1
|
Guo JY, Xu K, Wang XH, Li XM, Ku YP, Zeng L, Wan B, Yang GY, Wang J, Chu BB, Pan JJ, Hao WB. Host factor DIAPH1 regulates pseudorabivirus replication by modulating the dynamics of cytoskeleton. Int J Biol Macromol 2025; 298:140112. [PMID: 39842589 DOI: 10.1016/j.ijbiomac.2025.140112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 01/05/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025]
Abstract
As obligate parasites, viruses exploit host cell organelles and molecular components to complete their life cycle. Among which, viruses firstly hijack the cytoskeleton of host cells to ensure their efficiently cell entry and replication. Although formin family members play a key role in both microfilament and microtubule cytoskeletal remodeling, few studies addressed the detailed function and mechanism of formins in the process of viral infection. Here, we showed that sus scrofa DIAPH1 was involved in the regulation of cytoskeletal dynamics during PRV replication. Firstly, we found that DIAPH1 showed significant changes in the expression level and intracellular localization during PRV infection of PK-15 cells. Next, inhibition of DIAPH1 by RNA interference or small molecular inhibitor SMIFH2 was found to diminish the outcome of PRV infection. Besides, DIAPH1 partially co-localized with actin and tubulin in PRV-infected cells. Cross-talk occurred between microfilaments and microfilaments, which also had an influence on the intracellular localization of DIAPH1. What's more, inhibition of DIAPH1 induced the reorganization of microfilament and the stability of microtubule. These results suggested that DIAPH1 regulated PRV infection by remodeling microfilament and microtubule cytoskeletal dynamics.
Collapse
Affiliation(s)
- Jie-Yuan Guo
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Kun Xu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Xiao-Han Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Xin-Man Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Yan-Pei Ku
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Lei Zeng
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China
| | - Bo Wan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Guo-Yu Yang
- Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China; Henan University of Animal Husbandry and Economy, Zhengzhou 450047, China
| | - Jiang Wang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Bei-Bei Chu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China
| | - Jia-Jia Pan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China; Key Laboratory of Animal Biochemistry and Nutrition, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Zhengzhou 450046, China; Key Laboratory of Veterinary Biotechnology of Henan Province, Zhengzhou 450046, China; Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Zhengzhou 450046, China.
| | - Wen-Bo Hao
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou 450046, China.
| |
Collapse
|
2
|
Wang J, Daniszewski M, Hao MM, Hernández D, Pébay A, Gleeson PA, Fourriere L. Organelle mapping in dendrites of human iPSC-derived neurons reveals dynamic functional dendritic Golgi structures. Cell Rep 2023; 42:112709. [PMID: 37393622 DOI: 10.1016/j.celrep.2023.112709] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Revised: 05/15/2023] [Accepted: 06/12/2023] [Indexed: 07/04/2023] Open
Abstract
Secretory pathways within dendrites of neurons have been proposed for local transport of newly synthesized proteins. However, little is known about the dynamics of the local secretory system and whether the organelles are transient or stable structures. Here, we quantify the spatial and dynamic behavior of dendritic Golgi and endosomes during differentiation of human neurons generated from induced pluripotent stem cells (iPSCs). In early neuronal development, before and during migration, the entire Golgi apparatus transiently translocates from the soma into dendrites. In mature neurons, dynamic Golgi elements, containing cis and trans cisternae, are transported from the soma along dendrites, in an actin-dependent process. Dendritic Golgi outposts are dynamic and display bidirectional movement. Similar structures were observed in cerebral organoids. Using the retention using selective hooks (RUSH) system, Golgi resident proteins are transported efficiently into Golgi outposts from the endoplasmic reticulum. This study reveals dynamic, functional Golgi structures in dendrites and a spatial map for investigating dendrite trafficking in human neurons.
Collapse
Affiliation(s)
- Jingqi Wang
- The Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Maciej Daniszewski
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Marlene M Hao
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Damián Hernández
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Alice Pébay
- Department of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010, Australia; Department of Surgery, Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Paul A Gleeson
- The Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia.
| | - Lou Fourriere
- The Department of Biochemistry and Pharmacology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, VIC 3010, Australia.
| |
Collapse
|
3
|
Lungu C, Meyer F, Hörning M, Steudle J, Braun A, Noll B, Benz D, Fränkle F, Schmid S, Eisler SA, Olayioye MA. Golgi screen identifies the RhoGEF Solo as a novel regulator of RhoB and endocytic transport. Traffic 2023; 24:162-176. [PMID: 36562184 DOI: 10.1111/tra.12880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 12/14/2022] [Indexed: 12/24/2022]
Abstract
The control of intracellular membrane trafficking by Rho GTPases is central to cellular homeostasis. How specific guanine nucleotide exchange factors and GTPase-activating proteins locally balance GTPase activation in this process is nevertheless largely unclear. By performing a microscopy-based RNAi screen, we here identify the RhoGEF protein Solo as a functional counterplayer of DLC3, a RhoGAP protein with established roles in membrane trafficking. Biochemical, imaging and optogenetics assays further uncover Solo as a novel regulator of endosomal RhoB. Remarkably, we find that Solo and DLC3 control not only the activity, but also total protein levels of RhoB in an antagonistic manner. Together, the results of our study uncover the first functionally connected RhoGAP-RhoGEF pair at endomembranes, placing Solo and DLC3 at the core of endocytic trafficking.
Collapse
Affiliation(s)
- Cristiana Lungu
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - Florian Meyer
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Marcel Hörning
- Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany.,Institute of Biomaterials and Biomolecular Systems, Biobased Materials Group, University of Stuttgart, Stuttgart, Germany
| | - Jasmin Steudle
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Anja Braun
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Bettina Noll
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - David Benz
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Felix Fränkle
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Simone Schmid
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Stephan A Eisler
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| | - Monilola A Olayioye
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany.,Stuttgart Research Center Systems Biology (SRCSB), University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
4
|
Khuntia P, Rawal S, Marwaha R, Das T. Actin-driven Golgi apparatus dispersal during collective migration of epithelial cells. Proc Natl Acad Sci U S A 2022; 119:e2204808119. [PMID: 35749357 PMCID: PMC9245705 DOI: 10.1073/pnas.2204808119] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 04/20/2022] [Indexed: 12/26/2022] Open
Abstract
As a sedentary epithelium turns motile during wound healing, morphogenesis, and metastasis, the Golgi apparatus moves from an apical position, above the nucleus, to a basal position. This apical-to-basal repositioning of Golgi is critical for epithelial cell migration. Yet the molecular mechanism underlying it remains elusive, although microtubules are believed to play a role. Using live-cell and super-resolution imaging, we show that at the onset of collective migration of epithelial cells, Golgi stacks get dispersed to create an unpolarized transitional structure, and surprisingly, this dispersal process depends not on microtubules but on actin cytoskeleton. Golgi-actin interaction involves Arp2/3-driven actin projections emanating from the actin cortex, and a Golgi-localized actin elongation factor, MENA. While in sedentary epithelial cells, actin projections intermittently interact with the apically located Golgi, and the frequency of this event increases before the dispersion of Golgi stacks, at the onset of cell migration. Preventing Golgi-actin interaction with MENA-mutants eliminates Golgi dispersion and reduces the persistence of cell migration. Taken together, we show a process of actin-driven Golgi dispersion that is mechanistically different from the well-known Golgi apparatus fragmentation during mitosis and is essential for collective migration of epithelial cells.
Collapse
Affiliation(s)
- Purnati Khuntia
- Tata Institute of Fundamental Research Hyderabad, Hyderabad 500 046, India
| | - Simran Rawal
- Tata Institute of Fundamental Research Hyderabad, Hyderabad 500 046, India
| | - Rituraj Marwaha
- Tata Institute of Fundamental Research Hyderabad, Hyderabad 500 046, India
| | - Tamal Das
- Tata Institute of Fundamental Research Hyderabad, Hyderabad 500 046, India
| |
Collapse
|
5
|
Loss of KAP3 decreases intercellular adhesion and impairs intracellular transport of laminin in signet ring cell carcinoma of the stomach. Sci Rep 2022; 12:5050. [PMID: 35322078 PMCID: PMC8943207 DOI: 10.1038/s41598-022-08904-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 03/14/2022] [Indexed: 12/14/2022] Open
Abstract
Signet-ring cell carcinoma (SRCC) is a unique subtype of gastric cancer that is impaired for cell-cell adhesion. The pathogenesis of SRCC remains unclear. Here, we show that expression of kinesin-associated protein 3 (KAP3), a cargo adaptor subunit of the kinesin superfamily protein 3 (KIF3), a motor protein, is specifically decreased in SRCC of the stomach. CRISPR/Cas9-mediated gene knockout experiments indicated that loss of KAP3 impairs the formation of circumferential actomyosin cables by inactivating RhoA, leading to the weakening of cell-cell adhesion. Furthermore, in KAP3 knockout cells, post-Golgi transport of laminin, a key component of the basement membrane, was inhibited, resulting in impaired basement membrane formation. Together, these findings uncover a potential role for KAP3 in the pathogenesis of SRCC of the stomach.
Collapse
|
6
|
Mascanzoni F, Iannitti R, Colanzi A. Functional Coordination among the Golgi Complex, the Centrosome and the Microtubule Cytoskeleton during the Cell Cycle. Cells 2022; 11:cells11030354. [PMID: 35159164 PMCID: PMC8834581 DOI: 10.3390/cells11030354] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
The Golgi complex of mammalian cells is organized in a ribbon-like structure often closely associated with the centrosome during interphase. Conversely, the Golgi complex assumes a fragmented and dispersed configuration away from the centrosome during mitosis. The structure of the Golgi complex and the relative position to the centrosome are dynamically regulated by microtubules. Many pieces of evidence reveal that this microtubule-mediated dynamic association between the Golgi complex and centrosome is of functional significance in cell polarization and division. Here, we summarize findings indicating how the Golgi complex and the centrosome cooperate in organizing the microtubule network for the directional protein transport and centrosome positioning required for cell polarization and regulating fundamental cell division processes.
Collapse
|
7
|
Mahlandt EK, Arts JJG, van der Meer WJ, van der Linden FH, Tol S, van Buul JD, Gadella TWJ, Goedhart J. Visualizing endogenous Rho activity with an improved localization-based, genetically encoded biosensor. J Cell Sci 2021; 134:272101. [PMID: 34357388 PMCID: PMC8445605 DOI: 10.1242/jcs.258823] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 07/26/2021] [Indexed: 12/05/2022] Open
Abstract
Rho GTPases are regulatory proteins, which orchestrate cell features such as morphology, polarity and movement. Therefore, probing Rho GTPase activity is key to understanding processes such as development and cell migration. Localization-based reporters for active Rho GTPases are attractive probes to study Rho GTPase-mediated processes in real time with subcellular resolution in living cells and tissue. Until now, relocation Rho biosensors (sensors that relocalize to the native location of active Rho GTPase) seem to have been only useful in certain organisms and have not been characterized well. In this paper, we systematically examined the contribution of the fluorescent protein and Rho-binding peptides on the performance of localization-based sensors. To test the performance, we compared relocation efficiency and specificity in cell-based assays. We identified several improved localization-based, genetically encoded fluorescent biosensors for detecting endogenous Rho activity. This enables a broader application of Rho relocation biosensors, which was demonstrated by using the improved biosensor to visualize Rho activity during several cellular processes, such as cell division, migration and G protein-coupled receptor signaling. Owing to the improved avidity of the new biosensors for Rho activity, cellular processes regulated by Rho can be better understood. This article has an associated First Person interview with the first author of the paper. Summary: The dT-2xrGBD location-based Rho biosensor relocalizes more efficiently than other sensors of this type, and this sensor enables the observation of endogenous Rho activity in cultured cells.
Collapse
Affiliation(s)
- Eike K Mahlandt
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Janine J G Arts
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.,Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Werner J van der Meer
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Franka H van der Linden
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Simon Tol
- Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Jaap D van Buul
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands.,Molecular Cell Biology Lab at Dept. Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Theodorus W J Gadella
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| | - Joachim Goedhart
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, Science Park 904, 1098 XH, Amsterdam, The Netherlands
| |
Collapse
|
8
|
Tokuo H, Komaba S, Coluccio LM. In pancreatic β-cells myosin 1b regulates glucose-stimulated insulin secretion by modulating an early step in insulin granule trafficking from the Golgi. Mol Biol Cell 2021; 32:1210-1220. [PMID: 33826361 PMCID: PMC8351557 DOI: 10.1091/mbc.e21-03-0094] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Pancreatic β-cells secrete insulin, which controls blood glucose levels, and defects in insulin secretion are responsible for diabetes mellitus. The actin cytoskeleton and some myosins support insulin granule trafficking and release, although a role for the class I myosin Myo1b, an actin- and membrane-associated load-sensitive motor, in insulin biology is unknown. We found by immunohistochemistry that Myo1b is expressed in islet cells of the rat pancreas. In cultured rat insulinoma 832/13 cells, Myo1b localized near actin patches, the trans-Golgi network (TGN) marker TGN38, and insulin granules in the perinuclear region. Myo1b depletion by small interfering RNA in 832/13 cells reduced intracellular proinsulin and insulin content and glucose-stimulated insulin secretion (GSIS) and led to the accumulation of (pro)insulin secretory granules (SGs) at the TGN. Using an in situ fluorescent pulse-chase strategy to track nascent proinsulin, Myo1b depletion in insulinoma cells reduced the number of (pro)insulin-containing SGs budding from the TGN. The studies indicate for the first time that in pancreatic β-cells Myo1b controls GSIS at least in part by mediating an early stage in insulin granule trafficking from the TGN.
Collapse
Affiliation(s)
- Hiroshi Tokuo
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118-2518
| | - Shigeru Komaba
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118-2518
| | - Lynne M Coluccio
- Department of Physiology & Biophysics, Boston University School of Medicine, Boston, MA 02118-2518
| |
Collapse
|
9
|
Palander O, Trimble WS. DIAPH1 regulates ciliogenesis and trafficking in primary cilia. FASEB J 2020; 34:16516-16535. [PMID: 33124112 DOI: 10.1096/fj.202001178r] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 09/08/2020] [Accepted: 10/12/2020] [Indexed: 01/29/2023]
Abstract
Primary cilia are critical hubs for several signaling pathways, and defects in ciliogenesis or cilia maintenance produce a range of diseases collectively known as ciliopathies. Ciliogenesis requires vesicle trafficking along a network of microtubules and actin filaments to the basal body. The DIAPH1 (Diaphanous-related formin) family of formins promotes both actin polymerization and EB1-dependent microtubule (MT) stability. EB1 and EB3 have previously been implicated in cilia biogenesis to carry out centrosome-related functions. However, the role of DIAPH1 proteins had not been examined. Here we show that the depletion of DIAPH1 decreased ciliogenesis, cilia length, and reduced trafficking within cilia. Additionally, both actin nucleating and microtubule-stabilizing properties of DIAPH1 are important for their cilia functions. To assess their roles in ciliogenesis in isolation, we targeted DIAPH1 specifically to the basal body, which caused an increase in cilia length and increased trafficking within cilia. Intriguingly, expression of DIAPH1 mutants associated with human deafness and microcephaly impaired ciliation and caused cilia elongation and bulb formation. These results suggest that the actin and microtubule functions of DIAPH1 proteins regulate cilia maintenance in part by regulating vesicular trafficking to the base of the primary cilia.
Collapse
Affiliation(s)
- Oliva Palander
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - William S Trimble
- Cell Biology Program, Hospital for Sick Children, Toronto, ON, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Phuyal S, Baschieri F. Endomembranes: Unsung Heroes of Mechanobiology? Front Bioeng Biotechnol 2020; 8:597721. [PMID: 33195167 PMCID: PMC7642594 DOI: 10.3389/fbioe.2020.597721] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Accepted: 10/05/2020] [Indexed: 12/18/2022] Open
Abstract
Mechanical stimuli have profound effects on the cellular architecture and functions. Over the past two decades, considerable progress has been made in unraveling the molecular machineries that confer cells the ability to sense and transduce mechanical input into biochemical signals. This has resulted in the identification of several force-sensing proteins or mechanically activated ion channels distributed throughout most cell types, whereby the plasma membrane, cytoskeleton, and the nucleus have garnered much attention. Although organelles from the endomembrane system make up significant portion of cell volume and play pivotal roles in the spatiotemporal distribution of signaling molecules, they have received surprisingly little attention in mechanobiology. In this mini-review, we summarize results that document participation of the endomembrane system in sensing and responding to mechanical cues.
Collapse
Affiliation(s)
- Santosh Phuyal
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Francesco Baschieri
- Inserm U1279, Gustave Roussy Institute, Université Paris-Saclay, Villejuif, France
| |
Collapse
|
11
|
Abstract
Rho GTPases are known to play an essential role in fundamental processes such as defining cell shape, polarity and migration. As such, the majority of Rho GTPases localize and function at, or close to, the plasma membrane. However, it is becoming increasingly clear that a number of Rho family proteins are also associated with the Golgi complex, where they not only regulate events at this organelle but also more widely across the cell. Given the central location of this organelle, and the numerous membrane trafficking pathways that connect it to both the endocytic and secretory systems of cells, it is clear that the Golgi is fundamental for maintaining cellular homoeostasis. In this review, we describe these GTPases in the context of how they regulate Golgi architecture, membrane trafficking into and away from this organelle, and cell polarity and migration. We summarize the key findings that show the growing importance of the pool of Rho GTPases associated with Golgi function, namely Cdc42, RhoA, RhoD, RhoBTB1 and RhoBTB3, and we discuss how they act in concert with other key families of molecules associated with the Golgi, including Rab GTPases and matrix proteins.
Collapse
Affiliation(s)
- Margaritha M Mysior
- Cell Screening Laboratory, School of Biology & Environmental Science, University College Dublin (UCD), Dublin Ireland
| | - Jeremy C Simpson
- Cell Screening Laboratory, School of Biology & Environmental Science, University College Dublin (UCD), Dublin Ireland
| |
Collapse
|
12
|
Modzelewska K, Brown L, Culotti J, Moghal N. Sensory regulated Wnt production from neurons helps make organ development robust to environmental changes in C. elegans. Development 2020; 147:dev186080. [PMID: 32586974 DOI: 10.1242/dev.186080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Accepted: 06/13/2020] [Indexed: 11/20/2022]
Abstract
Long-term survival of an animal species depends on development being robust to environmental variations and climate changes. We used C. elegans to study how mechanisms that sense environmental changes trigger adaptive responses that ensure animals develop properly. In water, the nervous system induces an adaptive response that reinforces vulval development through an unknown backup signal for vulval induction. This response involves the heterotrimeric G-protein EGL-30//Gαq acting in motor neurons. It also requires body-wall muscle, which is excited by EGL-30-stimulated synaptic transmission, suggesting a behavioral function of neurons induces backup signal production from muscle. We now report that increased acetylcholine during liquid growth activates an EGL-30-Rho pathway, distinct from the synaptic transmission pathway, that increases Wnt production from motor neurons. We also provide evidence that this neuronal Wnt contributes to EGL-30-stimulated vulval development, with muscle producing a parallel developmental signal. As diverse sensory modalities stimulate motor neurons via acetylcholine, this mechanism enables broad sensory perception to enhance Wnt-dependent development. Thus, sensory perception improves animal fitness by activating distinct neuronal functions that trigger adaptive changes in both behavior and developmental processes.
Collapse
Affiliation(s)
- Katarzyna Modzelewska
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
| | - Louise Brown
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Joseph Culotti
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario, M5G 1X5, Canada
| | - Nadeem Moghal
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT 84112, USA
- Princess Margaret Cancer Centre/University Health Network, Toronto, Ontario, M5G 1L7, Canada
| |
Collapse
|
13
|
Lázaro-Diéguez F, Müsch A. Low Rho activity in hepatocytes prevents apical from basolateral cargo separation during trans-Golgi network to surface transport. Traffic 2020; 21:364-374. [PMID: 32124512 PMCID: PMC7959587 DOI: 10.1111/tra.12725] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Revised: 02/27/2020] [Accepted: 02/28/2020] [Indexed: 12/24/2022]
Abstract
Hepatocytes, the main epithelial cells of the liver, organize their polarized membrane domains differently from ductal epithelia. They also differ in their biosynthetic delivery of single-membrane-spanning and glycophosphatidylinositol-anchored proteins to the apical domain. While ductal epithelia target apical proteins to varying degrees from the trans-Golgi network (TGN) to the apical surface directly, hepatocytes target them first to the basolateral domain, from where they undergo basolateral-to-apical transcytosis. How TGN-to-surface transport differs in both scenarios is unknown. Here, we report that the basolateral detour of a hepatocyte apical protein is due, in part, to low RhoA activity at the TGN, which prevents its segregation from basolateral transport carriers. Activating Rho in hepatocytic cells, which switches their polarity from hepatocytic to ductal, also led to apical-basolateral cargo segregation at the TGN as is typical for ductal cells, affirming a central role for Rho-signaling in different aspects of the hepatocytic polarity phenotype. Nevertheless, Rho-induced cargo segregation was not sufficient to target the apical protein directly; thus, failure to recruit apical targeting machinery also contributes to its indirect itinerary.
Collapse
Affiliation(s)
- Francisco Lázaro-Diéguez
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| | - Anne Müsch
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, New York
| |
Collapse
|
14
|
Long M, Kranjc T, Mysior MM, Simpson JC. RNA Interference Screening Identifies Novel Roles for RhoBTB1 and RhoBTB3 in Membrane Trafficking Events in Mammalian Cells. Cells 2020; 9:cells9051089. [PMID: 32354068 PMCID: PMC7291084 DOI: 10.3390/cells9051089] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 04/22/2020] [Accepted: 04/25/2020] [Indexed: 12/19/2022] Open
Abstract
In the endomembrane system of mammalian cells, membrane traffic processes require a high degree of regulation in order to ensure their specificity. The range of molecules that participate in trafficking events is truly vast, and much attention to date has been given to the Rab family of small GTPases. However, in recent years, a role in membrane traffic for members of the Rho GTPase family, in particular Cdc42, has emerged. This prompted us to develop and apply an image-based high-content screen, initially focussing on the Golgi complex, using RNA interference to systematically perturb each of the 21 Rho family members and assess their importance to the overall organisation of this organelle. Analysis of our data revealed previously unreported roles for two atypical Rho family members, RhoBTB1 and RhoBTB3, in membrane traffic events. We find that depletion of RhoBTB3 affects the morphology of the Golgi complex and causes changes in the trafficking speeds of carriers operating at the interface of the Golgi and endoplasmic reticulum. In addition, RhoBTB3 was found to be present on these carriers. Depletion of RhoBTB1 was also found to cause a disturbance to the Golgi architecture, however, this phenotype seems to be linked to endocytosis and retrograde traffic pathways. RhoBTB1 was found to be associated with early endosomal intermediates, and changes in the levels of RhoBTB1 not only caused profound changes to the organisation and distribution of endosomes and lysosomes, but also resulted in defects in the delivery of two different classes of cargo molecules to downstream compartments. Together, our data reveal new roles for these atypical Rho family members in the endomembrane system.
Collapse
|
15
|
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: 18] [Impact Index Per Article: 3.6] [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
|
16
|
Beigl TB, Hellesvik M, Saraste J, Arnesen T, Aksnes H. N-terminal acetylation of actin by NAA80 is essential for structural integrity of the Golgi apparatus. Exp Cell Res 2020; 390:111961. [PMID: 32209306 DOI: 10.1016/j.yexcr.2020.111961] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 03/11/2020] [Accepted: 03/15/2020] [Indexed: 01/07/2023]
Abstract
N-alpha-acetyltransferase 80 (NAA80) was recently demonstrated to acetylate the N-terminus of actin, with NAA80 knockout cells showing actin cytoskeleton-related phenotypes, such as increased formation of membrane protrusions and accelerated migration. Here we report that NAA80 knockout cells additionally display fragmentation of the Golgi apparatus. We further employed rescue assays to demonstrate that this phenotype is connected to the ability of NAA80 to modify actin. Thus, re-expression of NAA80, which leads to re-establishment of actin's N-terminal acetyl group, rescued the Golgi fragmentation, whereas a catalytic dead NAA80 mutant could neither restore actin Nt-acetylation nor Golgi structure. The Golgi phenotype of NAA80 KO cells was shared by both migrating and non-migrating cells and live-cell imaging indicated increased Golgi dynamics in migrating NAA80 KO cells. Finally, we detected a drastic increase in the amount of F-actin in cells lacking NAA80, suggesting a causal relationship between this effect and the observed re-organization of Golgi structure. The findings further underscore the importance of actin Nt-acetylation and provide novel insight into its cellular roles, suggesting a mechanistic link between actin modification state and Golgi organization.
Collapse
Affiliation(s)
- Tobias B Beigl
- Department of Biomedicine, University of Bergen, Norway; Institute of Cell Biology and Immunology, University of Stuttgart, Germany
| | | | | | - Thomas Arnesen
- Department of Biomedicine, University of Bergen, Norway; Department of Biological Sciences, University of Bergen, Norway; Department of Surgery, Haukeland University Hospital, Norway
| | | |
Collapse
|
17
|
Wosik J, Suarez-Villagran M, Miller JH, Ghobrial RM, Kloc M. Macrophage phenotype bioengineered by magnetic, genetic, or pharmacologic interference. Immunol Res 2019; 67:1-11. [PMID: 30649660 DOI: 10.1007/s12026-019-9066-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In all eukaryotes, the cell shape depends on the actin filament cytoskeleton, which is regulated by the small GTPase RhoA. It is well known that the cell shape determines cell function and behavior. Inversely, any change in the cell behavior and/or function reverberates at the cell shape. In this review, we describe how mechanical/magnetic, genetic, or pharmacologic interference with the actin cytoskeleton enforces changes in cell shape and function and how such techniques can be used to control the phenotype and functions of immune cells such as macrophages and to develop novel anti-cancer and anti-rejection clinical therapies.
Collapse
Affiliation(s)
- Jarek Wosik
- Electrical and Computer Engineering Department, University of Houston, Houston, TX, 77204, USA. .,Texas Center for Superconductivity, University of Houston, HSC Bldg., Rm. 202, Houston, TX, 77204-5002, USA.
| | - Martha Suarez-Villagran
- Electrical and Computer Engineering Department, University of Houston, Houston, TX, 77204, USA.,Physics Department, University of Houston, Houston, TX, USA
| | - John H Miller
- Electrical and Computer Engineering Department, University of Houston, Houston, TX, 77204, USA.,Physics Department, University of Houston, Houston, TX, USA
| | - Rafik M Ghobrial
- The Houston Methodist Research Institute, Houston, TX, 77030, USA.,Department of Surgery, The Houston Methodist Hospital, 6550 Fannin St., Houston, TX, 77030, USA
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, 77030, USA. .,Department of Surgery, The Houston Methodist Hospital, 6550 Fannin St., Houston, TX, 77030, USA. .,M.D. Anderson Cancer Center, Department of Genetics, The University of Texas, Houston, TX, 77030, USA.
| |
Collapse
|
18
|
Higa N, Shinsato Y, Kamil M, Hirano T, Takajo T, Shimokawa M, Minami K, Yamamoto M, Kawahara K, Yonezawa H, Hirano H, Furukawa T, Yoshimoto K, Arita K. Formin-like 1 (FMNL1) Is Associated with Glioblastoma Multiforme Mesenchymal Subtype and Independently Predicts Poor Prognosis. Int J Mol Sci 2019; 20:ijms20246355. [PMID: 31861134 PMCID: PMC6940780 DOI: 10.3390/ijms20246355] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2019] [Revised: 12/11/2019] [Accepted: 12/14/2019] [Indexed: 12/18/2022] Open
Abstract
Glioblastoma multiforme (GBM), the most common primary malignant brain tumor in adults, is characterized by rapid proliferation, aggressive migration, and invasion into normal brain tissue. Formin proteins have been implicated in these processes. However, the role of formin-like 1 (FMNL1) in cancer remains unclear. We studied FMNL1 expression in glioblastoma samples using immunohistochemistry. We sought to analyze the correlation between FMNL1 expression, clinicopathologic variables, and patient survival. Migration and invasion assays were used to verify the effect of FMNL1 on glioblastoma cell lines. Microarray data were downloaded from The Cancer Genome Atlas and analyzed using gene set enrichment analysis (GSEA). FMNL1 was an independent predictor of poor prognosis in a cohort of 217 glioblastoma multiforme cases (p < 0.001). FMNL1 expression was significantly higher in the mesenchymal subtype. FMNL1 upregulation and downregulation were associated with mesenchymal and proneural markers in the GSEA, respectively. These data highlight the important role of FMNL1 in the neural-to-mesenchymal transition. Conversely, FMNL1 downregulation suppressed glioblastoma multiforme cell migration and invasion via DIAPH1 and GOLGA2, respectively. FMNL1 downregulation also suppressed actin fiber assembly, induced morphological changes, and diminished filamentous actin. FMNL1 is a promising therapeutic target and a useful biomarker for GBM progression.
Collapse
Affiliation(s)
- Nayuta Higa
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan (H.Y.); (H.H.); (K.Y.)
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
| | - Yoshinari Shinsato
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
| | - Muhammad Kamil
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan (H.Y.); (H.H.); (K.Y.)
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
- Department of Neurosurgery, Faculty of Medicine, Airlangga University, Surabaya 60132, Indonesia
| | - Takuro Hirano
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
- Department of Digestive Surgery, Breast and Thyroid Surgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan
| | - Tomoko Takajo
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan (H.Y.); (H.H.); (K.Y.)
| | - Michiko Shimokawa
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
| | - Kentaro Minami
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
| | - Masatatsu Yamamoto
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
| | - Kohichi Kawahara
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
| | - Hajime Yonezawa
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan (H.Y.); (H.H.); (K.Y.)
| | - Hirofumi Hirano
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan (H.Y.); (H.H.); (K.Y.)
| | - Tatsuhiko Furukawa
- Department of Molecular Oncology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan; (Y.S.); (T.H.); (M.S.); (K.M.); (M.Y.); (K.K.)
- Center for the Research of Advanced Diagnosis and Therapy of Cancer, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan
- Correspondence: ; Tel.: +81-99-275-5490
| | - Koji Yoshimoto
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan (H.Y.); (H.H.); (K.Y.)
| | - Kazunori Arita
- Department of Neurosurgery, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8520, Japan (H.Y.); (H.H.); (K.Y.)
| |
Collapse
|
19
|
Frisbie CP, Lushnikov AY, Krasnoslobodtsev AV, Riethoven JJM, Clarke JL, Stepchenkova EI, Petrosyan A. Post-ER Stress Biogenesis of Golgi Is Governed by Giantin. Cells 2019; 8:E1631. [PMID: 31847122 PMCID: PMC6953117 DOI: 10.3390/cells8121631] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Revised: 12/05/2019] [Accepted: 12/09/2019] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND The Golgi apparatus undergoes disorganization in response to stress, but it is able to restore compact and perinuclear structure under recovery. This self-organization mechanism is significant for cellular homeostasis, but remains mostly elusive, as does the role of giantin, the largest Golgi matrix dimeric protein. METHODS In HeLa and different prostate cancer cells, we used the model of cellular stress induced by Brefeldin A (BFA). The conformational structure of giantin was assessed by proximity ligation assay and atomic force microscopy. The post-BFA distribution of Golgi resident enzymes was examined by 3D SIM high-resolution microscopy. RESULTS We detected that giantin is rather flexible than an extended coiled-coil dimer and BFA-induced Golgi disassembly was associated with giantin monomerization. A fusion of the nascent Golgi membranes after BFA washout is forced by giantin re-dimerization via disulfide bond in its luminal domain and assisted by Rab6a GTPase. GM130-GRASP65-dependent enzymes are able to reach the nascent Golgi membranes, while giantin-sensitive enzymes appeared at the Golgi after its complete recovery via direct interaction of their cytoplasmic tail with N-terminus of giantin. CONCLUSION Post-stress recovery of Golgi is conducted by giantin dimer and Golgi proteins refill membranes according to their docking affiliation rather than their intra-Golgi location.
Collapse
Affiliation(s)
- Cole P. Frisbie
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA;
| | - Alexander Y. Lushnikov
- Nanoimaging Core Facility, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA; (A.Y.L.); (A.V.K.)
| | - Alexey V. Krasnoslobodtsev
- Nanoimaging Core Facility, University of Nebraska Medical Center, Omaha, NE 68198-6025, USA; (A.Y.L.); (A.V.K.)
- Department of Physics, University of Nebraska-Omaha, Omaha, NE 68182-0266, USA
| | - Jean-Jack M. Riethoven
- Center for Biotechnology, University of Nebraska-Lincoln, Lincoln, NE 68588-0665, USA;
- The Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA;
| | - Jennifer L. Clarke
- The Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA;
- Department of Statistics, University of Nebraska-Lincoln, Lincoln, NE 68583-0963, USA
| | - Elena I. Stepchenkova
- Vavilov Institute of General Genetics, Saint-Petersburg Branch, Russian Academy of Sciences, Saint-Petersburg 199034, Russia;
- Department of Genetics, Saint-Petersburg State University, Saint-Petersburg 199034, Russia
| | - Armen Petrosyan
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198-5870, USA;
- The Nebraska Center for Integrated Biomolecular Communication, University of Nebraska-Lincoln, Lincoln, NE 68588-0304, USA;
- The Fred and Pamela Buffett Cancer Center, Omaha, NE 68198-5870, USA
| |
Collapse
|
20
|
Olayioye MA, Noll B, Hausser A. Spatiotemporal Control of Intracellular Membrane Trafficking by Rho GTPases. Cells 2019; 8:cells8121478. [PMID: 31766364 PMCID: PMC6952795 DOI: 10.3390/cells8121478] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 11/15/2019] [Accepted: 11/18/2019] [Indexed: 12/17/2022] Open
Abstract
As membrane-associated master regulators of cytoskeletal remodeling, Rho GTPases coordinate a wide range of biological processes such as cell adhesion, motility, and polarity. In the last years, Rho GTPases have also been recognized to control intracellular membrane sorting and trafficking steps directly; however, how Rho GTPase signaling is regulated at endomembranes is still poorly understood. In this review, we will specifically address the local Rho GTPase pools coordinating intracellular membrane trafficking with a focus on the endo- and exocytic pathways. We will further highlight the spatiotemporal molecular regulation of Rho signaling at endomembrane sites through Rho regulatory proteins, the GEFs and GAPs. Finally, we will discuss the contribution of dysregulated Rho signaling emanating from endomembranes to the development and progression of cancer.
Collapse
|
21
|
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: 73] [Impact Index Per Article: 12.2] [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
|
22
|
Rodríguez-Cruz F, Torres-Cruz FM, Monroy-Ramírez HC, Escobar-Herrera J, Basurto-Islas G, Avila J, García-Sierra F. Fragmentation of the Golgi Apparatus in Neuroblastoma Cells Is Associated with Tau-Induced Ring-Shaped Microtubule Bundles. J Alzheimers Dis 2019; 65:1185-1207. [PMID: 30124450 DOI: 10.3233/jad-180547] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Abnormal fibrillary aggregation of tau protein is a pathological condition observed in Alzheimer's disease and other tauopathies; however, the presence and pathological significance of early non-fibrillary aggregates of tau remain under investigation. In cell and animal models expressing normal or modified tau, toxic effects altering the structure and function of several membranous organelles have also been reported in the absence of fibrillary structures; however, how these abnormalities are produced is an issue yet to be addressed. In order to obtain more insights into the mechanisms by which tau may disturb intracellular membranous elements, we transiently overexpressed human full-length tau and several truncated tau variants in cultured neuroblastoma cells. After 48 h of transfection, either full-length or truncated tau forms produced significant fragmentation of the Golgi apparatus (GA) with no changes in cell viability. Noteworthy is that in the majority of cells exhibiting dispersion of the GA, a ring-shaped array of cortical or perinuclear microtubule (Mt) bundles was also generated under the expression of either variant of tau. In contrast, Taxol treatment of non-transfected cells increased the amount of Mt bundles but not sufficiently to produce fragmentation of the GA. Tau-induced ring-shaped Mt bundles appeared to be well-organized and stable structures because they were resistant to Nocodazole post-treatment and displayed a high level of tubulin acetylation. These results further indicate that a mechanical force generated by tau-induced Mt-bundling may be responsible for Golgi fragmentation and that the repeated domain region of tau may be the main promoter of this effect.
Collapse
Affiliation(s)
- Fanny Rodríguez-Cruz
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | - Francisco Miguel Torres-Cruz
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | | | - Jaime Escobar-Herrera
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| | | | - Jesús Avila
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM) Universidad Autónoma de Madrid, Madrid, Spain
| | - Francisco García-Sierra
- Department of Cell Biology, Center of Research and Advanced Studies of the National Polytechnic Institute (CINVESTAV), Mexico City, Mexico
| |
Collapse
|
23
|
Kloc M, Uosef A, Wosik J, Kubiak JZ, Ghobrial RM. RhoA Pathway and Actin Regulation of the Golgi/Centriole Complex. Results Probl Cell Differ 2019; 67:81-93. [PMID: 31435793 DOI: 10.1007/978-3-030-23173-6_5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
In vertebrate cells, the Golgi apparatus is located in close proximity to the centriole. The architecture of the Golgi/centriole complex depends on a multitude of factors, including the actin filament cytoskeleton. In turn, both the Golgi and centriole act as the actin nucleation centers. Actin organization and polymerization also depend on the small GTPase RhoA pathway. In this chapter, we summarize the most current knowledge on how the genetic, magnetic, or pharmacologic interference with RhoA pathway and actin cytoskeleton directly or indirectly affects architecture, structure, and function of the Golgi/centriole complex.
Collapse
Affiliation(s)
- Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA.
- Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA.
- Department of Genetics, MD Anderson Cancer Center, The University of Texas, Houston, TX, USA.
| | - Ahmed Uosef
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA
| | - Jarek Wosik
- Department of Electrical and Computer Engineering, University of Houston, Houston, TX, USA
- Texas Center for Superconductivity, University of Houston, Houston, TX, USA
| | - Jacek Z Kubiak
- Laboratory of Epidemiology, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
- Department of Regenerative Medicine and Cell Biology, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
- Faculty of Medicine, Cell Cycle Group, Institute of Genetics and Development of Rennes, Univ Rennes, UMR 6290, CNRS, Rennes, France
| | - Rafik Mark Ghobrial
- The Houston Methodist Research Institute, Houston, TX, USA
- Department of Surgery, The Houston Methodist Hospital, Houston, TX, USA
| |
Collapse
|
24
|
Abstract
Formin homology proteins (formins) are a highly conserved family of cytoskeletal remodeling proteins that are involved in a diverse array of cellular functions. Formins are best known for their ability to regulate actin dynamics, but the same functional domains also govern stability and organization of microtubules. It is thought that this dual activity allows them to coordinate the activity of these two major cytoskeletal networks and thereby influence cellular architecture. Golgi ribbon assembly is dependent upon cooperative interactions between actin filaments and cytoplasmic microtubules originating both at the Golgi itself and from the centrosome. Similarly, centrosome assembly, centriole duplication, and centrosome positioning are also reliant on a dialogue between both cytoskeletal networks. As presented in this chapter, a growing body of evidence suggests that multiple formin proteins play essential roles in these central cellular processes.
Collapse
Affiliation(s)
- John Copeland
- Faculty of Medicine, Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
25
|
Multiple roles of the actin and microtubule-regulating formins in the developing brain. Neurosci Res 2019; 138:59-69. [DOI: 10.1016/j.neures.2018.09.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 08/22/2018] [Accepted: 08/23/2018] [Indexed: 01/08/2023]
|
26
|
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: 3.3] [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
|
27
|
Giantin Is Required for Post-Alcohol Recovery of Golgi in Liver Cells. Biomolecules 2018; 8:biom8040150. [PMID: 30453527 PMCID: PMC6316505 DOI: 10.3390/biom8040150] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/09/2018] [Accepted: 11/12/2018] [Indexed: 12/17/2022] Open
Abstract
In hepatocytes and alcohol-metabolizing cultured cells, Golgi undergoes ethanol (EtOH)-induced disorganization. Perinuclear and organized Golgi is important in liver homeostasis, but how the Golgi remains intact is unknown. Work from our laboratories showed that EtOH-altered cellular function could be reversed after alcohol removal; we wanted to determine whether this recovery would apply to Golgi. We used alcohol-metabolizing HepG2 (VA-13) cells (cultured with or without EtOH for 72 h) and rat hepatocytes (control and EtOH-fed (Lieber–DeCarli diet)). For recovery, EtOH was removed and replenished with control medium (48 h for VA-13 cells) or control diet (10 days for rats). Results: EtOH-induced Golgi disassembly was associated with de-dimerization of the largest Golgi matrix protein giantin, along with impaired transport of selected hepatic proteins. After recovery from EtOH, Golgi regained their compact structure, and alterations in giantin and protein transport were restored. In VA-13 cells, when we knocked down giantin, Rab6a GTPase or non-muscle myosin IIB, minimal changes were observed in control conditions, but post-EtOH recovery was impaired. Conclusions: These data provide a link between Golgi organization and plasma membrane protein expression and identify several proteins whose expression is important to maintain Golgi structure during the recovery phase after EtOH administration.
Collapse
|
28
|
Pan ZN, Lu Y, Tang F, Pan MH, Wan X, Lan M, Zhang Y, Sun SC. RAB8A GTPase regulates spindle migration and Golgi apparatus distribution via ROCK-mediated actin assembly in mouse oocyte meiosis†. Biol Reprod 2018; 100:711-720. [DOI: 10.1093/biolre/ioy217] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 09/03/2018] [Accepted: 09/30/2018] [Indexed: 12/13/2022] Open
Affiliation(s)
- Zhen-Nan Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yujie Lu
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Feng Tang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Meng-Hao Pan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiang Wan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Mei Lan
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yu Zhang
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Shao-Chen Sun
- College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
29
|
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: 2.9] [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
|
30
|
Eisler SA, Curado F, Link G, Schulz S, Noack M, Steinke M, Olayioye MA, Hausser A. A Rho signaling network links microtubules to PKD controlled carrier transport to focal adhesions. eLife 2018; 7:35907. [PMID: 30028295 PMCID: PMC6070338 DOI: 10.7554/elife.35907] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 07/19/2018] [Indexed: 12/22/2022] Open
Abstract
Protein kinase D (PKD) is a family of serine/threonine kinases that is required for the structural integrity and function of the Golgi complex. Despite its importance in the regulation of Golgi function, the molecular mechanisms regulating PKD activity are still incompletely understood. Using the genetically encoded PKD activity reporter G-PKDrep we now uncover a Rho signaling network comprising GEF-H1, the RhoGAP DLC3, and the Rho effector PLCε that regulate the activation of PKD at trans-Golgi membranes. We further show that this molecular network coordinates the formation of TGN-derived Rab6-positive transport carriers delivering cargo for localized exocytosis at focal adhesions.
Collapse
Affiliation(s)
- Stephan A Eisler
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Filipa Curado
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Gisela Link
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Sarah Schulz
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Melanie Noack
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany
| | - Maren Steinke
- Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Monilola A Olayioye
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.,Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Angelika Hausser
- Stuttgart Research Center Systems Biology, University of Stuttgart, Stuttgart, Germany.,Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| |
Collapse
|
31
|
Yao J, Wang J, Yao Y, Wang K, Zhou Q, Tang Y. miR‑133b regulates proliferation and apoptosis in high‑glucose‑induced human retinal endothelial cells by targeting ras homolog family member A. Int J Mol Med 2018; 42:839-850. [PMID: 29786744 PMCID: PMC6034913 DOI: 10.3892/ijmm.2018.3694] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 05/03/2018] [Indexed: 12/26/2022] Open
Abstract
The aim of the present study was to investigate the role of microRNA (miR)‑133b in high‑glucose‑induced human retinal endothelial cells (hRECs), particularly regarding its potential targeting of ras homolog family member A (RhoA). To establish the high‑glucose‑induced diabetic retinopathy (DR) model, hRECs were cultured in high‑glucose medium for 1, 2 and 3 days. An Annexin allophycocyanin (APC)/7‑aminoactinomycin D (7‑AAD) staining assay was performed to measure the apoptosis of hRECs. Next, the cells were transfected with miR‑133b inhibitors or mimics, and the cell proliferation and apoptosis were measured by MTT and Annexin‑APC/7‑AAD staining assays, respectively. In addition, reverse transcription‑quantitative polymerase chain reaction (RT‑qPCR), western blotting and immunocytochemistry were used to detect the expression levels of RhoA, Rho‑associated protein kinase 1 (ROCK1), LIM domain kinase 1 (LIMK), myosin light chain (MLC) and phosphorylated (p)‑MLC. It was observed that high‑glucose or miR‑133b inhibitor treatment attenuated the apoptosis of hRECs, and upregulated the mRNA and protein expression levels of RhoA, ROCK1 and LIMK, as well as the p‑MLC protein level, in the hRECs. However, miR‑133b overexpression inhibited the cell proliferation, promoted apoptosis, and downregulated the mRNA and protein levels of RhoA, ROCK1 and LIMK, as well as p‑MLC protein, in high‑glucose‑induced hRECs. In conclusion, overexpression of miR‑133b inhibited the proliferation and promoted apoptosis in a DR cell model by downregulating RhoA expression.
Collapse
Affiliation(s)
- Jun Yao
- Department of Ophthalmology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430014, P.R. China
| | - Jihong Wang
- Department of Ophthalmology, Wuxi Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu 214071, P.R. China
| | - Yong Yao
- Department of Ophthalmology, Wuxi People's Hospital of Nanjing Medical University, Wuxi, Jiangsu 214023, P.R. China
| | - Kelei Wang
- Department of Ophthalmology, Wuxi Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu 214071, P.R. China
| | - Qianqian Zhou
- Department of Ophthalmology, Wuxi Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu 214071, P.R. China
| | - Ying Tang
- Department of Ophthalmology, Wuxi Affiliated Hospital of Nanjing University of Traditional Chinese Medicine, Wuxi, Jiangsu 214071, P.R. China
| |
Collapse
|
32
|
BIG2-ARF1-RhoA-mDia1 Signaling Regulates Dendritic Golgi Polarization in Hippocampal Neurons. Mol Neurobiol 2018; 55:7701-7716. [PMID: 29455446 DOI: 10.1007/s12035-018-0954-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 02/05/2018] [Indexed: 01/08/2023]
Abstract
Proper dendrite development is essential for establishing neural circuitry, and Rho GTPases play key regulatory roles in this process. From mouse brain lysates, we identified Brefeldin A-inhibited guanine exchange factor 2 (BIG2) as a novel Rho GTPase regulatory protein involved in dendrite growth and maintenance. BIG2 was highly expressed during early development, and knockdown of the ARFGEF2 gene encoding BIG2 significantly reduced total dendrite length and the number of branches. Expression of the constitutively active ADP-ribosylation factor 1 ARF1 Q71L rescued the defective dendrite morphogenesis of ARFGEF2-null neurons, indicating that BIG2 controls dendrite growth and maintenance by activating ARF1. Moreover, BIG2 co-localizes with the Golgi apparatus and is required for Golgi deployment into major dendrites in cultured hippocampal neurons. Simultaneous overexpression of BIG2 and ARF1 activated RhoA, and treatment with the RhoA activator lysophosphatidic acid in neurons lacking BIG2 or ARF1 increased the number of cells with dendritic Golgi, suggesting that BIG2 and ARF1 activate RhoA to promote dendritic Golgi polarization. mDia1 was identified as a downstream effector of BIG2-ARF1-RhoA axis, mediating Golgi polarization and dendritic morphogenesis. Furthermore, in utero electroporation of ARFGEF2 shRNA into the embryonic mouse brain confirmed an in vivo role of BIG2 for Golgi deployment into the apical dendrite. Taken together, our results suggest that BIG2-ARF1-RhoA-mDia1 signaling regulates dendritic Golgi polarization and dendrite growth and maintenance in hippocampal neurons.
Collapse
|
33
|
Miserey-Lenkei S, Bousquet H, Pylypenko O, Bardin S, Dimitrov A, Bressanelli G, Bonifay R, Fraisier V, Guillou C, Bougeret C, Houdusse A, Echard A, Goud B. Coupling fission and exit of RAB6 vesicles at Golgi hotspots through kinesin-myosin interactions. Nat Commun 2017; 8:1254. [PMID: 29093437 PMCID: PMC5665954 DOI: 10.1038/s41467-017-01266-0] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Accepted: 09/04/2017] [Indexed: 01/08/2023] Open
Abstract
The actin and microtubule cytoskeletons play important roles in Golgi structure and function, but how they are connected remain poorly known. In this study, we investigated whether RAB6 GTPase, a Golgi-associated RAB involved in the regulation of several transport steps at the Golgi level, and two of its effectors, Myosin IIA and KIF20A participate in the coupling between actin and microtubule cytoskeleton. We have previously shown that RAB6–Myosin IIA interaction is critical for the fission of RAB6-positive transport carriers from Golgi/TGN membranes. Here we show that KIF20A is also involved in the fission process and serves to anchor RAB6 on Golgi/TGN membranes near microtubule nucleating sites. We provide evidence that the fission events occur at a limited number of hotspots sites. Our results suggest that coupling between actin and microtubule cytoskeletons driven by Myosin II and KIF20A ensures the spatial coordination between RAB6-positive vesicles fission from Golgi/TGN membranes and their exit along microtubules. Actin and microtubules play important roles in Golgi structure and function but how they are connected is poorly understood. Here the authors show that KIF20A is involved in the fission process and, in association with Myosin II, serves to anchor RAB6 on Golgi/TGN membranes near microtubules nucleating sites.
Collapse
Affiliation(s)
- Stéphanie Miserey-Lenkei
- Institut Curie, PSL Research University, CNRS, UMR 144, Molecular Mechanisms of Intracellular Transport, F-75005, Paris, France.
| | - Hugo Bousquet
- Institut Curie, PSL Research University, CNRS, UMR 144, Molecular Mechanisms of Intracellular Transport, F-75005, Paris, France
| | - Olena Pylypenko
- Institut Curie, PSL Research University, CNRS, UMR 144, Structural Motility, F-75005, Paris, France
| | - Sabine Bardin
- Institut Curie, PSL Research University, CNRS, UMR 144, Molecular Mechanisms of Intracellular Transport, F-75005, Paris, France
| | - Ariane Dimitrov
- Institut Curie, PSL Research University, CNRS, UMR 144, Molecular Mechanisms of Intracellular Transport, F-75005, Paris, France
| | - Gaëlle Bressanelli
- Institut Curie, PSL Research University, CNRS, UMR 144, Structural Motility, F-75005, Paris, France
| | - Raja Bonifay
- Institut Curie, PSL Research University, CNRS, UMR 144, Structural Motility, F-75005, Paris, France
| | - Vincent Fraisier
- Institut Curie, PSL Research University, CNRS, UMR 144, Cell and Tissue Imaging Facility (PICT-IBiSA), F-75005, Paris, France
| | | | | | - Anne Houdusse
- Institut Curie, PSL Research University, CNRS, UMR 144, Structural Motility, F-75005, Paris, France
| | - Arnaud Echard
- Institut Pasteur, CNRS UMR3691, Membrane Traffic and Cell Division, F-75015, Paris, France
| | - Bruno Goud
- Institut Curie, PSL Research University, CNRS, UMR 144, Molecular Mechanisms of Intracellular Transport, F-75005, Paris, France.
| |
Collapse
|
34
|
Chen W, Zhao Y, Li XC, Kubiak JZ, Ghobrial RM, Kloc M. Rho-specific Guanine nucleotide exchange factors (Rho-GEFs) inhibition affects macrophage phenotype and disrupts Golgi complex. Int J Biochem Cell Biol 2017; 93:12-24. [PMID: 29061365 DOI: 10.1016/j.biocel.2017.10.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/04/2017] [Accepted: 10/17/2017] [Indexed: 01/09/2023]
Abstract
Macrophages play crucial role in tissue homeostasis and the innate and adaptive immune response. Depending on the state of activation macrophages acquire distinct phenotypes that depend on actin, which is regulated by small GTPase RhoA. The naive M0 macrophages are slightly elongated, pro-inflammatory M1 are round and M2 anti-inflammatory macrophages are elongated. We showed previously that interference with RhoA pathway (RhoA deletion or RhoA/ROCK kinase inhibition) disrupted actin, produced extremely elongated (hummingbird) macrophage phenotype and inhibited macrophage movement toward transplanted hearts. The RhoA function depends on the family of guanine-nucleotide exchange factors (GEFs), which catalyze the exchange of GDP for GTP and activate RhoA that reorganizes actin cytoskeleton. Using actin staining, immunostaining, Western blotting, flow cytometry and transmission electron microscopy we studied how a direct inhibition of Rho-GEFs with Rhosin (Rho GEF-binding domain blocker) and Y16 (Rho GEF DH-PH domain blocker) affects M0, M1 and M2 macrophage phenotypes. We also studied how Rho-GEFs inhibition and RhoA deletion affects organization of Golgi complex that is crucial for normal macrophage functions such as phagocytosis, antigen presentation and receptor recycling. We found that GEFs inhibition differently affected M0, M1 and M2 macrophages phenotype and that GEFs inhibition and RhoA deletion both caused changes in the ultrastructure of the Golgi complex. These results suggest that actin/RhoA- dependent shaping of macrophage phenotype has different requirements for activity of RhoA/GEFs pathway in M0, M1 and M2 macrophages, and that RhoA and Rho-GEFs functions are necessary for the maintenance of actin-dependent organization of Golgi complex.
Collapse
Affiliation(s)
- Wei Chen
- The Houston Methodist Research Institute, Houston, TX, USA; Department of Nephrology, Second Xiangya Hospital, Central South University, Changsha 410011, China
| | - Yue Zhao
- The Houston Methodist Research Institute, Houston, TX, USA
| | - Xian C Li
- The Houston Methodist Research Institute, Houston, TX, USA; The Houston Methodist Hospital, Department of Surgery, Houston, TX, USA
| | - Jacek Z Kubiak
- CNRS UMR 6290, Institute of Genetics and Development of Rennes, Cell Cycle Group, IFR 140 GFAS, France; University of Rennes 1, Faculty of Medicine, Rennes, France; Department of Regenerative Medicine, Military Institute of Hygiene and Epidemiology (WIHE), Warsaw, Poland
| | - Rafik M Ghobrial
- The Houston Methodist Research Institute, Houston, TX, USA; The Houston Methodist Hospital, Department of Surgery, Houston, TX, USA
| | - Malgorzata Kloc
- The Houston Methodist Research Institute, Houston, TX, USA; The Houston Methodist Hospital, Department of Surgery, Houston, TX, USA; The University of Texas, MD Anderson Cancer Center, Department of Genetics, Houston, TX, USA.
| |
Collapse
|
35
|
Kage F, Steffen A, Ellinger A, Ranftler C, Gehre C, Brakebusch C, Pavelka M, Stradal T, Rottner K. FMNL2 and -3 regulate Golgi architecture and anterograde transport downstream of Cdc42. Sci Rep 2017; 7:9791. [PMID: 28852060 PMCID: PMC5575334 DOI: 10.1038/s41598-017-09952-1] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 07/26/2017] [Indexed: 01/08/2023] Open
Abstract
The Rho-family small GTPase Cdc42 localizes at plasma membrane and Golgi complex and aside from protrusion and migration operates in vesicle trafficking, endo- and exocytosis as well as establishment and/or maintenance of cell polarity. The formin family members FMNL2 and -3 are actin assembly factors established to regulate cell edge protrusion during migration and invasion. Here we report these formins to additionally accumulate and function at the Golgi apparatus. As opposed to lamellipodia, Golgi targeting of these proteins required both their N-terminal myristoylation and the interaction with Cdc42. Moreover, Golgi association of FMNL2 or -3 induced a phalloidin-detectable actin meshwork around the Golgi. Importantly, functional interference with FMNL2/3 formins by RNAi or CRISPR/Cas9-mediated gene deletion invariably induced Golgi fragmentation in different cell lines. Furthermore, absence of these proteins led to enlargement of endosomes as well as defective maturation and/or sorting into late endosomes and lysosomes. In line with Cdc42 - recently established to regulate anterograde transport through the Golgi by cargo sorting and carrier formation - FMNL2/3 depletion also affected anterograde trafficking of VSV-G from the Golgi to the plasma membrane. Our data thus link FMNL2/3 formins to actin assembly-dependent functions of Cdc42 in anterograde transport through the Golgi apparatus.
Collapse
Affiliation(s)
- Frieda Kage
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Anika Steffen
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Adolf Ellinger
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Carmen Ranftler
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Christian Gehre
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany.,Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Cord Brakebusch
- Biomedical Institute, BRIC, University of Copenhagen, DK-2200, Copenhagen, Denmark
| | - Margit Pavelka
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Theresia Stradal
- Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany
| | - Klemens Rottner
- Division of Molecular Cell Biology, Zoological Institute, Technische Universität Braunschweig, Spielmannstrasse 7, 38106, Braunschweig, Germany. .,Department of Cell Biology, Helmholtz Centre for Infection Research, Inhoffenstrasse 7, 38124, Braunschweig, Germany.
| |
Collapse
|
36
|
De Conto F, Fazzi A, Razin SV, Arcangeletti MC, Medici MC, Belletti S, Chezzi C, Calderaro A. Mammalian Diaphanous-related formin-1 restricts early phases of influenza A/NWS/33 virus (H1N1) infection in LLC-MK2 cells by affecting cytoskeleton dynamics. Mol Cell Biochem 2017; 437:185-201. [PMID: 28744815 DOI: 10.1007/s11010-017-3107-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Accepted: 07/01/2017] [Indexed: 12/15/2022]
Abstract
Viruses depend on cellular machinery to efficiently replicate. The host cytoskeleton is one of the first cellular systems hijacked by viruses in order to ensure their intracellular transport and promote the development of infection. Our previous results demonstrated that stable microfilaments and microtubules interfered with human influenza A/NWS/33 virus (H1N1) infection in semi-permissive LLC-MK2 cells. Although formins play a key role in cytoskeletal remodelling, few studies addressed a possible role of these proteins in development of viral infection. Here, we have demonstrated that mammalian Diaphanous-related formin-1 (mDia1) is involved in the control of cytoskeleton dynamics during human influenza A virus infection. First, by employing cytoskeleton-perturbing drugs, we evidenced a cross-talk occurring between microtubules and microfilaments that also has implications on the intracellular localization of mDia1. In influenza A/NWS/33 virus-infected LLC-MK2 cells, mDia1 showed a highly dynamic intracellular localization and partially co-localized with actin and tubulin. A depletion of mDia1 by RNA-mediated RNA interference was found to improve the outcome of influenza A/NWS/33 virus infection and to increase the dynamics of microfilament and microtubule networks in LLC-MK2 cells. Consistent with these findings, observations made in epithelial respiratory cells from paediatric patients with acute respiratory disease assessed that the expression of mDia1 is stimulated by influenza A virus but not by respiratory syncytial virus. Taken together, the obtained results suggest that mDia1 restricts the initiation of influenza A/NWS/33 virus infection in LLC-MK2 cells by counteracting cytoskeletal dynamics.
Collapse
Affiliation(s)
- Flora De Conto
- Department of Medicine and Surgery, University of Parma, Parma, Italy.
| | - Alessandra Fazzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Sergey V Razin
- Institute of Gene Biology, Russian Academy of Sciences and Lomonosow Moscow State University, Moscow, Russia
| | | | | | - Silvana Belletti
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Carlo Chezzi
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| | - Adriana Calderaro
- Department of Medicine and Surgery, University of Parma, Parma, Italy
| |
Collapse
|
37
|
Shibata S, Teshima Y, Niimi K, Inagaki S. Involvement of ARHGEF10, GEF for RhoA, in Rab6/Rab8-mediating membrane traffic. Small GTPases 2017; 10:169-177. [PMID: 28448737 DOI: 10.1080/21541248.2017.1302550] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Small GTPases play crucial roles in the maintenance of a homeostatic environment and appropriate movements of the cell. In these processes, the direct or indirect interaction between distinct small GTPases could be required for regulating mutual signaling pathways. In our recent study, ARHGEF10, known as a guanine nucleotide exchange factor (GEF) for RhoA, was indicated to interact with Rab6A and Rab8A, which are known to function in the exocytotic pathway, and colocalized with these Rabs at exocytotic vesicles. Moreover, it was suggested that ARHGEF10 is involved in the regulation of Rab6A and Rab8A localization and invasion of breast carcinoma cells, in which Rab8 also acts via regulation of membrane trafficking. These results may reveal the existence of a novel small GTPase cascade which connects the signaling of these Rabs with RhoA during membrane trafficking. In this mini-review, we consider the possible functions of ARHGEF10 and RhoA in the Rab6- and Rab8-mediated membrane trafficking pathway.
Collapse
Affiliation(s)
- Satoshi Shibata
- a Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine , Osaka University , Osaka , Japan
| | - Yui Teshima
- a Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine , Osaka University , Osaka , Japan
| | - Kenta Niimi
- a Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine , Osaka University , Osaka , Japan
| | - Shinobu Inagaki
- a Group of Neurobiology, Division of Health Sciences, Graduate School of Medicine , Osaka University , Osaka , Japan
| |
Collapse
|
38
|
Rho GTPases operating at the Golgi complex: Implications for membrane traffic and cancer biology. Tissue Cell 2016; 49:163-169. [PMID: 27720426 DOI: 10.1016/j.tice.2016.09.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 09/09/2016] [Accepted: 09/26/2016] [Indexed: 11/20/2022]
Abstract
The Golgi complex is the central unit of the secretory pathway, modifying, processing and sorting proteins and lipids to their correct cellular localisation. Changes to proteins at the Golgi complex can have deleterious effects on the function of this organelle, impeding trafficking routes through it, potentially resulting in disease. It is emerging that several Rho GTPase proteins, namely Cdc42, RhoBTB3, RhoA and RhoD are at least in part localised to the Golgi complex, and a number of studies have shown that dysregulation of their levels or activity can be associated with cellular changes which ultimately drive cancer progression. In this mini-review we highlight some of the recent work that explores links between form and function of the Golgi complex, Rho GTPases and cancer.
Collapse
|
39
|
Fokin AI, Klementeva TS, Nadezhdina ES, Burakov AV. SLK/LOSK kinase regulates cell motility independently of microtubule organization and Golgi polarization. Cytoskeleton (Hoboken) 2016; 73:83-92. [PMID: 26818812 DOI: 10.1002/cm.21276] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Revised: 10/16/2015] [Accepted: 01/20/2016] [Indexed: 12/31/2022]
Abstract
Cell motility is an essential complex process that requires actin and microtubule cytoskeleton reorganization and polarization. Such extensive rearrangement is closely related to cell polarization as a whole. The serine/threonine kinase SLK/LOSK is a potential regulator of cell motility, as it phosphorylates a series of cytoskeleton-bound proteins that collectively participate in the remodeling of migratory cell architecture. In this work, we report that SLK/LOSK is an indispensable regulator of cell locomotion that primarily acts through the small GTPase RhoA and the dynactin subunit p150(Glued). Both RhoA and dynactin affect cytoskeleton organization, polarization, and general cell locomotory activity to various extents. However, it seems that these events are independent of each other. Thus, SLK/LOSK kinase effectively functions as a switch that links all of the processes underlying cell motility to provide robust directional movement.
Collapse
Affiliation(s)
- Artem I Fokin
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskye Gory, Moscow, 119992, Russia
| | - Tatiana S Klementeva
- Institute of Protein Research of Russian Academy of Sciences, Moscow Region, Pushchino, Institutskaya Str, 4, 142290, Russia
| | - Elena S Nadezhdina
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskye Gory, Moscow, 119992, Russia.,Institute of Protein Research of Russian Academy of Sciences, Moscow Region, Pushchino, Institutskaya Str, 4, 142290, Russia
| | - Anton V Burakov
- A.N.Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskye Gory, Moscow, 119992, Russia
| |
Collapse
|
40
|
Copeland SJ, Thurston SF, Copeland JW. Actin- and microtubule-dependent regulation of Golgi morphology by FHDC1. Mol Biol Cell 2015; 27:260-76. [PMID: 26564798 PMCID: PMC4713130 DOI: 10.1091/mbc.e15-02-0070] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 11/04/2015] [Indexed: 01/08/2023] Open
Abstract
The coordinated action of the actin and microtubule cytoskeletal networks is required for Golgi ribbon assembly. The novel formin FHDC1 accumulates on the Golgi-derived microtubule network, where it acts to regulate Golgi ribbon assembly in an actin- and microtubule-dependent manner. The Golgi apparatus is the central hub of intracellular trafficking and consists of tethered stacks of cis, medial, and trans cisternae. In mammalian cells, these cisternae are stitched together as a perinuclear Golgi ribbon, which is required for the establishment of cell polarity and normal subcellular organization. We previously identified FHDC1 (also known as INF1) as a unique microtubule-binding member of the formin family of cytoskeletal-remodeling proteins. We show here that endogenous FHDC1 regulates Golgi ribbon formation and has an apparent preferential association with the Golgi-derived microtubule network. Knockdown of FHDC1 expression results in defective Golgi assembly and suggests a role for FHDC1 in maintenance of the Golgi-derived microtubule network. Similarly, overexpression of FHDC1 induces dispersion of the Golgi ribbon into functional ministacks. This effect is independent of centrosome-derived microtubules and instead likely requires the interaction between the FHDC1 microtubule-binding domain and the Golgi-derived microtubule network. These effects also depend on the interaction between the FHDC1 FH2 domain and the actin cytoskeleton. Thus our results suggest that the coordination of actin and microtubule dynamics by FHDC1 is required for normal Golgi ribbon formation.
Collapse
Affiliation(s)
- Sarah J Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - Susan F Thurston
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| | - John W Copeland
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON K1H 8M5, Canada
| |
Collapse
|
41
|
Tang D, Zhang X, Huang S, Yuan H, Li J, Wang Y. Mena-GRASP65 interaction couples actin polymerization to Golgi ribbon linking. Mol Biol Cell 2015; 27:137-52. [PMID: 26538023 PMCID: PMC4694753 DOI: 10.1091/mbc.e15-09-0650] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 10/27/2015] [Indexed: 01/08/2023] Open
Abstract
GRASP65 plays a role in Golgi ribbon formation. Because the gaps between Golgi stacks are heterogeneous and large, it is possible that other proteins may help GRASP65 in ribbon linking. Mena is a novel GRASP65-binding protein that promotes actin elongation and enhances GRASP65 oligomerization to link Golgi stacks into a ribbon. In mammalian cells, the Golgi reassembly stacking protein 65 (GRASP65) has been implicated in both Golgi stacking and ribbon linking by forming trans-oligomers through the N-terminal GRASP domain. Because the GRASP domain is globular and relatively small, but the gaps between stacks are large and heterogeneous, it remains puzzling how GRASP65 physically links Golgi stacks into a ribbon. To explore the possibility that other proteins may help GRASP65 in ribbon linking, we used biochemical methods and identified the actin elongation factor Mena as a novel GRASP65-binding protein. Mena is recruited onto the Golgi membranes through interaction with GRASP65. Depleting Mena or disrupting actin polymerization resulted in Golgi fragmentation. In cells, Mena and actin were required for Golgi ribbon formation after nocodazole washout; in vitro, Mena and microfilaments enhanced GRASP65 oligomerization and Golgi membrane fusion. Thus Mena interacts with GRASP65 to promote local actin polymerization, which facilitates Golgi ribbon linking.
Collapse
Affiliation(s)
- Danming Tang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048
| | - Xiaoyan Zhang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048
| | - Shijiao Huang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048
| | - Hebao Yuan
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048
| | - Jie Li
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048
| | - Yanzhuang Wang
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI 48109-1048 Department of Neurology, University of Michigan School of Medicine, Ann Arbor, MI 48109-1048
| |
Collapse
|
42
|
RhoA Ambivalently Controls Prominent Myofibroblast Characteritics by Involving Distinct Signaling Routes. PLoS One 2015; 10:e0137519. [PMID: 26448568 PMCID: PMC4598021 DOI: 10.1371/journal.pone.0137519] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2015] [Accepted: 08/18/2015] [Indexed: 11/19/2022] Open
Abstract
Introduction RhoA has been shown to be beneficial in cardiac disease models when overexpressed in cardiomyocytes, whereas its role in cardiac fibroblasts (CF) is still poorly understood. During cardiac remodeling CF undergo a transition towards a myofibroblast phenotype thereby showing an increased proliferation and migration rate. Both processes involve the remodeling of the cytoskeleton. Since RhoA is known to be a major regulator of the cytoskeleton, we analyzed its role in CF and its effect on myofibroblast characteristics in 2 D and 3D models. Results Downregulation of RhoA was shown to strongly affect the actin cytoskeleton. It decreased the myofibroblast marker α-sm-actin, but increased certain fibrosis-associated factors like TGF-β and collagens. Also, the detailed analysis of CTGF expression demonstrated that the outcome of RhoA signaling strongly depends on the involved stimulus. Furthermore, we show that proliferation of myofibroblasts rely on RhoA and tubulin acetylation. In assays accessing three different types of migration, we demonstrate that RhoA/ROCK/Dia1 are important for 2D migration and the repression of RhoA and Dia1 signaling accelerates 3D migration. Finally, we show that a downregulation of RhoA in CF impacts the viscoelastic and contractile properties of engineered tissues. Conclusion RhoA positively and negatively influences myofibroblast characteristics by differential signaling cascades and depending on environmental conditions. These include gene expression, migration and proliferation. Reduction of RhoA leads to an increased viscoelasticity and a decrease in contractile force in engineered cardiac tissue.
Collapse
|
43
|
van Unen J, Reinhard NR, Yin T, Wu YI, Postma M, Gadella TWJ, Goedhart J. Plasma membrane restricted RhoGEF activity is sufficient for RhoA-mediated actin polymerization. Sci Rep 2015; 5:14693. [PMID: 26435194 PMCID: PMC4592971 DOI: 10.1038/srep14693] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2015] [Accepted: 09/04/2015] [Indexed: 01/08/2023] Open
Abstract
The small GTPase RhoA is involved in cell morphology and migration. RhoA activity is tightly regulated in time and space and depends on guanine exchange factors (GEFs). However, the kinetics and subcellular localization of GEF activity towards RhoA are poorly defined. To study the mechanism underlying the spatiotemporal control of RhoA activity by GEFs, we performed single cell imaging with an improved FRET sensor reporting on the nucleotide loading state of RhoA. By employing the FRET sensor we show that a plasma membrane located RhoGEF, p63RhoGEF, can rapidly activate RhoA through endogenous GPCRs and that localized RhoA activity at the cell periphery correlates with actin polymerization. Moreover, synthetic recruitment of the catalytic domain derived from p63RhoGEF to the plasma membrane, but not to the Golgi apparatus, is sufficient to activate RhoA. The synthetic system enables local activation of endogenous RhoA and effectively induces actin polymerization and changes in cellular morphology. Together, our data demonstrate that GEF activity at the plasma membrane is sufficient for actin polymerization via local RhoA signaling.
Collapse
Affiliation(s)
- Jakobus van Unen
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, P.O. Box 94215, NL-1090 GE Amsterdam, The Netherlands
| | - Nathalie R Reinhard
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, P.O. Box 94215, NL-1090 GE Amsterdam, The Netherlands
| | - Taofei Yin
- Center for Cell Analysis and Modeling, University of Connecticut Health Center, 400 Farmington Avenue, Farmington, CT 06032-6406
| | - Yi I Wu
- Center for Cell Analysis and Modeling, University of Connecticut Health Center, 400 Farmington Avenue, Farmington, CT 06032-6406
| | - Marten Postma
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, P.O. Box 94215, NL-1090 GE Amsterdam, The Netherlands
| | - Theodorus W J Gadella
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, P.O. Box 94215, NL-1090 GE Amsterdam, The Netherlands
| | - Joachim Goedhart
- Swammerdam Institute for Life Sciences, Section of Molecular Cytology, van Leeuwenhoek Centre for Advanced Microscopy, University of Amsterdam, P.O. Box 94215, NL-1090 GE Amsterdam, The Netherlands
| |
Collapse
|
44
|
Gurel PS, Hatch AL, Higgs HN. Connecting the cytoskeleton to the endoplasmic reticulum and Golgi. Curr Biol 2015; 24:R660-R672. [PMID: 25050967 DOI: 10.1016/j.cub.2014.05.033] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
A tendency in cell biology is to divide and conquer. For example, decades of painstaking work have led to an understanding of endoplasmic reticulum (ER) and Golgi structure, dynamics, and transport. In parallel, cytoskeletal researchers have revealed a fantastic diversity of structure and cellular function in both actin and microtubules. Increasingly, these areas overlap, necessitating an understanding of both organelle and cytoskeletal biology. This review addresses connections between the actin/microtubule cytoskeletons and organelles in animal cells, focusing on three key areas: ER structure and function; ER-to-Golgi transport; and Golgi structure and function. Making these connections has been challenging for several reasons: the small sizes and dynamic characteristics of some components; the fact that organelle-specific cytoskeletal elements can easily be obscured by more abundant cytoskeletal structures; and the difficulties in imaging membranes and cytoskeleton simultaneously, especially at the ultrastructural level. One major concept is that the cytoskeleton is frequently used to generate force for membrane movement, with two potential consequences: translocation of the organelle, or deformation of the organelle membrane. While initially discussing issues common to metazoan cells in general, we subsequently highlight specific features of neurons, since these highly polarized cells present unique challenges for organellar distribution and dynamics.
Collapse
Affiliation(s)
- Pinar S Gurel
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Anna L Hatch
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA
| | - Henry N Higgs
- Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover NH 03755, USA.
| |
Collapse
|
45
|
Arden JD, Lavik KI, Rubinic KA, Chiaia N, Khuder SA, Howard MJ, Nestor-Kalinoski AL, Alberts AS, Eisenmann KM. Small-molecule agonists of mammalian Diaphanous-related (mDia) formins reveal an effective glioblastoma anti-invasion strategy. Mol Biol Cell 2015; 26:3704-18. [PMID: 26354425 PMCID: PMC4626057 DOI: 10.1091/mbc.e14-11-1502] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Accepted: 09/04/2015] [Indexed: 12/26/2022] Open
Abstract
Formin agonists impede the most dangerous aspect of glioblastoma—tumor spread into surrounding healthy tissue. Formin activation impairs a novel aspect of the transformed cell and informs the development of antitumor strategies for a cancer needing a more effective therapy. The extensive invasive capacity of glioblastoma (GBM) makes it resistant to surgery, radiotherapy, and chemotherapy and thus makes it lethal. In vivo, GBM invasion is mediated by Rho GTPases through unidentified downstream effectors. Mammalian Diaphanous (mDia) family formins are Rho-directed effectors that regulate the F-actin cytoskeleton to support tumor cell motility. Historically, anti-invasion strategies focused upon mDia inhibition, whereas activation remained unexplored. The recent development of small molecules directly inhibiting or activating mDia-driven F-actin assembly that supports motility allows for exploration of their role in GBM. We used the formin inhibitor SMIFH2 and mDia agonists IMM-01/-02 and mDia2-DAD peptides, which disrupt autoinhibition, to examine the roles of mDia inactivation versus activation in GBM cell migration and invasion in vitro and in an ex vivo brain slice invasion model. Inhibiting mDia suppressed directional migration and spheroid invasion while preserving intrinsic random migration. mDia agonism abrogated both random intrinsic and directional migration and halted U87 spheroid invasion in ex vivo brain slices. Thus mDia agonism is a superior GBM anti-invasion strategy. We conclude that formin agonism impedes the most dangerous GBM component—tumor spread into surrounding healthy tissue. Formin activation impairs novel aspects of transformed cells and informs the development of anti-GBM invasion strategies.
Collapse
Affiliation(s)
- Jessica D Arden
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Kari I Lavik
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Kaitlin A Rubinic
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Nicolas Chiaia
- Department of Neurosciences, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Sadik A Khuder
- Departments of Medicine and Public Health and Homeland Security, University of Toledo Health Science Campus, Toledo, OH 43614
| | - Marthe J Howard
- Department of Neurosciences, University of Toledo Health Science Campus, Toledo, OH 43614
| | | | - Arthur S Alberts
- Laboratory of Cell Structure and Signal Integration, Van Andel Research Institute, Grand Rapids, MI 49503
| | - Kathryn M Eisenmann
- Department of Biochemistry and Cancer Biology, University of Toledo Health Science Campus, Toledo, OH 43614 )
| |
Collapse
|
46
|
Aittaleb M, Chen PJ, Akaaboune M. Failure of lysosome clustering and positioning in the juxtanuclear region in cells deficient in rapsyn. J Cell Sci 2015; 128:3744-56. [PMID: 26330529 DOI: 10.1242/jcs.172536] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/23/2015] [Indexed: 01/06/2023] Open
Abstract
Rapsyn, a scaffold protein, is required for the clustering of acetylcholine receptors (AChRs) at contacts between motor neurons and differentiating muscle cells. Rapsyn is also expressed in cells that do not express AChRs. However, its function in these cells remains unknown. Here, we show that rapsyn plays an AChR-independent role in organizing the distribution and mobility of lysosomes. In cells devoid of AChRs, rapsyn selectively induces the clustering of lysosomes at high density in the juxtanuclear region without affecting the distribution of other intracellular organelles. However, when the same cells overexpress AChRs, rapsyn is recruited away from lysosomes to colocalize with AChR clusters on the cell surface. In rapsyn-deficient (Rapsn(-/-)) myoblasts or cells overexpressing rapsyn mutants, lysosomes are scattered within the cell and highly dynamic. The increased mobility of lysosomes in Rapsn(-/-) cells is associated with a significant increase in lysosomal exocytosis, as evidenced by increased release of lysosomal enzymes and plasma membrane damage when cells were challenged with the bacterial pore-forming toxin streptolysin-O. These findings uncover a new link between rapsyn, lysosome positioning, exocytosis and plasma membrane integrity.
Collapse
Affiliation(s)
- Mohamed Aittaleb
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Po-Ju Chen
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Mohammed Akaaboune
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48109, USA Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
47
|
Zaidel-Bar R, Zhenhuan G, Luxenburg C. The contractome – a systems view of actomyosin contractility in non-muscle cells. J Cell Sci 2015; 128:2209-17. [DOI: 10.1242/jcs.170068] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 04/27/2015] [Indexed: 12/21/2022] Open
Abstract
ABSTRACT
Actomyosin contractility is a highly regulated process that affects many fundamental biological processes in each and every cell in our body. In this Cell Science at a Glance article and the accompanying poster, we mined the literature and databases to map the contractome of non-muscle cells. Actomyosin contractility is involved in at least 49 distinct cellular functions that range from providing cell architecture to signal transduction and nuclear activity. Containing over 100 scaffolding and regulatory proteins, the contractome forms a highly complex network with more than 230 direct interactions between its components, 86 of them involving phosphorylation. Mapping these interactions, we identify the key regulatory pathways involved in the assembly of actomyosin structures and in activating myosin to produce contractile forces within non-muscle cells at the exact time and place necessary for cellular function.
Collapse
Affiliation(s)
- Ronen Zaidel-Bar
- Mechanobiology Institute, National University of Singapore, T-lab building #05-01, 5A Engineering Drive 1, 117411, Singapore
| | - Guo Zhenhuan
- Mechanobiology Institute, National University of Singapore, T-lab building #05-01, 5A Engineering Drive 1, 117411, Singapore
| | - Chen Luxenburg
- Department of Cell and Developmental Biology, Sackler Faculty of Medicine, Tel Aviv University, P.O. Box 39040, Tel Aviv 69978, Israel
| |
Collapse
|
48
|
Goins LM, Mullins RD. A novel tropomyosin isoform functions at the mitotic spindle and Golgi in Drosophila. Mol Biol Cell 2015; 26:2491-504. [PMID: 25971803 PMCID: PMC4571303 DOI: 10.1091/mbc.e14-12-1619] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Accepted: 05/05/2015] [Indexed: 12/28/2022] Open
Abstract
Most eukaryotic cells express multiple isoforms of the actin-binding protein tropomyosin that help construct a variety of cytoskeletal networks. Only one nonmuscle tropomyosin (Tm1A) has previously been described in Drosophila, but developmental defects caused by insertion of P-elements near tropomyosin genes imply the existence of additional, nonmuscle isoforms. Using biochemical and molecular genetic approaches, we identified three tropomyosins expressed in Drosophila S2 cells: Tm1A, Tm1J, and Tm2A. The Tm1A isoform localizes to the cell cortex, lamellar actin networks, and the cleavage furrow of dividing cells--always together with myosin-II. Isoforms Tm1J and Tm2A colocalize around the Golgi apparatus with the formin-family protein Diaphanous, and loss of either isoform perturbs cell cycle progression. During mitosis, Tm1J localizes to the mitotic spindle, where it promotes chromosome segregation. Using chimeras, we identified the determinants of tropomyosin localization near the C-terminus. This work 1) identifies and characterizes previously unknown nonmuscle tropomyosins in Drosophila, 2) reveals a function for tropomyosin in the mitotic spindle, and 3) uncovers sequence elements that specify isoform-specific localizations and functions of tropomyosin.
Collapse
Affiliation(s)
- Lauren M Goins
- Department of Cellular and Molecular Pharmacology, School of Medicine, University of California, San Francisco, San Francisco, CA 94158
| | - R Dyche Mullins
- Department of Cellular and Molecular Pharmacology, School of Medicine, University of California, San Francisco, San Francisco, CA 94158
| |
Collapse
|
49
|
Mechanical stimulation induces formin-dependent assembly of a perinuclear actin rim. Proc Natl Acad Sci U S A 2015; 112:E2595-601. [PMID: 25941386 DOI: 10.1073/pnas.1504837112] [Citation(s) in RCA: 94] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Cells constantly sense and respond to mechanical signals by reorganizing their actin cytoskeleton. Although a number of studies have explored the effects of mechanical stimuli on actin dynamics, the immediate response of actin after force application has not been studied. We designed a method to monitor the spatiotemporal reorganization of actin after cell stimulation by local force application. We found that force could induce transient actin accumulation in the perinuclear region within ∼ 2 min. This actin reorganization was triggered by an intracellular Ca(2+) burst induced by force application. Treatment with the calcium ionophore A23187 recapitulated the force-induced perinuclear actin remodeling. Blocking of actin polymerization abolished this process. Overexpression of Klarsicht, ANC-1, Syne Homology (KASH) domain to displace nesprins from the nuclear envelope did not abolish Ca(2+)-dependent perinuclear actin assembly. However, the endoplasmic reticulum- and nuclear membrane-associated inverted formin-2 (INF2), a potent actin polymerization activator (mutations of which are associated with several genetic diseases), was found to be important for perinuclear actin assembly. The perinuclear actin rim structure colocalized with INF2 on stimulation, and INF2 depletion resulted in attenuation of the rim formation. Our study suggests that cells can respond rapidly to external force by remodeling perinuclear actin in a unique Ca(2+)- and INF2-dependent manner.
Collapse
|
50
|
SMIFH2 has effects on Formins and p53 that perturb the cell cytoskeleton. Sci Rep 2015; 5:9802. [PMID: 25925024 PMCID: PMC5386218 DOI: 10.1038/srep09802] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 03/19/2015] [Indexed: 01/08/2023] Open
Abstract
Formin proteins are key regulators of the cytoskeleton involved in developmental and homeostatic programs, and human disease. For these reasons, small molecules interfering with Formins' activity have gained increasing attention. Among them, small molecule inhibitor of Formin Homology 2 domains (SMIFH2) is often used as a pharmacological Formin blocker. Although SMIFH2 inhibits actin polymerization by Formins and affects the actin cytoskeleton, its cellular mechanism of action and target specificity remain unclear. Here we show that SMIFH2 induces remodelling of actin filaments, microtubules and the Golgi complex as a result of its effects on Formins and p53. We found that SMIFH2 triggers alternated depolymerization-repolymerization cycles of actin and tubulin, increases cell migration, causes scattering of the Golgi complex, and also cytotoxicity at high dose. Moreover, SMIFH2 reduces expression and activity of p53 through a post-transcriptional, proteasome-independent mechanism that influences remodelling of the cytoskeleton. As the action of SMIFH2 may go beyond Formin inhibition, only short-term and low-dose SMIFH2 treatments minimize confounding effects induced by loss of p53 and cytotoxicity.
Collapse
|