumiyoshi, Osaka, Japan
- * E-mail:
| |
Collapse
|
29
|
Mascanzoni F, Ayala I, Colanzi A. Organelle Inheritance Control of Mitotic Entry and Progression: Implications for Tissue Homeostasis and Disease. Front Cell Dev Biol 2019; 7:133. [PMID: 31396510 PMCID: PMC6664238 DOI: 10.3389/fcell.2019.00133] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 07/04/2019] [Indexed: 12/12/2022] Open
Abstract
The Golgi complex (GC), in addition to its well-known role in membrane traffic, is also actively involved in the regulation of mitotic entry and progression. In particular, during the G2 phase of the cell cycle, the Golgi ribbon is unlinked into isolated stacks. Importantly, this ribbon cleavage is required for G2/M transition, indicating that a "Golgi mitotic checkpoint" controls the correct segregation of this organelle. Then, during mitosis, the isolated Golgi stacks are disassembled, and this process is required for spindle formation. Moreover, recent evidence indicates that also proper mitotic segregation of other organelles, such as mitochondria, endosomes, and peroxisomes, is required for correct mitotic progression and/or spindle formation. Collectively, these observations imply that in addition to the control of chromosomes segregation, which is required to preserve the genetic information, the cells actively monitor the disassembly and redistribution of subcellular organelles in mitosis. Here, we provide an overview of the major structural reorganization of the GC and other organelles during G2/M transition and of their regulatory mechanisms, focusing on novel findings that have shed light on the basic processes that link organelle inheritance to mitotic progression and spindle formation, and discussing their implications for tissue homeostasis and diseases.
Collapse
Affiliation(s)
| | | | - Antonino Colanzi
- Institute of Biochemistry and Cell Biology, National Research Council, Naples, Italy
| |
Collapse
|
30
|
Nardi F, Franco OE, Fitchev P, Morales A, Vickman RE, Hayward SW, Crawford SE. DGAT1 Inhibitor Suppresses Prostate Tumor Growth and Migration by Regulating Intracellular Lipids and Non-Centrosomal MTOC Protein GM130. Sci Rep 2019; 9:3035. [PMID: 30816200 PMCID: PMC6395665 DOI: 10.1038/s41598-019-39537-z] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 12/31/2018] [Indexed: 11/10/2022] Open
Abstract
Acyl-CoA:diacylglycerol acyltransferase I (DGAT1) is a key enzyme in lipogenesis which is increased in metabolically active cells to meet nutrient requirements. DGAT1 has been recognized as an anti-obesity target; however, its role in the tumor microenvironment remains unclear. We postulated that, in prostate cancer (PCa) cells, augmented lipogenesis and growth are due to increased DGAT1 expression leading to microtubule-organizing center (MTOC) amplification. Thus, therapeutic targeting of DGAT1 potentially has tumor suppressive activity. We tested whether blocking DGAT1 in PCa cells altered MTOC and lipid signaling. Western blot and immunofluorescence were performed for MTOC and triglyceride mediators. Treatment with a DGAT1 inhibitor was evaluated. We found a stepwise increase in DGAT1 protein levels when comparing normal prostate epithelial cells to PCa cells, LNCaP and PC-3. Lipid droplets, MTOCs, and microtubule-regulating proteins were reduced in tumor cells treated with a DGAT1 inhibitor. Depletion of the non-centrosomal MTOC protein GM130 reduced PCa cell proliferation and migration. Inhibition of DGAT1 reduced tumor growth both in vitro and in vivo, and a negative feedback loop was discovered between DGAT1, PEDF, and GM130. These data identify DGAT1 as a promising new target for suppressing PCa growth by regulating GM130, MTOC number and disrupting microtubule integrity.
Collapse
Affiliation(s)
- Francesca Nardi
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Omar E Franco
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Philip Fitchev
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Alejandro Morales
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Renee E Vickman
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Simon W Hayward
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States
| | - Susan E Crawford
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL, 60201, United States.
| |
Collapse
|
31
|
Cheng HW, Hsiao CT, Chen YQ, Huang CM, Chan SI, Chiou A, Kuo JC. Centrosome guides spatial activation of Rac to control cell polarization and directed cell migration. Life Sci Alliance 2019; 2:2/1/e201800135. [PMID: 30737247 PMCID: PMC6369537 DOI: 10.26508/lsa.201800135] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 01/27/2019] [Accepted: 01/28/2019] [Indexed: 01/01/2023] Open
Abstract
The centrosome acts as a controller by balancing the formation of centrosomal and acentrosomal microtubules, the modulation of focal adhesion signaling and the activation of local Rac1 at the cell front, which then coordinates cell polarization during directed cell migration. Directed cell migration requires centrosome-mediated cell polarization and dynamical control of focal adhesions (FAs). To examine how FAs cooperate with centrosomes for directed cell migration, we used centrosome-deficient cells and found that loss of centrosomes enhanced the formation of acentrosomal microtubules, which failed to form polarized structures in wound-edge cells. In acentrosomal cells, we detected higher levels of Rac1-guanine nucleotide exchange factor TRIO (Triple Functional Domain Protein) on microtubules and FAs. Acentrosomal microtubules deliver TRIO to FAs for Rac1 regulation. Indeed, centrosome disruption induced excessive Rac1 activation around the cell periphery via TRIO, causing rapid FA turnover, a disorganized actin meshwork, randomly protruding lamellipodia, and loss of cell polarity. This study reveals the importance of centrosomes to balance the assembly of centrosomal and acentrosomal microtubules and to deliver microtubule-associated TRIO proteins to FAs at the cell front for proper spatial activation of Rac1, FA turnover, lamillipodial protrusion, and cell polarization, thereby allowing directed cell migration.
Collapse
Affiliation(s)
- Hung-Wei Cheng
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Cheng-Te Hsiao
- Institute of Biological Chemistry, Academia Sinica, Taipei, Taiwan
| | - Yin-Quan Chen
- Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan.,Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| | - Chi-Ming Huang
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Seng-I Chan
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan
| | - Arthur Chiou
- Institute of Biophotonics, National Yang-Ming University, Taipei, Taiwan
| | - Jean-Cheng Kuo
- Institute of Biochemistry and Molecular Biology, National Yang-Ming University, Taipei, Taiwan .,Cancer Progression Research Center, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
32
|
Abstract
Neurons are polarized cells with long branched axons and dendrites. Microtubule generation and organization machineries are crucial to grow and pattern these complex cellular extensions. Microtubule organizing centers (MTOCs) concentrate the molecular machinery for templating microtubules, stabilizing the nascent polymer, and organizing the resultant microtubules into higher-order structures. MTOC formation and function are well described at the centrosome, in the spindle, and at interphase Golgi; we review these studies and then describe recent results about how the machineries acting at these classic MTOCs are repurposed in the postmitotic neuron for axon and dendrite differentiation. We further discuss a constant tug-of-war interplay between different MTOC activities in the cell and how this process can be used as a substrate for transcription factor-mediated diversification of neuron types.
Collapse
Affiliation(s)
- Jason Y Tann
- Laboratory for Neurodiversity, RIKEN Centre for Brain Science, Saitama, Japan
| | - Adrian W Moore
- Laboratory for Neurodiversity, RIKEN Centre for Brain Science, Saitama, Japan.
| |
Collapse
|
33
|
Abstract
The Golgi apparatus is a central intracellular membrane-bound organelle with key functions in trafficking, processing, and sorting of newly synthesized membrane and secretory proteins and lipids. To best perform these functions, Golgi membranes form a unique stacked structure. The Golgi structure is dynamic but tightly regulated; it undergoes rapid disassembly and reassembly during the cell cycle of mammalian cells and is disrupted under certain stress and pathological conditions. In the past decade, significant amount of effort has been made to reveal the molecular mechanisms that regulate the Golgi membrane architecture and function. Here we review the major discoveries in the mechanisms of Golgi structure formation, regulation, and alteration in relation to its functions in physiological and pathological conditions to further our understanding of Golgi structure and function in health and diseases.
Collapse
Affiliation(s)
- Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Erpan Ahat
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
- Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI, USA.
| |
Collapse
|
34
|
Fuertes-Alvarez S, Maeso-Alonso L, Villoch-Fernandez J, Wildung M, Martin-Lopez M, Marshall C, Villena-Cortes AJ, Diez-Prieto I, Pietenpol JA, Tissir F, Lizé M, Marques MM, Marin MC. p73 regulates ependymal planar cell polarity by modulating actin and microtubule cytoskeleton. Cell Death Dis 2018; 9:1183. [PMID: 30518789 PMCID: PMC6281643 DOI: 10.1038/s41419-018-1205-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 11/02/2018] [Indexed: 02/07/2023]
Abstract
Planar cell polarity (PCP) and intercellular junctional complexes establish tissue structure and coordinated behaviors across epithelial sheets. In multiciliated ependymal cells, rotational and translational PCP coordinate cilia beating and direct cerebrospinal fluid circulation. Thus, PCP disruption results in ciliopathies and hydrocephalus. PCP establishment depends on the polarization of cytoskeleton and requires the asymmetric localization of core and global regulatory modules, including membrane proteins like Vangl1/2 or Frizzled. We analyzed the subcellular localization of select proteins that make up these modules in ependymal cells and the effect of Trp73 loss on their localization. We identify a novel function of the Trp73 tumor suppressor gene, the TAp73 isoform in particular, as an essential regulator of PCP through the modulation of actin and microtubule cytoskeleton dynamics, demonstrating that Trp73 is a key player in the organization of ependymal ciliated epithelia. Mechanistically, we show that p73 regulates translational PCP and actin dynamics through TAp73-dependent modulation of non-musclemyosin-II activity. In addition, TAp73 is required for the asymmetric localization of PCP-core and global signaling modules and regulates polarized microtubule dynamics, which in turn set up the rotational PCP. Therefore, TAp73 modulates, directly and/or indirectly, transcriptional programs regulating actin and microtubules dynamics and Golgi organization signaling pathways. These results shed light into the mechanism of ependymal cell planar polarization and reveal p73 as an epithelial architect during development regulating the cellular cytoskeleton.
Collapse
Affiliation(s)
- Sandra Fuertes-Alvarez
- Instituto de Biomedicina (IBIOMED) and Departamento de Biología Molecular, Universidad de León, Campus de Vegazana, 24071, León, Spain
| | - Laura Maeso-Alonso
- Instituto de Biomedicina (IBIOMED) and Departamento de Biología Molecular, Universidad de León, Campus de Vegazana, 24071, León, Spain
| | - Javier Villoch-Fernandez
- Instituto de Biomedicina (IBIOMED) and Departamento de Biología Molecular, Universidad de León, Campus de Vegazana, 24071, León, Spain
| | - Merit Wildung
- Molecular and Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center, 37077, Göttingen, Germany.,Institute of Molecular Oncology, Clinic for Cardiology and Pneumology, Department of Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Marta Martin-Lopez
- Instituto de Biomedicina (IBIOMED) and Departamento de Biología Molecular, Universidad de León, Campus de Vegazana, 24071, León, Spain
| | - Clayton Marshall
- Department of Biochemistry and Vanderbilt-Ingram Cancer Center, Vanderbilt University and Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Alberto J Villena-Cortes
- Instituto de Biomedicina (IBIOMED) and Departamento de Biología Molecular, Universidad de León, Campus de Vegazana, 24071, León, Spain
| | - Inmaculada Diez-Prieto
- Departamento de Medicina, Cirugía y Anatomía Veterinaria, Universidad de León, Campus de Vegazana, 24071, León, Spain
| | - Jennifer A Pietenpol
- Department of Biochemistry and Vanderbilt-Ingram Cancer Center, Vanderbilt University and Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Fadel Tissir
- Developmental Neurobiology, Institute of Neuroscience, Universite Catholique de Louvain, Avenue E. Mounier, 73, Box B1.73.16, B1200, Brussels, Belgium
| | - Muriel Lizé
- Molecular and Experimental Pneumology Group, Clinic for Cardiology and Pneumology, University Medical Center, 37077, Göttingen, Germany.,Institute of Molecular Oncology, Clinic for Cardiology and Pneumology, Department of Pneumology, University Medical Center Göttingen, Göttingen, Germany
| | - Margarita M Marques
- Instituto de Desarrollo Ganadero (INDEGSAL) and Departamento de Producción Animal, Universidad de León, Campus de Vegazana, 24071, León, Spain
| | - Maria C Marin
- Instituto de Biomedicina (IBIOMED) and Departamento de Biología Molecular, Universidad de León, Campus de Vegazana, 24071, León, Spain.
| |
Collapse
|
35
|
Chang B, Svoboda KKH, Liu X. Cell polarization: From epithelial cells to odontoblasts. Eur J Cell Biol 2018; 98:1-11. [PMID: 30473389 DOI: 10.1016/j.ejcb.2018.11.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Revised: 10/04/2018] [Accepted: 11/16/2018] [Indexed: 12/29/2022] Open
Abstract
Cell polarity identifies the asymmetry of a cell. Various types of cells, including odontoblasts and epithelial cells, polarize to fulfil their destined functions. Odontoblast polarization is a prerequisite and fundamental step for tooth development and tubular dentin formation. Current knowledge of odontoblast polarization, however, is very limited, which greatly impedes the development of novel approaches for regenerative endodontics. Compared to odontoblasts, epithelial cell polarization has been extensively studied over the last several decades. The knowledge obtained from epithelia polarization has been found applicable to other cell types, which is particularly useful considering the remarkable similarities of the morphological and compositional features between polarized odontoblasts and epithelia. In this review, we first discuss the characteristics, the key regulatory factors, and the process of epithelial polarity. Next, we compare the known facts of odontoblast polarization with epithelial cells. Lastly, we clarify knowledge gaps in odontoblast polarization and propose the directions for future research to fill the gaps, leading to the advancement of regenerative endodontics.
Collapse
Affiliation(s)
- Bei Chang
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Kathy K H Svoboda
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA
| | - Xiaohua Liu
- Department of Biomedical Sciences, Texas A&M University College of Dentistry, Dallas, TX, 75246, USA.
| |
Collapse
|
36
|
LaFlamme SE, Mathew-Steiner S, Singh N, Colello-Borges D, Nieves B. Integrin and microtubule crosstalk in the regulation of cellular processes. Cell Mol Life Sci 2018; 75:4177-4185. [PMID: 30206641 PMCID: PMC6182340 DOI: 10.1007/s00018-018-2913-x] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 08/14/2018] [Accepted: 08/27/2018] [Indexed: 11/25/2022]
Abstract
Integrins engage components of the extracellular matrix, and in collaboration with other receptors, regulate signaling cascades that impact cell behavior in part by modulating the cell's cytoskeleton. Integrins have long been known to function together with the actin cytoskeleton to promote cell adhesion, migration, and invasion, and with the intermediate filament cytoskeleton to mediate the strong adhesion needed for the maintenance and integrity of epithelial tissues. Recent studies have shed light on the crosstalk between integrin and the microtubule cytoskeleton. Integrins promote microtubule nucleation, growth, and stabilization at the cell cortex, whereas microtubules regulate integrin activity and remodeling of adhesion sites. Integrin-dependent stabilization of microtubules at the cell cortex is critical to the establishment of apical-basal polarity required for the formation of epithelial tissues. During cell migration, integrin-dependent microtubule stabilization contributes to front-rear polarity, whereas microtubules promote the turnover of integrin-mediated adhesions. This review focuses on this interdependent relationship and its impact on cell behavior and function.
Collapse
Affiliation(s)
- Susan E LaFlamme
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA.
| | - Shomita Mathew-Steiner
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
- Indiana University, 975 W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Neetu Singh
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Diane Colello-Borges
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| | - Bethsaida Nieves
- Department of Regenerative and Cancer Cell Biology, Albany Medical College, 47 New Scotland Avenue, Albany, NY, 12208, USA
| |
Collapse
|
37
|
The Golgi architecture and cell sensing. Biochem Soc Trans 2018; 46:1063-1072. [DOI: 10.1042/bst20180323] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/06/2018] [Accepted: 08/21/2018] [Indexed: 12/23/2022]
Abstract
An array of signalling molecules are located at the Golgi apparatus, including phosphoinositides, small GTPases, kinases, and phosphatases, which are linked to multiple signalling pathways. Initially considered to be associated predominantly with membrane trafficking, signalling pathways at the Golgi are now recognised to regulate a diverse range of higher-order functions. Many of these signalling pathways are influenced by the architecture of the Golgi. In vertebrate cells, the Golgi consists of individual stacks fused together into a compact ribbon structure and the function of this ribbon structure has been enigmatic. Notably, recent advances have identified a role for the Golgi ribbon in regulation of cellular processes. Fragmentation of the Golgi ribbon results in modulation of many signalling pathways. Various diseases and disorders, including cancer and neurodegeneration, are associated with the loss of the Golgi ribbon and the appearance of a dispersed fragmented Golgi. Here, we review the emerging theme of the Golgi as a cell sensor and highlight the relationship between the morphological status of the Golgi in vertebrate cells and the modulation of signalling networks.
Collapse
|
38
|
Singh V, Erady C, Balasubramanian N. Cell-matrix adhesion controls Golgi organization and function through Arf1 activation in anchorage-dependent cells. J Cell Sci 2018; 131:jcs.215855. [PMID: 30054383 PMCID: PMC6127727 DOI: 10.1242/jcs.215855] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Accepted: 06/27/2018] [Indexed: 12/15/2022] Open
Abstract
Cell-matrix adhesion regulates membrane trafficking controlling anchorage-dependent signaling. While a dynamic Golgi complex can contribute to this pathway, its regulation by adhesion remains unclear. Here we report that loss of adhesion dramatically disorganized the Golgi in mouse and human fibroblast cells. Golgi integrity is restored rapidly upon integrin-mediated re-adhesion to FN and is disrupted by integrin blocking antibody. In suspended cells, the cis, cis-medial and trans-Golgi networks differentially disorganize along the microtubule network but show no overlap with the ER, making this disorganization distinct from known Golgi fragmentation. This pathway is regulated by an adhesion-dependent reduction and recovery of Arf1 activation. Constitutively active Arf1 disrupts this regulation and prevents Golgi disorganization due to loss of adhesion. Adhesion-dependent Arf1 activation regulates its binding to the microtubule minus-end motor protein dynein to control Golgi reorganization, which is blocked by ciliobrevin. Adhesion-dependent Golgi organization controls its function, regulating cell surface glycosylation due to loss of adhesion, which is blocked by constitutively active Arf1. This study, hence, identified integrin-dependent cell-matrix adhesion to be a novel regulator of Arf1 activation, controlling Golgi organization and function in anchorage-dependent cells.
This article has an associated First Person interview with the first author of the paper. Summary: Integrin-dependent cell-matrix adhesion activates Arf1, which then recruits dynein to regulate Golgi organization and function along the microtubule network.
Collapse
Affiliation(s)
- Vibha Singh
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
| | - Chaitanya Erady
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
| | - Nagaraj Balasubramanian
- Indian Institute of Science Education and Research, Dr. Homi Bhabha Road, Pune, Maharashtra 411008, India
| |
Collapse
|
39
|
Functions and dysfunctions of the mammalian centrosome in health, disorders, disease, and aging. Histochem Cell Biol 2018; 150:303-325. [PMID: 30062583 DOI: 10.1007/s00418-018-1698-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/19/2018] [Indexed: 01/17/2023]
Abstract
Since its discovery well over 100 years ago (Flemming, in Sitzungsber Akad Wissensch Wien 71:81-147, 1875; Van Beneden, in Bull Acad R Belg 42:35-97, 1876) the centrosome is increasingly being recognized as a most impactful organelle for its role not only as primary microtubule organizing center (MTOC) but also as a major communication center for signal transduction pathways and as a center for proteolytic activities. Its significance for cell cycle regulation has been well studied and we now also know that centrosome dysfunctions are implicated in numerous diseases and disorders including cancer, Alstrom syndrome, Bardet-Biedl syndrome, Huntington's disease, reproductive disorders, and several other diseases and disorders. The present review is meant to build on information presented in the previous review (Schatten, in Histochem Cell Biol 129:667-686, 2008) and to highlight functions of the mammalian centrosome in health, and dysfunctions in disorders, disease, and aging with six sections focused on (1) centrosome structure and functions, and new insights into the role of centrosomes in cell cycle progression; (2) the role of centrosomes in tumor initiation and progression; (3) primary cilia, centrosome-primary cilia interactions, and consequences for cell cycle functions in health and disease; (4) transitions from centrosome to non-centrosome functions during cellular polarization; (5) other centrosome dysfunctions associated with the pathogenesis of human disease; and (6) centrosome functions in oocyte germ cells and dysfunctions in reproductive disorders and reproductive aging.
Collapse
|
40
|
Nardi F, Fitchev P, Franco OE, Ivanisevic J, Scheibler A, Hayward SW, Brendler CB, Welte MA, Crawford SE. PEDF regulates plasticity of a novel lipid-MTOC axis in prostate cancer-associated fibroblasts. J Cell Sci 2018; 131:jcs.213579. [PMID: 29792311 DOI: 10.1242/jcs.213579] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Accepted: 05/10/2018] [Indexed: 12/27/2022] Open
Abstract
Prostate tumors make metabolic adaptations to ensure adequate energy and amplify cell cycle regulators, such as centrosomes, to sustain their proliferative capacity. It is not known whether cancer-associated fibroblasts (CAFs) undergo metabolic re-programming. We postulated that CAFs augment lipid storage and amplify centrosomal or non-centrosomal microtubule-organizing centers (MTOCs) through a pigment epithelium-derived factor (PEDF)-dependent lipid-MTOC signaling axis. Primary human normal prostate fibroblasts (NFs) and CAFs were evaluated for lipid content, triacylglycerol-regulating proteins, MTOC number and distribution. CAFs were found to store more neutral lipids than NFs. Adipose triglyceride lipase (ATGL) and PEDF were strongly expressed in NFs, whereas CAFs had minimal to undetectable levels of PEDF or ATGL protein. At baseline, CAFs demonstrated MTOC amplification when compared to 1-2 perinuclear MTOCs consistently observed in NFs. Treatment with PEDF or blockade of lipogenesis suppressed lipid content and MTOC number. In summary, our data support that CAFs have acquired a tumor-like phenotype by re-programming lipid metabolism and amplifying MTOCs. Normalization of MTOCs by restoring PEDF or by blocking lipogenesis highlights a previously unrecognized plasticity in centrosomes, which is regulated through a new lipid-MTOC axis.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Francesca Nardi
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Philip Fitchev
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Omar E Franco
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Jelena Ivanisevic
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Adrian Scheibler
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Simon W Hayward
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Charles B Brendler
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| | - Michael A Welte
- Department of Biology, University of Rochester, Rochester, NY 14627, United States
| | - Susan E Crawford
- Department of Surgery, NorthShore University Research Institute, Affiliate of University of Chicago Pritzker School of Medicine, Evanston, IL 60201, United States
| |
Collapse
|
41
|
Wei YL, Yang WX. The acroframosome-acroplaxome-manchette axis may function in sperm head shaping and male fertility. Gene 2018; 660:28-40. [DOI: 10.1016/j.gene.2018.03.059] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 03/09/2018] [Accepted: 03/19/2018] [Indexed: 12/27/2022]
|
42
|
Abstract
A portfolio is presented documenting economic, high-resolution correlative focused ion beam scanning electron microscopy (FIB/SEM) in routine, comprising: (i) the use of custom-labeled slides and coverslips, (ii) embedding of cells in thin, or ultra-thin resin layers for correlative light and electron microscopy (CLEM) and (iii) the claim to reach the highest resolution possible with FIB/SEM in xyz. Regions of interest (ROIs) defined in light microscope (LM), can be relocated quickly and precisely in SEM. As proof of principle, HeLa cells were investigated in 3D context at all stages of the cell cycle, documenting ultrastructural changes during mitosis: nuclear envelope breakdown and reassembly, Golgi degradation and reconstitution and the formation of the midzone and midbody.
Collapse
|
43
|
Coming into Focus: Mechanisms of Microtubule Minus-End Organization. Trends Cell Biol 2018; 28:574-588. [PMID: 29571882 DOI: 10.1016/j.tcb.2018.02.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2017] [Revised: 02/17/2018] [Accepted: 02/27/2018] [Indexed: 11/22/2022]
Abstract
Microtubule organization has a crucial role in regulating cell architecture. The geometry of microtubule arrays strongly depends on the distribution of sites responsible for microtubule nucleation and minus-end attachment. In cycling animal cells, the centrosome often represents a dominant microtubule-organizing center (MTOC). However, even in cells with a radial microtubule system, many microtubules are not anchored at the centrosome, but are instead linked to the Golgi apparatus or other structures. Non-centrosomal microtubules predominate in many types of differentiated cell and in mitotic spindles. In this review, we discuss recent advances in understanding how the organization of centrosomal and non-centrosomal microtubule networks is controlled by proteins involved in microtubule nucleation and specific factors that recognize free microtubule minus ends and regulate their localization and dynamics.
Collapse
|
44
|
Martin M, Veloso A, Wu J, Katrukha EA, Akhmanova A. Control of endothelial cell polarity and sprouting angiogenesis by non-centrosomal microtubules. eLife 2018; 7:33864. [PMID: 29547120 PMCID: PMC5898915 DOI: 10.7554/elife.33864] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2017] [Accepted: 03/13/2018] [Indexed: 12/11/2022] Open
Abstract
Microtubules control different aspects of cell polarization. In cells with a radial microtubule system, a pivotal role in setting up asymmetry is attributed to the relative positioning of the centrosome and the nucleus. Here, we show that centrosome loss had no effect on the ability of endothelial cells to polarize and move in 2D and 3D environments. In contrast, non-centrosomal microtubules stabilized by the microtubule minus-end-binding protein CAMSAP2 were required for directional migration on 2D substrates and for the establishment of polarized cell morphology in soft 3D matrices. CAMSAP2 was also important for persistent endothelial cell sprouting during in vivo zebrafish vessel development. In the absence of CAMSAP2, cell polarization in 3D could be partly rescued by centrosome depletion, indicating that in these conditions the centrosome inhibited cell polarity. We propose that CAMSAP2-protected non-centrosomal microtubules are needed for establishing cell asymmetry by enabling microtubule enrichment in a single-cell protrusion. Networks of blood vessels grow like trees. Sprouts appear on existing vessels, stretching out to form new branches in a process called angiogenesis. The cells responsible are the same cells that line the finished vessels. These “endothelial cells” start the process by reorganizing themselves to face the direction of the new sprout, changing shape to become asymmetrical, and then they begin to migrate. Beneath the surface, a network of protein scaffolding supports each migrating cell. The scaffolding includes tube-like fibers called microtubules that extend towards the cell membrane and organize the inside of the cell. Destroying microtubules damages blood vessel formation, but their exact role remains unclear. A structure called the centrosome can organize microtubules within cells. The centrosome was generally believed to act like a compass, pointing in the direction that the cell will move. Microtubules can anchor to the centrosome, and this structure is thought to play an important role in cell migration. Yet, many microtubules organize without it; these microtubules instead are organized by a compartment of the cell called the Golgi apparatus and are stabilized by a protein named CAMSAP2. Martin et al. now report that removing the cells’ centrosomes did not affect cell migration, but getting rid of CAMSAP2 did. Analysis of cell shape and movement in cells grown in the laboratory and in living animals revealed that cells cannot become asymmetrical, or “polarize”, and migrate without CAMSAP2. In a two-dimensional wound-healing assay, a sheet of cells originally grown from the vessels of a human umbilical cord was scratched, and a microscope was then used to record the cell’s movement as they repaired the injury. Normally, the cells on either side move in a straight line using their microtubules, and though the process was not affected in cells without centrosomes, it was in those without CAMSAP2. Even more striking results were seen in three-dimensional assays. When the same blood vessel cells from human umbilical cords are grown as spheres inside collagen gels, they form sprouts as they would in the body. Without CAMSAP2, the cells could not organize their microtubules and they were unable to elongate in one direction and form stable sprouts. Lastly, depleting CAMSAP2 also prevented the normal formation of blood vessels in zebrafish embryos. Taken together, these findings change our understanding of how microtubules affect cell movement and how important the centrosome is for this process. Further work could have an impact on human health, not least in cancer research. Tumors need a good blood supply to grow, so understanding how to block blood vessel formation could lead to new treatments. Microtubules are already a target for cancer therapy, so future work could help to optimize the use of existing drugs.
Collapse
Affiliation(s)
- Maud Martin
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Alexandra Veloso
- Interdisciplinary Cluster for Applied Genoproteomics, University of Liège, Liège, Belgium.,GIGA-Molecular Biology in Diseases, University of Liège, Liège, Belgium
| | - Jingchao Wu
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Eugene A Katrukha
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
45
|
Ong ST, Chalasani MLS, Fazil MHUT, Prasannan P, Kizhakeyil A, Wright GD, Kelleher D, Verma NK. Centrosome- and Golgi-Localized Protein Kinase N-Associated Protein Serves As a Docking Platform for Protein Kinase A Signaling and Microtubule Nucleation in Migrating T-Cells. Front Immunol 2018; 9:397. [PMID: 29545805 PMCID: PMC5837996 DOI: 10.3389/fimmu.2018.00397] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Accepted: 02/13/2018] [Indexed: 02/01/2023] Open
Abstract
Centrosome- and Golgi-localized protein kinase N-associated protein (CG-NAP), also known as AKAP450, is a cytosolic scaffolding protein involved in the targeted positioning of multiple signaling molecules, which are critical for cellular functioning. Here, we show that CG-NAP is predominantly expressed in human primary T-lymphocytes, localizes in close proximity (<0.2 μm) with centrosomal and Golgi structures and serves as a docking platform for Protein Kinase A (PKA). GapmeR-mediated knockdown of CG-NAP inhibits LFA-1-induced T-cell migration and impairs T-cell chemotaxis toward the chemokine SDF-1α. Depletion of CG-NAP dislocates PKARIIα, disrupts centrosomal and non-centrosomal microtubule nucleation, causes Golgi fragmentation, and impedes α-tubulin tyrosination and acetylation, which are important for microtubule dynamics and stability in migrating T-cells. Furthermore, we show that CG-NAP coordinates PKA-mediated phosphorylation of pericentrin and dynein in T-cells. Overall, our findings provide critical insights into the roles of CG-NAP in regulating cytoskeletal architecture and T-cell migration.
Collapse
Affiliation(s)
- Seow Theng Ong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - M H U Turabe Fazil
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Praseetha Prasannan
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Atish Kizhakeyil
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | | | - Dermot Kelleher
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Department of Medicine, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Navin Kumar Verma
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore.,Singapore Eye Research Institute, Singapore, Singapore
| |
Collapse
|
46
|
Dubois F, Alpha K, Turner CE. Paxillin regulates cell polarization and anterograde vesicle trafficking during cell migration. Mol Biol Cell 2017; 28:3815-3831. [PMID: 29046398 PMCID: PMC5739297 DOI: 10.1091/mbc.e17-08-0488] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 09/26/2017] [Accepted: 10/13/2017] [Indexed: 12/25/2022] Open
Abstract
Cell polarization and directed migration play pivotal roles in diverse physiological and pathological processes. Herein, we identify new roles for paxillin-mediated HDAC6 inhibition in regulating key aspects of cell polarization in both two-dimensional and one-dimensional matrix environments. Paxillin, by modulating microtubule acetylation through HDAC6 regulation, was shown to control centrosome and Golgi reorientation toward the leading edge, a hallmark of cell polarization to ensure directed trafficking of promigratory factors. Paxillin was also required for pericentrosomal Golgi localization and centrosome cohesion, independent of its localization to, and role in, focal adhesion signaling. In addition, we provide evidence of an accumulation of paxillin at the centrosome that is dependent on focal adhesion kinase (FAK) and identify an important collaboration between paxillin and FAK signaling in the modulation of microtubule acetylation, as well as centrosome and Golgi organization and polarization. Finally, paxillin was also shown to be required for optimal anterograde vesicular trafficking to the plasma membrane.
Collapse
Affiliation(s)
- Fatemeh Dubois
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Kyle Alpha
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| | - Christopher E Turner
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, NY 13210
| |
Collapse
|
47
|
Yang C, Wu J, de Heus C, Grigoriev I, Liv N, Yao Y, Smal I, Meijering E, Klumperman J, Qi RZ, Akhmanova A. EB1 and EB3 regulate microtubule minus end organization and Golgi morphology. J Cell Biol 2017; 216:3179-3198. [PMID: 28814570 PMCID: PMC5626540 DOI: 10.1083/jcb.201701024] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 06/08/2017] [Accepted: 07/18/2017] [Indexed: 12/19/2022] Open
Abstract
End-binding proteins regulate the dynamics and function of microtubule plus ends by recruiting a plethora of diverse factors. Yang et al. show that EB1 and EB3 also affect microtubule minus ends by participating in their attachment to Golgi membranes. This function is important for cell polarity and migration. End-binding proteins (EBs) are the core components of microtubule plus end tracking protein complexes, but it is currently unknown whether they are essential for mammalian microtubule organization. Here, by using CRISPR/Cas9-mediated knockout technology, we generated stable cell lines lacking EB2 and EB3 and the C-terminal partner-binding half of EB1. These cell lines show only mild defects in cell division and microtubule polymerization. However, the length of CAMSAP2-decorated stretches at noncentrosomal microtubule minus ends in these cells is reduced, microtubules are detached from Golgi membranes, and the Golgi complex is more compact. Coorganization of microtubules and Golgi membranes depends on the EB1/EB3–myomegalin complex, which acts as membrane–microtubule tether and counteracts tight clustering of individual Golgi stacks. Disruption of EB1 and EB3 also perturbs cell migration, polarity, and the distribution of focal adhesions. EB1 and EB3 thus affect multiple interphase processes and have a major impact on microtubule minus end organization.
Collapse
Affiliation(s)
- Chao Yang
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Jingchao Wu
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Cecilia de Heus
- Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Ilya Grigoriev
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Nalan Liv
- Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Yao Yao
- Department of Medical Informatics, Biomedical Imaging Group Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Radiology, Biomedical Imaging Group Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Ihor Smal
- Department of Medical Informatics, Biomedical Imaging Group Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Radiology, Biomedical Imaging Group Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Erik Meijering
- Department of Medical Informatics, Biomedical Imaging Group Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands.,Department of Radiology, Biomedical Imaging Group Rotterdam, Erasmus University Medical Center, Rotterdam, Netherlands
| | - Judith Klumperman
- Department of Cell Biology, Center for Molecular Medicine, University Medical Center Utrecht, Utrecht, Netherlands
| | - Robert Z Qi
- Division of Life Science, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
48
|
Jiang P, Li Y, Poleshko A, Medvedeva V, Baulina N, Zhang Y, Zhou Y, Slater CM, Pellegrin T, Wasserman J, Lindy M, Efimov A, Daly M, Katz RA, Chen X. The Protein Encoded by the CCDC170 Breast Cancer Gene Functions to Organize the Golgi-Microtubule Network. EBioMedicine 2017; 22:28-43. [PMID: 28687497 PMCID: PMC5552109 DOI: 10.1016/j.ebiom.2017.06.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Revised: 06/23/2017] [Accepted: 06/23/2017] [Indexed: 12/17/2022] Open
Abstract
Genome-Wide Association Studies (GWAS) and subsequent fine-mapping studies (>50) have implicated single nucleotide polymorphisms (SNPs) located at the CCDC170/C6ORF97-ESR1 locus (6q25.1) as being associated with the risk of breast cancer. Surprisingly, our analysis using genome-wide differential allele-specific expression (DASE), an indicator for breast cancer susceptibility, suggested that the genetic alterations of CCDC170, but not ESR1, account for GWAS-associated breast cancer risk at this locus. Breast cancer-associated CCDC170 nonsense mutations and rearrangements have also been detected, with the latter being specifically implicated in driving breast cancer. Here we report that the wild type CCDC170 protein localizes to the region of the Golgi apparatus and binds Golgi-associated microtubules (MTs), and that breast cancer-linked truncations of CCDC170 result in loss of Golgi localization. Overexpression of wild type CCDC170 triggers Golgi reorganization, and enhances Golgi-associated MT stabilization and acetyltransferase ATAT1-dependent α-tubulin acetylation. Golgi-derived MTs regulate cellular polarity and motility, and we provide evidence that dysregulation of CCDC170 affects polarized cell migration. Taken together, our findings demonstrate that CCDC170 plays an essential role in Golgi-associated MT organization and stabilization, and implicate a mechanism for how perturbations in the CCDC170 gene may contribute to the hallmark changes in cell polarity and motility seen in breast cancer.
Collapse
Affiliation(s)
- Pengtao Jiang
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Yueran Li
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Andrey Poleshko
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Valentina Medvedeva
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Natalia Baulina
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Yongchao Zhang
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Yan Zhou
- Department of Biostatistics and Bioinformatics, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Carolyn M Slater
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Trinity Pellegrin
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Jason Wasserman
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Michael Lindy
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Andrey Efimov
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Mary Daly
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Richard A Katz
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| | - Xiaowei Chen
- Cancer Epigenetics Program, Fox Chase Cancer Center, Philadelphia, PA 19111, United States.
| |
Collapse
|
49
|
Abstract
The organization of microtubule networks is crucial for controlling chromosome segregation during cell division, for positioning and transport of different organelles, and for cell polarity and morphogenesis. The geometry of microtubule arrays strongly depends on the localization and activity of the sites where microtubules are nucleated and where their minus ends are anchored. Such sites are often clustered into structures known as microtubule-organizing centers, which include the centrosomes in animals and spindle pole bodies in fungi. In addition, other microtubules, as well as membrane compartments such as the cell nucleus, the Golgi apparatus, and the cell cortex, can nucleate, stabilize, and tether microtubule minus ends. These activities depend on microtubule-nucleating factors, such as γ-tubulin-containing complexes and their activators and receptors, and microtubule minus end-stabilizing proteins with their binding partners. Here, we provide an overview of the current knowledge on how such factors work together to control microtubule organization in different systems.
Collapse
Affiliation(s)
- Jingchao Wu
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands; ,
| | - Anna Akhmanova
- Cell Biology, Department of Biology, Faculty of Science, Utrecht University, 3584 Utrecht, The Netherlands; ,
| |
Collapse
|
50
|
Golgi trafficking defects in postnatal microcephaly: The evidence for “Golgipathies”. Prog Neurobiol 2017; 153:46-63. [DOI: 10.1016/j.pneurobio.2017.03.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 02/22/2017] [Accepted: 03/29/2017] [Indexed: 12/17/2022]
|