1
|
Ocket E, Matthaeus C. Insights in caveolae protein structure arrangements and their local lipid environment. Biol Chem 2024; 0:hsz-2024-0046. [PMID: 38970809 DOI: 10.1515/hsz-2024-0046] [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: 03/19/2024] [Accepted: 06/19/2024] [Indexed: 07/08/2024]
Abstract
Caveolae are 50-80 nm sized plasma membrane invaginations found in adipocytes, endothelial cells or fibroblasts. They are involved in endocytosis, lipid uptake and the regulation of the cellular lipid metabolism as well as sensing and adapting to changes in plasma membrane tension. Caveolae are characterized by their unique lipid composition and their specific protein coat consisting of caveolin and cavin proteins. Recently, detailed structural information was obtained for the major caveolae protein caveolin1 showing the formation of a disc-like 11-mer protein complex. Furthermore, the importance of the cavin disordered regions in the generation of cavin trimers and caveolae at the plasma membrane were revealed. Thus, finally, structural insights about the assembly of the caveolar coat can be elucidated. Here, we review recent developments in caveolae structural biology with regard to caveolae coat formation and caveolae curvature generation. Secondly, we discuss the importance of specific lipid species necessary for caveolae curvature and formation. In the last years, it was shown that specifically sphingolipids, cholesterol and fatty acids can accumulate in caveolae invaginations and may drive caveolae endocytosis. Throughout, we summarize recent studies in the field and highlight future research directions.
Collapse
Affiliation(s)
- Esther Ocket
- Institute of Nutritional Science, Cellular Physiology of Nutrition, University of Potsdam, Karl-Liebknecht-Str. 24/25, Building 29, Room 0.08, D-14476 Potsdam, Germany
| | - Claudia Matthaeus
- Institute of Nutritional Science, Cellular Physiology of Nutrition, University of Potsdam, Karl-Liebknecht-Str. 24/25, Building 29, Room 0.08, D-14476 Potsdam, Germany
| |
Collapse
|
2
|
Lin J, Guo W, Luo Q, Zhang Q, Wan T, Jiang C, Ye Y, Lin H, Fan G. Senolytics prevent caveolar Ca V 3.2-RyR axis malfunction in old vascular smooth muscle. Aging Cell 2023; 22:e14002. [PMID: 37837625 PMCID: PMC10652315 DOI: 10.1111/acel.14002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 07/18/2023] [Accepted: 08/07/2023] [Indexed: 10/16/2023] Open
Abstract
Aging is a major risk factor for cardiovascular diseases. Our previous studies demonstrate that aging impairs the caveolar T-type CaV 3.2-RyR axis for extracellular Ca2+ influx to trigger Ca2+ sparks in vascular smooth muscle cells (VSMCs). We hypothesize that the administration of senolytics, which can selectively clear senescent cells, could preserve the caveolar CaV 3.2-RyR axis in aging VSMCs. In this study, 10-month-old mice were administered the senolytics cocktail consisting of dasatinib (5 mg/kg) and quercetin (50 mg/kg) or vehicle bi-weekly for 4 months. Using VSMCs from mouse mesenteric arteries, we found that Ca2+ sparks were diminished after caveolae disruption by methyl-β-cyclodextrin (10 mM) in cells from D + Q treated but not vehicle-treated 14-month-old mice. D + Q treatment promoted the expression of CaV 3.2 in 14-month-old mesenteric arteries. Structural analysis using electron tomography and immunofluorescence staining revealed the remodeling of caveolae and co-localization of CaV 3.2-Cav-1 in D + Q treatment aged mesenteric arteries. In keeping with theoretical observations, Cav 3.2 channel inhibition by Ni2+ (50 μM) suppressed Ca2+ in VSMCs from the D + Q group, with no effect observed in vehicle-treated arteries. Our study provides evidence that age-related caveolar CaV 3.2-RyR axis malfunction can be alleviated by pharmaceutical intervention targeting cellular senescence. Our findings support the potential of senolytics for ameliorating age-associated cardiovascular disease.
Collapse
Affiliation(s)
- Jie Lin
- Cardiology DepartmentThe first Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Weiming Guo
- Sports Medicine CenterHuazhong University of Science and Technology Union Shenzhen Hospital, the 6th affiliated Hospital of Shenzhen University Medical SchoolShenzhenChina
| | - Qingtian Luo
- Department of GastroenterologyHuazhong University of Science and Technology Union Shenzhen Hospital, the 6th affiliated Hospital of Shenzhen University Medical SchoolShenzhenChina
| | - Qingping Zhang
- Neurology DepartmentHuazhong University of Science and Technology Union Shenzhen Hospital, the 6th affiliated Hospital of Shenzhen University Medical SchoolShenzhenChina
| | - Teng Wan
- Sports Medicine CenterHuazhong University of Science and Technology Union Shenzhen Hospital, the 6th affiliated Hospital of Shenzhen University Medical SchoolShenzhenChina
| | - Changyu Jiang
- Department of Pain Medicine and Shenzhen Municipal Key Laboratory for Pain MedicineHuazhong University of Science and Technology Union Shenzhen Hospital, the 6th affiliated Hospital of Shenzhen University Medical SchoolShenzhenChina
| | - Yuanchun Ye
- Quanzhou First Hospital Affiliated to Fujian Medical UniversityQuanzhouFujian ProvinceChina
| | - Haihuan Lin
- Cardiology DepartmentThe first Affiliated Hospital of Wenzhou Medical UniversityWenzhouChina
| | - Gang Fan
- Urology department, Huazhong University of Science and Technology Union Shenzhen Hospitalthe 6th affiliated Hospital of Shenzhen University Medical SchoolShenzhenChina
- Hunan Cancer Hospital, the Affiliated Cancer Hospital of Xiangya School of MedicineCentral South UniversityChangshaChina
| |
Collapse
|
3
|
Dhanda AS, Guttman JA. Localization of host endocytic and actin-associated proteins during Shigella flexneri intracellular motility and intercellular spreading. Anat Rec (Hoboken) 2022; 306:1088-1110. [PMID: 35582740 DOI: 10.1002/ar.24955] [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: 02/28/2022] [Revised: 04/24/2022] [Accepted: 04/27/2022] [Indexed: 11/10/2022]
Abstract
Shigella flexneri (S. flexneri), the causative agent of bacillary dysentery, uses an effector-mediated strategy to hijack host cells and cause disease. To propagate and spread within human tissues, S. flexneri bacteria commandeer the host actin cytoskeleton to generate slender actin-rich comet tails to move intracellularly, and later, plasma membrane actin-based protrusions to move directly between adjacent host cells. To facilitate intercellular bacterial spreading, large micron-sized endocytic-like membrane invaginations form at the periphery of neighboring host cells that come into contact with S. flexneri-containing membrane protrusions. While S. flexneri comet tails and membrane protrusions consist primarily of host actin cytoskeletal proteins, S. flexneri membrane invaginations remain poorly understood with only clathrin and the clathrin adapter epsin-1 localized to the structures. Tangentially, we recently reported that Listeria monocytogenes, another actin-hijacking pathogen, exploits an assortment of caveolar and actin-bundling proteins at their micron-sized membrane invaginations formed during their cell-to-cell movement. Thus, to further characterize the S. flexneri disease process, we set out to catalog the distribution of a variety of actin-associated and caveolar proteins during S. flexneri actin-based motility and cell-to-cell spreading. Here we show that actin-associated proteins found at L. monocytogenes comet tails and membrane protrusions mimic those present at S. flexneri comet tails with the exception of α-actinins 1 and 4, which were shed from S. flexneri membrane protrusions. We also demonstrate that all known host endocytic components found at L. monocytogenes membrane invaginations are also present at those formed during S. flexneri infections.
Collapse
Affiliation(s)
- Aaron Singh Dhanda
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| | - Julian Andrew Guttman
- Department of Biological Sciences, Centre for Cell Biology, Development, and Disease, Simon Fraser University, Burnaby, British Columbia, Canada
| |
Collapse
|
4
|
mDia1 Assembles a Linear F-Actin Coat at Membrane Invaginations To Drive Listeria monocytogenes Cell-to-Cell Spreading. mBio 2021; 12:e0293921. [PMID: 34781738 PMCID: PMC8593688 DOI: 10.1128/mbio.02939-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Direct cell-to-cell spreading of Listeria monocytogenes requires the bacteria to induce actin-based finger-like membrane protrusions in donor host cells that are endocytosed through caveolin-rich membrane invaginations by adjacent receiving cells. An actin shell surrounds these endocytic sites; however, its structure, composition, and functional significance remain elusive. Here, we show that the formin mDia1, but surprisingly not the Arp2/3 complex, is enriched at the membrane invaginations generated by L. monocytogenes during HeLa and Jeg-3 cell infections. Electron microscopy reveals a band of linear actin filaments that run along the longitudinal axis of the invagination membrane. Mechanistically, mDia1 expression is vital for the assembly of this F-actin shell. mDia1 is also required for the recruitment of Filamin A, a caveola-associated F-actin cross-linking protein, and caveolin-1 to the invaginations. Importantly, mixed-cell infection assays show that optimal caveolin-based L. monocytogenes cell-to-cell spreading correlates with the formation of the linear actin filament-containing shell by mDia1. IMPORTANCE Listeria monocytogenes spreads from one cell to another to colonize tissues. This cell-to-cell movement requires the propulsive force of an actin-rich comet tail behind the advancing bacterium, which ultimately distends the host plasma membrane into a slender bacterium-containing membrane protrusion. These membrane protrusions induce a corresponding invagination in the membrane of the adjacent host cell. The host cell that receives the protrusion utilizes caveolin-based endocytosis to internalize the structures, and filamentous actin lines these membrane invaginations. Here, we set out to determine the structure and function of this filamentous actin "shell." We demonstrate that the formin mDia1, but not the Arp2/3 complex, localizes to the invaginations. Morphologically, we show that this actin is organized into linear arrays and not branched dendritic networks. Mechanistically, we show that the actin shell is assembled by mDia1 and that mDia1 is required for efficient cell-to-cell transfer of L. monocytogenes.
Collapse
|
5
|
Charpentier JC, King PD. Mechanisms and functions of endocytosis in T cells. Cell Commun Signal 2021; 19:92. [PMID: 34503523 PMCID: PMC8427877 DOI: 10.1186/s12964-021-00766-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 07/17/2021] [Indexed: 11/11/2022] Open
Abstract
Once thought of primarily as a means to neutralize pathogens or to facilitate feeding, endocytosis is now known to regulate a wide range of eukaryotic cell processes. Among these are regulation of signal transduction, mitosis, lipid homeostasis, and directed migration, among others. Less well-appreciated are the roles various forms of endocytosis plays in regulating αβ and, especially, γδ T cell functions, such as T cell receptor signaling, antigen discovery by trogocytosis, and activated cell growth. Herein we examine the contribution of both clathrin-mediated and clathrin-independent mechanisms of endocytosis to T cell biology. Video Abstract
Collapse
Affiliation(s)
- John C Charpentier
- Department of Microbiology and Immunology, University of Michigan Medical School, 6606 Med Sci II, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5620, USA
| | - Philip D King
- Department of Microbiology and Immunology, University of Michigan Medical School, 6606 Med Sci II, 1150 West Medical Center Drive, Ann Arbor, MI, 48109-5620, USA.
| |
Collapse
|
6
|
Graceffa V. Physical and mechanical cues affecting biomaterial-mediated plasmid DNA delivery: insights into non-viral delivery systems. J Genet Eng Biotechnol 2021; 19:90. [PMID: 34142237 PMCID: PMC8211807 DOI: 10.1186/s43141-021-00194-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/09/2021] [Indexed: 12/17/2022]
Abstract
BACKGROUND Whilst traditional strategies to increase transfection efficiency of non-viral systems aimed at modifying the vector or the polyplexes/lipoplexes, biomaterial-mediated gene delivery has recently sparked increased interest. This review aims at discussing biomaterial properties and unravelling underlying mechanisms of action, for biomaterial-mediated gene delivery. DNA internalisation and cytoplasmic transport are initially discussed. DNA immobilisation, encapsulation and surface-mediated gene delivery (SMD), the role of extracellular matrix (ECM) and topographical cues, biomaterial stiffness and mechanical stimulation are finally outlined. MAIN TEXT Endocytic pathways and mechanisms to escape the lysosomal network are highly variable. They depend on cell and DNA complex types but can be diverted using appropriate biomaterials. 3D scaffolds are generally fabricated via DNA immobilisation or encapsulation. Degradation rate and interaction with the vector affect temporal patterns of DNA release and transgene expression. In SMD, DNA is instead coated on 2D surfaces. SMD allows the incorporation of topographical cues, which, by inducing cytoskeletal re-arrangements, modulate DNA endocytosis. Incorporation of ECM mimetics allows cell type-specific transfection, whereas in spite of discordances in terms of optimal loading regimens, it is recognised that mechanical loading facilitates gene transfection. Finally, stiffer 2D substrates enhance DNA internalisation, whereas in 3D scaffolds, the role of stiffness is still dubious. CONCLUSION Although it is recognised that biomaterials allow the creation of tailored non-viral gene delivery systems, there still are many outstanding questions. A better characterisation of endocytic pathways would allow the diversion of cell adhesion processes and cytoskeletal dynamics, in order to increase cellular transfection. Further research on optimal biomaterial mechanical properties, cell ligand density and loading regimens is limited by the fact that such parameters influence a plethora of other different processes (e.g. cellular adhesion, spreading, migration, infiltration, and proliferation, DNA diffusion and release) which may in turn modulate gene delivery. Only a better understanding of these processes may allow the creation of novel robust engineered systems, potentially opening up a whole new area of biomaterial-guided gene delivery for non-viral systems.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland.
- Department of Life Sciences, Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland.
| |
Collapse
|
7
|
Allard A, Lopes Dos Santos R, Campillo C. Remodelling of membrane tubules by the actin cytoskeleton. Biol Cell 2021; 113:329-343. [PMID: 33826772 DOI: 10.1111/boc.202000148] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/25/2021] [Accepted: 03/27/2021] [Indexed: 12/14/2022]
Abstract
Inside living cells, the remodelling of membrane tubules by actomyosin networks is crucial for processes such as intracellular trafficking or organelle reshaping. In this review, we first present various in vivo situations in which actin affects membrane tubule remodelling, then we recall some results on force production by actin dynamics and on membrane tubules physics. Finally, we show that our knowledge of the underlying mechanisms by which actomyosin dynamics affect tubule morphology has recently been moved forward. This is thanks to in vitro experiments that mimic cellular membranes and actin dynamics and allow deciphering the physics of tubule remodelling in biochemically controlled conditions, and shed new light on tubule shape regulation.
Collapse
Affiliation(s)
- Antoine Allard
- LAMBE, Université d'Évry, CNRS, CEA, Université Paris-Saclay, Évry-Courcouronnes, 91025, France.,Sorbonne Université, UPMC, Paris 06, Paris, France.,Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France.,Department of Physics, University of Warwick, Gibbet Hill Road, Coventry, CV4 7AL, UK
| | | | - Clément Campillo
- LAMBE, Université d'Évry, CNRS, CEA, Université Paris-Saclay, Évry-Courcouronnes, 91025, France
| |
Collapse
|
8
|
Shi X, Wen Z, Wang Y, Liu YJ, Shi K, Jiu Y. Feedback-Driven Mechanisms Between Phosphorylated Caveolin-1 and Contractile Actin Assemblies Instruct Persistent Cell Migration. Front Cell Dev Biol 2021; 9:665919. [PMID: 33928090 PMCID: PMC8076160 DOI: 10.3389/fcell.2021.665919] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/22/2021] [Indexed: 12/23/2022] Open
Abstract
The actin cytoskeleton and membrane-associated caveolae contribute to active processes, such as cell morphogenesis and motility. How these two systems interact and control directional cell migration is an outstanding question but remains understudied. Here we identified a negative feedback between contractile actin assemblies and phosphorylated caveolin-1 (CAV-1) in migrating cells. Cytoplasmic CAV-1 vesicles display actin-associated motilities by sliding along actin filaments or/and coupling to do retrograde flow with actomyosin bundles. Inhibition of contractile stress fibers, but not Arp2/3-dependent branched actin filaments, diminished the phosphorylation of CAV-1 on site Tyr14, and resulted in substantially increased size and decreased motility of cytoplasmic CAV-1 vesicles. Reciprocally, both the CAV-1 phospho-deficient mutation on site Tyr14 and CAV-1 knockout resulted in dramatic AMPK phosphorylation, further causing reduced active level of RhoA-myosin II and increased active level of Rac1-PAK1-Cofilin, consequently led to disordered contractile stress fibers and prominent lamellipodia. As a result, cells displayed depolarized morphology and compromised directional migration. Collectively, we propose a model in which feedback-driven regulation between actin and CAV-1 instructs persistent cell migration.
Collapse
Affiliation(s)
- Xuemeng Shi
- The Joint Program in Infection and Immunity, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,The Joint Program in Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Zeyu Wen
- Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yajun Wang
- Shanghai Institute of Cardiovascular Diseases, and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yan-Jun Liu
- Shanghai Institute of Cardiovascular Diseases, and Institutes of Biomedical Sciences, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Kun Shi
- The Joint Program in Infection and Immunity, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,The Joint Program in Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
| | - Yaming Jiu
- The Joint Program in Infection and Immunity, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China.,The Joint Program in Infection and Immunity, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,Key Laboratory of Molecular Virology and Immunology, The Center for Microbes, Development and Health, Institut Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
9
|
Abstract
Since the initial reports implicating caveolin-1 (CAV1) in neoplasia, the scientific community has made tremendous strides towards understanding how CAV1-dependent signaling and caveolae assembly modulate solid tumor growth. Once a solid neoplastic tumor reaches a certain size, it will increasingly rely on its stroma to meet the metabolic demands of the rapidly proliferating cancer cells, a limitation typically but not exclusively addressed via the formation of new blood vessels. Landmark studies using xenograft tumor models have highlighted the importance of stromal CAV1 during neoplastic blood vessel growth from preexisting vasculature, a process called angiogenesis, and helped identify endothelium-specific signaling events regulated by CAV1, such as vascular endothelial growth factor (VEGF) receptors as well as the endothelial nitric oxide (NO) synthase (eNOS) systems. This chapter provides a glimpse into the signaling events modulated by CAV1 and its scaffolding domain (CSD) during endothelial-specific aspects of neoplastic growth, such as vascular permeability, angiogenesis, and mechanotransduction.
Collapse
Affiliation(s)
- Pascal Bernatchez
- Department of Anesthesiology, Pharmacology & Therapeutics, Faculty of Medicine, University of British Columbia (UBC), 2176 Health Sciences mall, room 217, Vancouver, BC, V6T 1Z3, Canada. .,Centre for Heart & Lung Innovation, St. Paul's Hospital, Vancouver, Canada.
| |
Collapse
|
10
|
Lolo FN, Jiménez-Jiménez V, Sánchez-Álvarez M, Del Pozo MÁ. Tumor-stroma biomechanical crosstalk: a perspective on the role of caveolin-1 in tumor progression. Cancer Metastasis Rev 2021; 39:485-503. [PMID: 32514892 DOI: 10.1007/s10555-020-09900-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Tumor stiffening is a hallmark of malignancy that actively drives tumor progression and aggressiveness. Recent research has shed light onto several molecular underpinnings of this biomechanical process, which has a reciprocal crosstalk between tumor cells, stromal fibroblasts, and extracellular matrix remodeling at its core. This dynamic communication shapes the tumor microenvironment; significantly determines disease features including therapeutic resistance, relapse, or metastasis; and potentially holds the key for novel antitumor strategies. Caveolae and their components emerge as integrators of different aspects of cell function, mechanotransduction, and ECM-cell interaction. Here, we review our current knowledge on the several pivotal roles of the essential caveolar component caveolin-1 in this multidirectional biomechanical crosstalk and highlight standing questions in the field.
Collapse
Affiliation(s)
- Fidel Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Víctor Jiménez-Jiménez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Sánchez-Álvarez
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Miguel Ángel Del Pozo
- Mechanoadaptation and Caveolae Biology Lab, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| |
Collapse
|
11
|
Matthaeus C, Taraska JW. Energy and Dynamics of Caveolae Trafficking. Front Cell Dev Biol 2021; 8:614472. [PMID: 33692993 PMCID: PMC7939723 DOI: 10.3389/fcell.2020.614472] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 12/21/2020] [Indexed: 12/19/2022] Open
Abstract
Caveolae are 70–100 nm diameter plasma membrane invaginations found in abundance in adipocytes, endothelial cells, myocytes, and fibroblasts. Their bulb-shaped membrane domain is characterized and formed by specific lipid binding proteins including Caveolins, Cavins, Pacsin2, and EHD2. Likewise, an enrichment of cholesterol and other lipids makes caveolae a distinct membrane environment that supports proteins involved in cell-type specific signaling pathways. Their ability to detach from the plasma membrane and move through the cytosol has been shown to be important for lipid trafficking and metabolism. Here, we review recent concepts in caveolae trafficking and dynamics. Second, we discuss how ATP and GTP-regulated proteins including dynamin and EHD2 control caveolae behavior. Throughout, we summarize the potential physiological and cell biological roles of caveolae internalization and trafficking and highlight open questions in the field and future directions for study.
Collapse
Affiliation(s)
- Claudia Matthaeus
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| | - Justin W Taraska
- Biochemistry and Biophysics Center, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
12
|
Del Pozo MA, Lolo FN, Echarri A. Caveolae: Mechanosensing and mechanotransduction devices linking membrane trafficking to mechanoadaptation. Curr Opin Cell Biol 2020; 68:113-123. [PMID: 33188985 DOI: 10.1016/j.ceb.2020.10.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 09/21/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023]
Abstract
Mechanical forces (extracellular matrix stiffness, vascular shear stress, and muscle stretching) reaching the plasma membrane (PM) determine cell behavior. Caveolae are PM-invaginated nanodomains with specific lipid and protein composition. Being highly abundant in mechanically challenged tissues (muscles, lungs, vessels, and adipose tissues), they protect cells from mechanical stress damage. Caveolae flatten upon increased PM tension, enabling both force sensing and accommodation, critical for cell mechanoprotection and homeostasis. Thus, caveolae are highly plastic, ranging in complexity from flattened membranes to vacuolar invaginations surrounded by caveolae-rosettes-which also contribute to mechanoprotection. Caveolar components crosstalk with mechanotransduction pathways and recent studies show that they translocate from the PM to the nucleus to convey stress information. Furthermore, caveolae components can regulate membrane traffic from/to the PM to adapt to environmental mechanical forces. The interdependence between lipids and caveolae starts to be understood, and the relevance of caveolae-dependent membrane trafficking linked to mechanoadaption to different physiopathological processes is emerging.
Collapse
Affiliation(s)
- Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| | - Fidel-Nicolás Lolo
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Asier Echarri
- Mechanoadaptation and Caveolae Biology Laboratory, Area of Cell & Developmental Biology, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| |
Collapse
|
13
|
Allard A, Valentino F, Sykes C, Betz T, Campillo C. Fluctuations of a membrane nanotube covered with an actin sleeve. Phys Rev E 2020; 102:052402. [PMID: 33327147 DOI: 10.1103/physreve.102.052402] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Accepted: 08/11/2020] [Indexed: 06/12/2023]
Abstract
Many biological functions rely on the reshaping of cell membranes, in particular into nanotubes, which are covered in vivo by dynamic actin networks. Nanotubes are subject to thermal fluctuations, but the effect of these on cell functions is unknown. Here, we form nanotubes from liposomes using an optically trapped bead adhering to the liposome membrane. From the power spectral density of this bead, we study the nanotube fluctuations in the range of membrane tensions measured in vivo. We show that an actin sleeve covering the nanotube damps its high-frequency fluctuations because of the network viscoelasticity. Our work paves the way for further studies of the effect of nanotube fluctuations on cellular functions.
Collapse
Affiliation(s)
- A Allard
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France
- Sorbonne Université, UPMC, Paris 06, Paris, France
- LAMBE, Université d'Évry, CNRS, CEA, Université Paris-Saclay, 91025 Évry-Courcouronnes, France
| | - F Valentino
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France
- Sorbonne Université, UPMC, Paris 06, Paris, France
| | - C Sykes
- Laboratoire Physico Chimie Curie, Institut Curie, PSL Research University, CNRS UMR168, Paris, France
- Sorbonne Université, UPMC, Paris 06, Paris, France
| | - T Betz
- Institute of Cell Biology, Cells in Motion Interfaculty Center, Centre for Molecular Biology of Inflammation, 48149 Münster, Germany
| | - C Campillo
- LAMBE, Université d'Évry, CNRS, CEA, Université Paris-Saclay, 91025 Évry-Courcouronnes, France
| |
Collapse
|
14
|
Kessels MM, Qualmann B. Interplay between membrane curvature and the actin cytoskeleton. Curr Opin Cell Biol 2020; 68:10-19. [PMID: 32927373 DOI: 10.1016/j.ceb.2020.08.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/26/2022]
Abstract
An intimate interplay of the plasma membrane with curvature-sensing and curvature-inducing proteins would allow for defining specific sites or nanodomains of action at the plasma membrane, for example, for protrusion, invagination, and polarization. In addition, such connections are predestined to ensure spatial and temporal order and sequences. The combined forces of membrane shapers and the cortical actin cytoskeleton might hereby in particular be required to overcome the strong resistance against membrane rearrangements in case of high plasma membrane tension or cellular turgor. Interestingly, also the opposite might be necessary, the inhibition of both membrane shapers and cytoskeletal reinforcement structures to relieve membrane tension to protect cells from membrane damage and rupturing during mechanical stress. In this review article, we discuss recent conceptual advances enlightening the interplay of plasma membrane curvature and the cortical actin cytoskeleton during endocytosis, modulations of membrane tensions, and the shaping of entire cells.
Collapse
Affiliation(s)
- Michael M Kessels
- Institute of Biochemistry I, Jena University Hospital, Friedrich Schiller University Jena, Nonnenplan 2-4, 07743, Jena, Germany
| | - Britta Qualmann
- Institute of Biochemistry I, Jena University Hospital, Friedrich Schiller University Jena, Nonnenplan 2-4, 07743, Jena, Germany.
| |
Collapse
|
15
|
Tang K, Li S, Li P, Xia Q, Yang R, Li T, Li L, Jiang Y, Qin X, Yang H, Wu C, You F, Tan Y, Liu Y. Shear stress stimulates integrin β1 trafficking and increases directional migration of cancer cells via promoting deacetylation of microtubules. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2020; 1867:118676. [DOI: 10.1016/j.bbamcr.2020.118676] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 01/09/2020] [Accepted: 02/05/2020] [Indexed: 12/17/2022]
|
16
|
Putative Receptors for Gravity Sensing in Mammalian Cells: The Effects of Microgravity. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10062028] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Gravity is a constitutive force that influences life on Earth. It is sensed and translated into biochemical stimuli through the so called “mechanosensors”, proteins able to change their molecular conformation in order to amplify external cues causing several intracellular responses. Mechanosensors are widely represented in the human body with important structures such as otholiths in hair cells of vestibular system and statoliths in plants. Moreover, they are also present in the bone, where mechanical cues can cause bone resorption or formation and in muscle in which mechanical stimuli can increase the sensibility for mechanical stretch. In this review, we discuss the role of mechanosensors in two different conditions: normogravity and microgravity, emphasizing their emerging role in microgravity. Microgravity is a singular condition in which many molecular changes occur, strictly connected with the modified gravity force and free fall of bodies. Here, we first summarize the most important mechanosensors involved in normogravity and microgravity. Subsequently, we propose muscle LIM protein (MLP) and sirtuins as new actors in mechanosensing and signaling transduction under microgravity.
Collapse
|
17
|
Echarri A, Pavón DM, Sánchez S, García-García M, Calvo E, Huerta-López C, Velázquez-Carreras D, Viaris de Lesegno C, Ariotti N, Lázaro-Carrillo A, Strippoli R, De Sancho D, Alegre-Cebollada J, Lamaze C, Parton RG, Del Pozo MA. An Abl-FBP17 mechanosensing system couples local plasma membrane curvature and stress fiber remodeling during mechanoadaptation. Nat Commun 2019; 10:5828. [PMID: 31862885 PMCID: PMC6925243 DOI: 10.1038/s41467-019-13782-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 11/22/2019] [Indexed: 12/19/2022] Open
Abstract
Cells remodel their structure in response to mechanical strain. However, how mechanical forces are translated into biochemical signals that coordinate the structural changes observed at the plasma membrane (PM) and the underlying cytoskeleton during mechanoadaptation is unclear. Here, we show that PM mechanoadaptation is controlled by a tension-sensing pathway composed of c-Abl tyrosine kinase and membrane curvature regulator FBP17. FBP17 is recruited to caveolae to induce the formation of caveolar rosettes. FBP17 deficient cells have reduced rosette density, lack PM tension buffering capacity under osmotic shock, and cannot adapt to mechanical strain. Mechanistically, tension is transduced to the FBP17 F-BAR domain by direct phosphorylation mediated by c-Abl, a mechanosensitive molecule. This modification inhibits FBP17 membrane bending activity and releases FBP17-controlled inhibition of mDia1-dependent stress fibers, favoring membrane adaptation to increased tension. This mechanoprotective mechanism adapts the cell to changes in mechanical tension by coupling PM and actin cytoskeleton remodeling. Mechanical forces are sensed by cells and can alter plasma membrane properties, but biochemical changes underlying this are not clear. Here the authors show tension is sensed by c-Abl and FBP17, which couples changes in mechanical tension to remodelling of the plasma membrane and actin cytoskeleton.
Collapse
Affiliation(s)
- Asier Echarri
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| | - Dácil M Pavón
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Sara Sánchez
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - María García-García
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Enrique Calvo
- Proteomics Unit, Vascular Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Carla Huerta-López
- Molecular Mechanics of the Cardiovascular System Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Diana Velázquez-Carreras
- Molecular Mechanics of the Cardiovascular System Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Christine Viaris de Lesegno
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie - Centre de Recherche, PSL Research University, CNRS UMR3666, INSERM U1143, 75248, Paris, France
| | - Nicholas Ariotti
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ana Lázaro-Carrillo
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.,Departamento de Biología, Universidad Autónoma de Madrid, Cantoblanco, 28049, Madrid, Spain
| | | | - David De Sancho
- Departamento de Ciencia y Tecnología de Polímeros, Euskal Herriko Unibertsitatea, 20018, Donostia-San Sebastián, Spain.,Donostia International Physics Center, Manuel Lardizabal Ibilbidea, 4, 20018, Donostia-San Sebastián, Spain
| | - Jorge Alegre-Cebollada
- Molecular Mechanics of the Cardiovascular System Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain
| | - Christophe Lamaze
- Membrane Mechanics and Dynamics of Intracellular Signaling Laboratory, Institut Curie - Centre de Recherche, PSL Research University, CNRS UMR3666, INSERM U1143, 75248, Paris, France
| | - Robert G Parton
- The Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.,The Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Miguel A Del Pozo
- Mechanoadaptation and Caveolae Biology Laboratory, Cell and Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, Madrid, Spain.
| |
Collapse
|
18
|
Okuda A, Tahara S, Hirose H, Takeuchi T, Nakase I, Ono A, Takehashi M, Tanaka S, Futaki S. Oligoarginine-Bearing Tandem Repeat Penetration-Accelerating Sequence Delivers Protein to Cytosol via Caveolae-Mediated Endocytosis. Biomacromolecules 2019; 20:1849-1859. [PMID: 30893557 DOI: 10.1021/acs.biomac.8b01299] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
To facilitate the cytosolic delivery of larger molecules such as proteins, we developed a new cell-penetrating peptide sequence, named Pas2r12, consisting of a repeated Pas sequence (FFLIG-FFLIG) and d-dodeca-arginine (r12). This peptide significantly enhanced the cellular uptake and cytosolic release of enhanced green fluorescent protein and immunoglobulin G as cargos. We found that simply mixing Pas2r12 with cargos could generate cytosolic introducible forms. The cytosolic delivery of cargos by Pas2r12 was found to be an energy-requiring process, to rely on actin polymerization, and to be suppressed by caveolae-mediated endocytosis inhibitors (genistein and methyl-β-cyclodextrin) and small interfering RNA against caveolin-1. These results suggest that Pas2r12 enhances membrane penetration of cargos without the need for cross-linking and that caveolae-mediated endocytosis may be the route by which cytosolic delivery is enhanced.
Collapse
Affiliation(s)
- Akiko Okuda
- Department of Medical Technology, Graduate School of Health Sciences , Niigata University , 746 Asahimachidori-2 , Chuo-ku, Niigata , Niigata 951-8518 , Japan
| | - Shinya Tahara
- Department of Medical Technology, Graduate School of Health Sciences , Niigata University , 746 Asahimachidori-2 , Chuo-ku, Niigata , Niigata 951-8518 , Japan
| | - Hisaaki Hirose
- Institute for Chemical Research , Kyoto University , Uji , Kyoto 611-0011 , Japan
| | - Toshihide Takeuchi
- Institute for Chemical Research , Kyoto University , Uji , Kyoto 611-0011 , Japan
| | - Ikuhiko Nakase
- Graduate School of Science , Osaka Prefecture University , Naka-ku, Sakai , Osaka 599-8570 , Japan
| | - Atsushi Ono
- Department of Medical Technology, Graduate School of Health Sciences , Niigata University , 746 Asahimachidori-2 , Chuo-ku, Niigata , Niigata 951-8518 , Japan
| | - Masanori Takehashi
- Laboratory of Pathophysiology and Pharmacotherapeutics, Faculty of Pharmacy , Osaka Ohtani University , Tondabayashi , Osaka 584-8540 , Japan
| | - Seigo Tanaka
- Laboratory of Pathophysiology and Pharmacotherapeutics, Faculty of Pharmacy , Osaka Ohtani University , Tondabayashi , Osaka 584-8540 , Japan
| | - Shiroh Futaki
- Institute for Chemical Research , Kyoto University , Uji , Kyoto 611-0011 , Japan
| |
Collapse
|
19
|
Role of the Endocytosis of Caveolae in Intracellular Signaling and Metabolism. PROGRESS IN MOLECULAR AND SUBCELLULAR BIOLOGY 2019; 57:203-234. [PMID: 30097777 DOI: 10.1007/978-3-319-96704-2_8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Caveolae are 60-80 nm invaginated plasma membrane (PM) nanodomains, with a specific lipid and protein composition, which assist and regulate multiple processes in the plasma membrane-ranging from the organization of signalling complexes to the mechanical adaptation to changes in PM tension. However, since their initial descriptions, these structures have additionally been found tightly linked to internalization processes, mechanoadaptation, to the regulation of signalling events and of endosomal trafficking. Here, we review caveolae biology from this perspective, and its implications for cell physiology and disease.
Collapse
|
20
|
Rangel L, Bernabé-Rubio M, Fernández-Barrera J, Casares-Arias J, Millán J, Alonso MA, Correas I. Caveolin-1α regulates primary cilium length by controlling RhoA GTPase activity. Sci Rep 2019; 9:1116. [PMID: 30718762 PMCID: PMC6362014 DOI: 10.1038/s41598-018-38020-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2018] [Accepted: 10/26/2018] [Indexed: 11/08/2022] Open
Abstract
The primary cilium is a single non-motile protrusion of the plasma membrane of most types of mammalian cell. The structure, length and function of the primary cilium must be tightly controlled because their dysfunction is associated with disease. Caveolin 1 (Cav1), which is best known as a component of membrane invaginations called caveolae, is also present in non-caveolar membrane domains whose function is beginning to be understood. We show that silencing of α and β Cav1 isoforms in different cell lines increases ciliary length regardless of the route of primary ciliogenesis. The sole expression of Cav1α, which is distributed at the apical membrane, restores normal cilium size in Cav1 KO MDCK cells. Cells KO for only Cav1α, which also show long cilia, have a disrupted actin cytoskeleton and reduced RhoA GTPase activity at the apical membrane, and a greater accumulation of Rab11 vesicles at the centrosome. Subsequent experiments showed that DIA1 and ROCK help regulate ciliary length. Since MDCK cells lack apical caveolae, our results imply that non-caveolar apical Cav1α is an important regulator of ciliary length, exerting its effect via RhoA and its effectors, ROCK and DIA1.
Collapse
Affiliation(s)
- Laura Rangel
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel Bernabé-Rubio
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Jaime Fernández-Barrera
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Javier Casares-Arias
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Jaime Millán
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain
| | - Miguel A Alonso
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.
| | - Isabel Correas
- Department of Cell Biology and Immunology, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, Madrid, Spain.
- Department of Molecular Biology, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
21
|
Hozumi K, Teranishi Y, Enomoto S, Katagiri F, Kikkawa Y, Nomizu M. Identification of specific integrin cross-talk for dermal fibroblast cell adhesion using a mixed peptide-chitosan matrix. J Biomater Appl 2019; 33:893-902. [PMID: 30638115 DOI: 10.1177/0885328218823457] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Extracellular matrix molecules are recognized by several integrin subtypes, making identification of cross-talk among different integrin subtypes difficult. Here, we evaluated the cross-talk of integrin subtypes using four different integrin-binding peptides (FIB1; integrin αvβ3/α5β1, A2G10; integrin α6β1, EF1zz; integrin α2β1, or 531; integrin α3β1) derived from extracellular matrix molecules. Various combinations of two different integrin-binding peptides were mixed and conjugated on a chitosan matrix at various molar ratios and were evaluated for cell attachment activity. FIB1/A2G10 (molar ratio 5:5; total 10 nmol/well)-chitosan matrix significantly enhanced cell attachment activity compared with sum of the cell attachment activity on FIB1 (5 nmol/well)-chitosan matrices and A2G10 (5 nmol/well)-chitosan matrices, respectively. However, none of the other peptides showed a significant activity change when they were mixed and conjugated on a chitosan matrix. We investigated the mechanisms of this enhancement. FIB1/A2G10 (8:2 or 6:4)-chitosan matrix increased the cell spreading, phosphorylation of focal adhesion kinase at Y397, and slightly decreased phosphorylation of caveolin-1 at Y14 in fibroblasts compared with FIB1-chitosan and A2G10-chitosan matrices. These results indicate that FIB1/A2G10 (8:2 or 6:4)-chitosan matrix synergistically enhances cell attachment, suggesting that integrins αvβ3/α5β1 and α6β1 are involved in a cross-talk and synergistically enhance cell attachment. These findings also suggest that the mixed peptide-chitosan matrix system can regulate the ratio of two different peptides and is useful for evaluating cellular functions through receptor-specific cross-talk. Further, FIB1/A2G10 (8:2 or 6:4)-chitosan matrix could be a useful material for tissue engineering.
Collapse
Affiliation(s)
- Kentaro Hozumi
- 1 Department of Applied Clinical Dietetics, Kitasato Junior College of Health and Hygienic Sciences, Minamiuonuma, Niigata, Japan.,2 Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Yui Teranishi
- 2 Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Sayaka Enomoto
- 2 Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Fumihiko Katagiri
- 2 Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Yamato Kikkawa
- 2 Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| | - Motoyoshi Nomizu
- 2 Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences, Hachioji, Tokyo, Japan
| |
Collapse
|
22
|
Mendoza-Topaz C, Yeow I, Riento K, Nichols BJ. BioID identifies proteins involved in the cell biology of caveolae. PLoS One 2018; 13:e0209856. [PMID: 30589899 PMCID: PMC6307745 DOI: 10.1371/journal.pone.0209856] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 12/12/2018] [Indexed: 01/20/2023] Open
Abstract
The mechanisms controlling the abundance and sub-cellular distribution of caveolae are not well described. A first step towards determining such mechanisms would be identification of relevant proteins that interact with known components of caveolae. Here, we applied proximity biotinylation (BioID) to identify a list of proteins that may interact with the caveolar protein cavin1. Screening of these candidates using siRNA to reduce their expression revealed that one of them, CSDE1, regulates the levels of mRNAs and protein expression for multiple components of caveolae. A second candidate, CD2AP, co-precipitated with cavin1. Caveolar proteins were observed in characteristic and previously un-described linear arrays adjacent to cell-cell junctions in both MDCK cells, and in HeLa cells overexpressing an active form of the small GTPase Rac1. CD2AP was required for the recruitment of caveolar proteins to these linear arrays. We conclude that BioID will be useful in identification of new proteins involved in the cell biology of caveolae, and that interaction between CD2AP and cavin1 may have an important role in regulating the sub-cellular distribution of caveolae.
Collapse
Affiliation(s)
| | - I. Yeow
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - K. Riento
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
| | - B. J. Nichols
- MRC Laboratory of Molecular Biology, Cambridge, United Kingdom
- * E-mail:
| |
Collapse
|
23
|
Thottacherry JJ, Kosmalska AJ, Kumar A, Vishen AS, Elosegui-Artola A, Pradhan S, Sharma S, Singh PP, Guadamillas MC, Chaudhary N, Vishwakarma R, Trepat X, Del Pozo MA, Parton RG, Rao M, Pullarkat P, Roca-Cusachs P, Mayor S. Mechanochemical feedback control of dynamin independent endocytosis modulates membrane tension in adherent cells. Nat Commun 2018; 9:4217. [PMID: 30310066 PMCID: PMC6181995 DOI: 10.1038/s41467-018-06738-5] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 09/04/2018] [Indexed: 12/31/2022] Open
Abstract
Plasma membrane tension regulates many key cellular processes. It is modulated by, and can modulate, membrane trafficking. However, the cellular pathway(s) involved in this interplay is poorly understood. Here we find that, among a number of endocytic processes operating simultaneously at the cell surface, a dynamin independent pathway, the CLIC/GEEC (CG) pathway, is rapidly and specifically upregulated upon a sudden reduction of tension. Moreover, inhibition (activation) of the CG pathway results in lower (higher) membrane tension. However, alteration in membrane tension does not directly modulate CG endocytosis. This requires vinculin, a mechano-transducer recruited to focal adhesion in adherent cells. Vinculin acts by controlling the levels of a key regulator of the CG pathway, GBF1, at the plasma membrane. Thus, the CG pathway directly regulates membrane tension and is in turn controlled via a mechano-chemical feedback inhibition, potentially leading to homeostatic regulation of membrane tension in adherent cells. Plasma membrane tension is an important factor that regulates many key cellular processes. Here authors show that a specific dynamin-independent endocytic pathway is modulated by changes in tension via the mechano-transducer vinculin.
Collapse
Affiliation(s)
- Joseph Jose Thottacherry
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bengaluru, 560065, India
| | - Anita Joanna Kosmalska
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, 08028, Spain.,University of Barcelona, Barcelona, 08036, Spain
| | - Amit Kumar
- Raman Research Institute, C. V. Raman Avenue, Bengaluru, 560080, India
| | - Amit Singh Vishen
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bengaluru, 560065, India.,Simons Centre for the Study of Living Machines, National Centre for Biological Sciences (NCBS), Bengaluru, 560065, India
| | | | - Susav Pradhan
- Raman Research Institute, C. V. Raman Avenue, Bengaluru, 560080, India
| | - Sumit Sharma
- CSIR - Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Parvinder P Singh
- CSIR - Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Marta C Guadamillas
- Integrin Signalling Lab, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029, Spain
| | - Natasha Chaudhary
- University of Queensland, Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, St Lucia, QLD, 4072, Australia.,Department of Biochemistry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Ram Vishwakarma
- CSIR - Indian Institute of Integrative Medicine, Jammu, 180001, India
| | - Xavier Trepat
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, 08028, Spain.,University of Barcelona, Barcelona, 08036, Spain.,Centro de Investigación Biomédica en Red en Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN) and Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, 08010, Spain
| | - Miguel A Del Pozo
- Integrin Signalling Lab, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, 28029, Spain
| | - Robert G Parton
- University of Queensland, Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, St Lucia, QLD, 4072, Australia
| | - Madan Rao
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bengaluru, 560065, India.,Simons Centre for the Study of Living Machines, National Centre for Biological Sciences (NCBS), Bengaluru, 560065, India
| | - Pramod Pullarkat
- Raman Research Institute, C. V. Raman Avenue, Bengaluru, 560080, India
| | - Pere Roca-Cusachs
- Institute for Bioengineering of Catalonia (IBEC), Barcelona, 08028, Spain.,University of Barcelona, Barcelona, 08036, Spain
| | - Satyajit Mayor
- National Centre for Biological Sciences (NCBS), Tata Institute of Fundamental Research (TIFR), Bellary Road, Bengaluru, 560065, India. .,Institute for Stem Cell Biology and Regenerative Medicine, Tata Institute of Fundamental Research (TIFR), Bengaluru, 560065, India.
| |
Collapse
|
24
|
Local actin polymerization during endocytic carrier formation. Biochem Soc Trans 2018; 46:565-576. [DOI: 10.1042/bst20170355] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/06/2018] [Accepted: 03/16/2018] [Indexed: 12/20/2022]
Abstract
Extracellular macromolecules, pathogens and cell surface proteins rely on endocytosis to enter cells. Key steps of endocytic carrier formation are cargo molecule selection, plasma membrane folding and detachment from the cell surface. While dedicated proteins mediate each step, the actin cytoskeleton contributes to all. However, its role can be indirect to the actual molecular events driving endocytosis. Here, we review our understanding of the molecular steps mediating local actin polymerization during the formation of endocytic carriers. Clathrin-mediated endocytosis is the least reliant on local actin polymerization, as it is only engaged to counter forces induced by membrane tension or cytoplasmic pressure. Two opposite situations are coated pit formation in yeast and at the basolateral surface of polarized mammalian cells which are, respectively, dependent and independent on actin polymerization. Conversely, clathrin-independent endocytosis forming both nanometer [CLIC (clathrin-independent carriers)/GEEC (glycosylphosphatidylinositol (GPI)-anchored protein enriched endocytic compartments), caveolae, FEME (fast endophilin-mediated endocytosis) and IL-2β (interleukin-2β) uptake] and micrometer carriers (macropinocytosis) are dependent on actin polymerization to power local membrane deformation and carrier budding. A variety of endocytic adaptors can recruit and activate the Cdc42/N-WASP or Rac1/WAVE complexes, which, in turn, engage the Arp2/3 complex, thereby mediating local actin polymerization at the membrane. However, the molecular steps for RhoA and formin-mediated actin bundling during endocytic pit formation remain unclear.
Collapse
|
25
|
ITGB1-dependent upregulation of Caveolin-1 switches TGFβ signalling from tumour-suppressive to oncogenic in prostate cancer. Sci Rep 2018; 8:2338. [PMID: 29402961 PMCID: PMC5799174 DOI: 10.1038/s41598-018-20161-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Accepted: 01/15/2018] [Indexed: 01/10/2023] Open
Abstract
Caveolin-1 (CAV1) is over-expressed in prostate cancer (PCa) and is associated with adverse prognosis, but the molecular mechanisms linking CAV1 expression to disease progression are poorly understood. Extensive gene expression correlation analysis, quantitative multiplex imaging of clinical samples, and analysis of the CAV1-dependent transcriptome, supported that CAV1 re-programmes TGFβ signalling from tumour suppressive to oncogenic (i.e. induction of SLUG, PAI-1 and suppression of CDH1, DSP, CDKN1A). Supporting such a role, CAV1 knockdown led to growth arrest and inhibition of cell invasion in prostate cancer cell lines. Rationalized RNAi screening and high-content microscopy in search for CAV1 upstream regulators revealed integrin beta1 (ITGB1) and integrin associated proteins as CAV1 regulators. Our work suggests TGFβ signalling and beta1 integrins as potential therapeutic targets in PCa over-expressing CAV1, and contributes to better understand the paradoxical dual role of TGFβ in tumour biology.
Collapse
|
26
|
Pagnozzi LA, Butcher JT. Mechanotransduction Mechanisms in Mitral Valve Physiology and Disease Pathogenesis. Front Cardiovasc Med 2017; 4:83. [PMID: 29312958 PMCID: PMC5744129 DOI: 10.3389/fcvm.2017.00083] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2017] [Accepted: 12/07/2017] [Indexed: 01/13/2023] Open
Abstract
The mitral valve exists in a mechanically demanding environment, with the stress of each cardiac cycle deforming and shearing the native fibroblasts and endothelial cells. Cells and their extracellular matrix exhibit a dynamic reciprocity in the growth and formation of tissue through mechanotransduction and continuously adapt to physical cues in their environment through gene, protein, and cytokine expression. Valve disease is the most common congenital heart defect with watchful waiting and valve replacement surgery the only treatment option. Mitral valve disease (MVD) has been linked to a variety of mechano-active genes ranging from extracellular components, mechanotransductive elements, and cytoplasmic and nuclear transcription factors. Specialized cell receptors, such as adherens junctions, cadherins, integrins, primary cilia, ion channels, caveolae, and the glycocalyx, convert mechanical cues into biochemical responses via a complex of mechanoresponsive elements, shared signaling modalities, and integrated frameworks. Understanding mechanosensing and transduction in mitral valve-specific cells may allow us to discover unique signal transduction pathways between cells and their environment, leading to cell or tissue specific mechanically targeted therapeutics for MVD.
Collapse
Affiliation(s)
- Leah A. Pagnozzi
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| | - Jonathan T. Butcher
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, United States
| |
Collapse
|
27
|
WIP Drives Tumor Progression through YAP/TAZ-Dependent Autonomous Cell Growth. Cell Rep 2017; 17:1962-1977. [PMID: 27851961 DOI: 10.1016/j.celrep.2016.10.064] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 09/12/2016] [Accepted: 10/18/2016] [Indexed: 11/21/2022] Open
Abstract
In cancer, the deregulation of growth signaling pathways drives changes in the cell's architecture and its environment that allow autonomous growth of tumors. These cells then acquire a tumor-initiating "stemness" phenotype responsible for disease advancement to more aggressive stages. Here, we show that high levels of the actin cytoskeleton-associated protein WIP (WASP-interacting protein) correlates with tumor growth, both of which are linked to the tumor-initiating cell phenotype. We find that WIP controls tumor growth by boosting signals that stabilize the YAP/TAZ complex via a mechanism mediated by the endocytic/endosomal system. When WIP levels are high, the β-catenin Adenomatous polyposis coli (APC)-axin-GSK3 destruction complex is sequestered to the multi-vesicular body compartment, where its capacity to degrade YAP/TAZ is inhibited. YAP/TAZ stability is dependent on Rac, p21-activated kinase (PAK) and mammalian diaphanous-related formin (mDia), and is Hippo independent. This close biochemical relationship indicates an oncogenic role for WIP in the physiology of cancer pathology by increasing YAP/TAZ stability.
Collapse
|
28
|
Hissa B, Oakes PW, Pontes B, Ramírez-San Juan G, Gardel ML. Cholesterol depletion impairs contractile machinery in neonatal rat cardiomyocytes. Sci Rep 2017; 7:43764. [PMID: 28256617 PMCID: PMC5335656 DOI: 10.1038/srep43764] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 01/27/2017] [Indexed: 02/06/2023] Open
Abstract
Cholesterol regulates numerous cellular processes. Depleting its synthesis in skeletal myofibers induces vacuolization and contraction impairment. However, little is known about how cholesterol reduction affects cardiomyocyte behavior. Here, we deplete cholesterol by incubating neonatal cardiomyocytes with methyl-beta-cyclodextrin. Traction force microscopy shows that lowering cholesterol increases the rate of cell contraction and generates defects in cell relaxation. Cholesterol depletion also increases membrane tension, Ca2+ spikes frequency and intracellular Ca2+ concentration. These changes can be correlated with modifications in caveolin-3 and L-Type Ca2+ channel distributions across the sarcolemma. Channel regulation is also compromised since cAMP-dependent PKA activity is enhanced, increasing the probability of L-Type Ca2+ channel opening events. Immunofluorescence reveals that cholesterol depletion abrogates sarcomeric organization, changing spacing and alignment of α-actinin bands due to increase in proteolytic activity of calpain. We propose a mechanism in which cholesterol depletion triggers a signaling cascade, culminating with contraction impairment and myofibril disruption in cardiomyocytes.
Collapse
Affiliation(s)
- Barbara Hissa
- James Franck Institute, Institute for Biophysical Dynamics and Physics Department, University of Chicago, Chicago, IL, United States
| | - Patrick W. Oakes
- James Franck Institute, Institute for Biophysical Dynamics and Physics Department, University of Chicago, Chicago, IL, United States
| | - Bruno Pontes
- LPO-COPEA, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Guillermina Ramírez-San Juan
- James Franck Institute, Institute for Biophysical Dynamics and Physics Department, University of Chicago, Chicago, IL, United States
| | - Margaret L. Gardel
- James Franck Institute, Institute for Biophysical Dynamics and Physics Department, University of Chicago, Chicago, IL, United States
| |
Collapse
|
29
|
Senju Y, Suetsugu S. Possible regulation of caveolar endocytosis and flattening by phosphorylation of F-BAR domain protein PACSIN2/Syndapin II. BIOARCHITECTURE 2016; 5:70-7. [PMID: 26745030 PMCID: PMC4832444 DOI: 10.1080/19490992.2015.1128604] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
ABSTRACT. Caveolae are flask-shaped invaginations of the plasma membrane. The BAR domain proteins form crescent-shaped dimers, and their oligomeric filaments are considered to form spirals at the necks of invaginations, such as clathrin-coated pits and caveolae. PACSIN2/Syndapin II is one of the BAR domain-containing proteins, and is localized at the necks of caveolae. PACSIN2 is thought to function in the scission and stabilization of caveolae, through binding to dynamin-2 and EHD2, respectively. These two functions are considered to be switched by PACSIN2 phosphorylation by protein kinase C (PKC) upon hypotonic stress and sheer stress. The phosphorylation decreases the membrane binding affinity of PACSIN2, leading to its removal from caveolae. The removal of the putative oligomeric spiral of PACSIN2 from caveolar membrane invaginations could lead to the deformation of caveolae. Indeed, PACSIN2 removal from caveolae is accompanied by the recruitment of dynamin-2, suggesting that the removal provides space for the function of dynamin-2. Otherwise, the removal of PACSIN2 decreases the stability of caveolae, which could result in the flattening of caveolae. In contrast, an increase in the amount of EHD2 restored caveolar stability. Therefore, PACSIN2 at caveolae stabilizes caveolae, but its removal by phosphorylation could induce both caveolar endocytosis and flattening.
Collapse
Affiliation(s)
- Yosuke Senju
- a Institute of Biotechnology; University of Helsinki ; Helsinki , Finland
| | - Shiro Suetsugu
- b Laboratory of Molecular Medicine and Cell Biology; Graduate School of Biosciences; Nara Institute of Science and Technology ; Ikoma , Japan
| |
Collapse
|
30
|
Abstract
The Abelson tyrosine kinases were initially identified as drivers of leukemia in mice and humans. The Abl family kinases Abl1 and Abl2 regulate diverse cellular processes during development and normal homeostasis, and their functions are subverted during inflammation, cancer and other pathologies. Abl kinases can be activated by multiple stimuli leading to cytoskeletal reorganization required for cell morphogenesis, motility, adhesion and polarity. Depending on the cellular context, Abl kinases regulate cell survival and proliferation. Emerging data support important roles for Abl kinases in pathologies linked to inflammation. Among these are neurodegenerative diseases and inflammatory pathologies. Unexpectedly, Abl kinases have also been identified as important players in mammalian host cells during microbial pathogenesis. Thus, the use of Abl kinase inhibitors might prove to be effective in the treatment of pathologies beyond leukemia and solid tumors. In this Cell Science at a Glance article and in the accompanying poster, we highlight the emerging roles of Abl kinases in the regulation of cellular processes in normal cells and diverse pathologies ranging from cancer to microbial pathogenesis.
Collapse
Affiliation(s)
- Aaditya Khatri
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jun Wang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ann Marie Pendergast
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
31
|
Xu Q, Cao M, Song H, Chen S, Qian X, Zhao P, Ren H, Tang H, Wang Y, Wei Y, Zhu Y, Qi Z. Caveolin-1-mediated Japanese encephalitis virus entry requires a two-step regulation of actin reorganization. Future Microbiol 2016; 11:1227-1248. [DOI: 10.2217/fmb-2016-0002] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Aim: To investigate the detailed mechanism of Japanese encephalitis virus (JEV) cell entry. Materials & methods: Utilize a siRNA library targeting cellular membrane trafficking genes to identify key molecules that mediate JEV entry into human neuronal cells. Results: JEV enters human neuronal cells by caveolin-1-mediated endocytosis, which depends on a two-step regulation of actin cytoskeleton remodeling triggered by RhoA and Rac1: RhoA activation promoted the phosphorylation of caveolin-1, and then Rac1 activation facilitated caveolin-associated viral internalization. Specifically, virus attachment activates the EGFR–PI3K signaling pathway, thereby leading to RhoA activation. Conclusion: This work provides a detailed picture of the entry route and intricate cellular events following the entry of JEV into human neuronal cells, and promotes a better understanding of JEV entry.
Collapse
Affiliation(s)
- Qingqiang Xu
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Mingmei Cao
- Department of Medical Microbiology & Parasitology, Second Military Medical University, Shanghai 200433, China
| | - Hongyuan Song
- Department of Ophthalmology, Changhai Hospital, Second Military Medical University, Shanghai 200433, China
| | - Shenglin Chen
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Xijing Qian
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Ping Zhao
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Hao Ren
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Hailin Tang
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Yan Wang
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Youheng Wei
- State Key Laboratory of Genetic Engineering, Institute of Genetics, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Yongzhe Zhu
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| | - Zhongtian Qi
- Department of Microbiology, Second Military Medical University, Shanghai Key Laboratory of Medical Biodefense, Shanghai 200433, China
| |
Collapse
|
32
|
Scaffolding protein IQGAP1: an insulin-dependent link between caveolae and the cytoskeleton in primary human adipocytes? Biochem J 2016; 473:3177-88. [PMID: 27458251 DOI: 10.1042/bcj20160581] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2016] [Accepted: 07/25/2016] [Indexed: 12/15/2022]
Abstract
The ubiquitously expressed IQ motif-containing GTPase activating protein-1 (IQGAP1) is a scaffolding protein implicated in an array of cellular functions, in particular by binding to cytoskeletal elements and signaling proteins. A role of IQGAP1 in adipocytes has not been reported. We therefore investigated the cellular IQGAP1 interactome in primary human adipocytes. Immunoprecipitation and quantitative mass spectrometry identified caveolae and caveolae-associated proteins as the major IQGAP1 interactors alongside cytoskeletal proteins. We confirmed co-localization of IQGAP1 with the defining caveolar marker protein caveolin-1 by confocal microscopy and proximity ligation assay. Most interestingly, insulin enhanced the number of IQGAP1 interactions with caveolin-1 by five-fold. Moreover, we found a significantly reduced abundance of IQGAP1 in adipocytes from patients with type 2 diabetes compared with cells from nondiabetic control subjects. Both the abundance of IQGAP1 protein and mRNA were reduced, indicating a transcriptional defect in diabetes. Our findings suggest a novel role of IQGAP1 in insulin-regulated interaction between caveolae and cytoskeletal elements of the adipocyte, and that this is quelled in the diabetic state.
Collapse
|
33
|
Sharonov GV, Balatskaya MN, Tkachuk VA. Glycosylphosphatidylinositol-anchored proteins as regulators of cortical cytoskeleton. BIOCHEMISTRY (MOSCOW) 2016; 81:636-50. [DOI: 10.1134/s0006297916060110] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
34
|
Interphase adhesion geometry is transmitted to an internal regulator for spindle orientation via caveolin-1. Nat Commun 2016; 7:ncomms11858. [PMID: 27292265 PMCID: PMC4910015 DOI: 10.1038/ncomms11858] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2015] [Accepted: 05/09/2016] [Indexed: 12/31/2022] Open
Abstract
Despite theoretical and physical studies implying that cell-extracellular matrix adhesion geometry governs the orientation of the cell division axis, the molecular mechanisms that translate interphase adhesion geometry to the mitotic spindle orientation remain elusive. Here, we show that the cellular edge retraction during mitotic cell rounding correlates with the spindle axis. At the onset of mitotic cell rounding, caveolin-1 is targeted to the retracting cortical region at the proximal end of retraction fibres, where ganglioside GM1-enriched membrane domains with clusters of caveola-like structures are formed in an integrin and RhoA-dependent manner. Furthermore, Gαi1–LGN–NuMA, a well-known regulatory complex of spindle orientation, is targeted to the caveolin-1-enriched cortical region to guide the spindle axis towards the cellular edge retraction. We propose that retraction-induced cortical heterogeneity of caveolin-1 during mitotic cell rounding sets the spindle orientation in the context of adhesion geometry. Studies imply that cell adhesion geometry during interphase dictates the orientation of the cell division axis. Here the authors show that accumulation of caveolin-1 to rapidly retracting regions during cell rounding sets the spindle orientation by recruiting Gαi1-LGN-NuMA to the cortex.
Collapse
|
35
|
Echarri A, Del Pozo MA. Caveolae - mechanosensitive membrane invaginations linked to actin filaments. J Cell Sci 2015; 128:2747-58. [PMID: 26159735 DOI: 10.1242/jcs.153940] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
An essential property of the plasma membrane of mammalian cells is its plasticity, which is required for sensing and transmitting of signals, and for accommodating the tensional changes imposed by its environment or its own biomechanics. Caveolae are unique invaginated membrane nanodomains that play a major role in organizing signaling, lipid homeostasis and adaptation to membrane tension. Caveolae are frequently associated with stress fibers, a major regulator of membrane tension and cell shape. In this Commentary, we discuss recent studies that have provided new insights into the function of caveolae and have shown that trafficking and organization of caveolae are tightly regulated by stress-fiber regulators, providing a functional link between caveolae and stress fibers. Furthermore, the tension in the plasma membrane determines the curvature of caveolae because they flatten at high tension and invaginate at low tension, thus providing a tension-buffering system. Caveolae also regulate multiple cellular pathways, including RhoA-driven actomyosin contractility and other mechanosensitive pathways, suggesting that caveolae could couple mechanotransduction pathways to actin-controlled changes in tension through their association with stress fibers. Therefore, we argue here that the association of caveolae with stress fibers could provide an important strategy for cells to deal with mechanical stress.
Collapse
Affiliation(s)
- Asier Echarri
- Integrin Signaling Laboratory, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Miguel A Del Pozo
- Integrin Signaling Laboratory, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| |
Collapse
|
36
|
Senju Y, Rosenbaum E, Shah C, Hamada-Nakahara S, Itoh Y, Yamamoto K, Hanawa-Suetsugu K, Daumke O, Suetsugu S. Phosphorylation of PACSIN2 by protein kinase C triggers the removal of caveolae from the plasma membrane. J Cell Sci 2015; 128:2766-80. [PMID: 26092940 DOI: 10.1242/jcs.167775] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Accepted: 06/12/2015] [Indexed: 01/08/2023] Open
Abstract
PACSIN2, a membrane-sculpting BAR domain protein, localizes to caveolae. Here, we found that protein kinase C (PKC) phosphorylates PACSIN2 at serine 313, thereby decreasing its membrane binding and tubulation capacities. Concomitantly, phosphorylation decreased the time span for which caveolae could be tracked at the plasma membrane (the 'tracking duration'). Analyses of the phospho-mimetic S313E mutant suggested that PACSIN2 phosphorylation was sufficient to reduce caveolar-tracking durations. Both hypotonic treatment and isotonic drug-induced PKC activation increased PACSIN2 phosphorylation at serine 313 and shortened caveolar-tracking durations. Caveolar-tracking durations were also reduced upon the expression of other membrane-binding-deficient PACSIN2 mutants or upon RNA interference (RNAi)-mediated PACSIN2 depletion, pointing to a role for PACSIN2 levels in modulating the lifetime of caveolae. Interestingly, the decrease in membrane-bound PACSIN2 was inversely correlated with the recruitment and activity of dynamin 2, a GTPase that mediates membrane scission. Furthermore, expression of EHD2, which stabilizes caveolae and binds to PACSIN2, restored the tracking durations of cells with reduced PACSIN2 levels. These findings suggest that the PACSIN2 phosphorylation decreases its membrane-binding activity, thereby decreasing its stabilizing effect on caveolae and triggering dynamin-mediated removal of caveolae.
Collapse
Affiliation(s)
- Yosuke Senju
- Laboratory of Membrane and Cytoskeleton Dynamics, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Eva Rosenbaum
- Crystallography, Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Claudio Shah
- Crystallography, Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Sayaka Hamada-Nakahara
- Laboratory of Membrane and Cytoskeleton Dynamics, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Yuzuru Itoh
- Laboratory of Membrane and Cytoskeleton Dynamics, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan
| | - Kimiko Yamamoto
- Laboratory of System Physiology, Department of Biomedical Engineering, Graduate School of Medicine, University of Tokyo, Tokyo 113-0033, Japan
| | - Kyoko Hanawa-Suetsugu
- Laboratory of Membrane and Cytoskeleton Dynamics, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan Laboratory of Molecular Medicine and Cell Biology, Graduate School of Biosciences, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| | - Oliver Daumke
- Crystallography, Max Delbrück Center for Molecular Medicine, Berlin 13125, Germany
| | - Shiro Suetsugu
- Laboratory of Membrane and Cytoskeleton Dynamics, Institute of Molecular and Cellular Biosciences, University of Tokyo, 1-1-1 Yayoi, Bunkyo-ku, Tokyo 113-0032, Japan Laboratory of Molecular Medicine and Cell Biology, Graduate School of Biosciences, Nara Institute of Science and Technology, Ikoma 630-0192, Japan
| |
Collapse
|
37
|
Mougeolle A, Poussard S, Decossas M, Lamaze C, Lambert O, Dargelos E. Oxidative stress induces caveolin 1 degradation and impairs caveolae functions in skeletal muscle cells. PLoS One 2015; 10:e0122654. [PMID: 25799323 PMCID: PMC4370508 DOI: 10.1371/journal.pone.0122654] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2014] [Accepted: 02/21/2015] [Indexed: 11/19/2022] Open
Abstract
Increased level of oxidative stress, a major actor of cellular aging, impairs the regenerative capacity of skeletal muscle and leads to the reduction in the number and size of muscle fibers causing sarcopenia. Caveolin 1 is the major component of caveolae, small membrane invaginations involved in signaling and endocytic trafficking. Their role has recently expanded to mechanosensing and to the regulation of oxidative stress-induced pathways. Here, we increased the amount of reactive oxidative species in myoblasts by addition of hydrogen peroxide (H2O2) at non-toxic concentrations. The expression level of caveolin 1 was significantly decreased as early as 10 min after 500 μM H2O2 treatment. This reduction was not observed in the presence of a proteasome inhibitor, suggesting that caveolin 1 was rapidly degraded by the proteasome. In spite of caveolin 1 decrease, caveolae were still able to assemble at the plasma membrane. Their functions however were significantly perturbed by oxidative stress. Endocytosis of a ceramide analog monitored by flow cytometry was significantly diminished after H2O2 treatment, indicating that oxidative stress impaired its selective internalization via caveolae. The contribution of caveolae to the plasma membrane reservoir has been monitored after osmotic cell swelling. H2O2 treatment increased membrane fragility revealing that treated cells were more sensitive to an acute mechanical stress. Altogether, our results indicate that H2O2 decreased caveolin 1 expression and impaired caveolae functions. These data give new insights on age-related deficiencies in skeletal muscle.
Collapse
Affiliation(s)
- Alexis Mougeolle
- Univ Bordeaux, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; CNRS, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; Bordeaux INP, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France
| | - Sylvie Poussard
- Univ Bordeaux, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; CNRS, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; Bordeaux INP, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France
| | - Marion Decossas
- Univ Bordeaux, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; CNRS, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; Bordeaux INP, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France
| | - Christophe Lamaze
- Institut Curie—Centre de Recherche, Membrane Dynamics and Mechanics of Intracellular Signaling Team, INSERM U1143, CNRS UMR 3666, Paris, France
| | - Olivier Lambert
- Univ Bordeaux, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; CNRS, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; Bordeaux INP, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France
| | - Elise Dargelos
- Univ Bordeaux, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; CNRS, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France; Bordeaux INP, Chimie et Biologie des Membranes et Nanoobjets, UMR 5248, F-33600 Pessac, France
- * E-mail:
| |
Collapse
|
38
|
Hirata H, Tatsumi H, Hayakawa K, Sokabe M. Non-channel mechanosensors working at focal adhesion-stress fiber complex. Pflugers Arch 2014; 467:141-55. [PMID: 24965068 DOI: 10.1007/s00424-014-1558-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Revised: 06/10/2014] [Accepted: 06/12/2014] [Indexed: 01/05/2023]
Abstract
Mechanosensitive ion channels (MSCs) have long been the only established molecular class of cell mechanosensors; however, in the last decade, a variety of non-channel type mechanosensor molecules have been identified. Many of them are focal adhesion-associated proteins that include integrin, talin, and actin. Mechanosensors must be non-soluble molecules firmly interacting with relatively rigid cellular structures such as membranes (in terms of lateral stiffness), cytoskeletons, and adhesion structures. The partner of MSCs is the membrane in which MSC proteins efficiently transduce changes in the membrane tension into conformational changes that lead to channel opening. By contrast, the integrin, talin, and actin filament form a linear complex of which both ends are typically anchored to the extracellular matrices via integrins. Upon cell deformation by forces, this structure turns out to be a portion that efficiently transduces the generated stress into conformational changes of composite molecules, leading to the activation of integrin (catch bond with extracellular matrices) and talin (unfolding to induce vinculin bindings). Importantly, this structure also serves as an "active" mechanosensor to detect substrate rigidity by pulling the substrate with contraction of actin stress fibers (SFs), which may induce talin unfolding and an activation of MSCs in the vicinity of integrins. A recent study demonstrates that the actin filament acts as a mechanosensor with unique characteristics; the filament behaves as a negative tension sensor in which increased torsional fluctuations by tension decrease accelerate ADF/cofilin binding, leading to filament disruption. Here, we review the latest progress in the study of those non-channel mechanosensors and discuss their activation mechanisms and physiological roles.
Collapse
Affiliation(s)
- Hiroaki Hirata
- Mechanobiology Institute, National University of Singapore, 117411, Singapore, Singapore
| | | | | | | |
Collapse
|
39
|
Involvement of microtubular network and its motors in productive endocytic trafficking of mouse polyomavirus. PLoS One 2014; 9:e96922. [PMID: 24810588 PMCID: PMC4014599 DOI: 10.1371/journal.pone.0096922] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2013] [Accepted: 04/14/2014] [Indexed: 12/18/2022] Open
Abstract
Infection of non-enveloped polyomaviruses depends on an intact microtubular network. Here we focus on mouse polyomavirus (MPyV). We show that the dynamics of MPyV cytoplasmic transport reflects the characteristics of microtubular motor-driven transport with bi-directional saltatory movements. In cells treated with microtubule-disrupting agents, localization of MPyV was significantly perturbed, the virus was retained at the cell periphery, mostly within membrane structures resembling multicaveolar complexes, and at later times post-infection, only a fraction of the virus was found in Rab7-positive endosomes and multivesicular bodies. Inhibition of cytoplasmic dynein-based motility by overexpression of dynamitin affected perinuclear translocation of the virus, delivery of virions to the ER and substantially reduced the numbers of infected cells, while overexpression of dominant-negative form of kinesin-1 or kinesin-2 had no significant impact on virus localization and infectivity. We also found that transport along microtubules was important for MPyV-containing endosome sequential acquisition of Rab5, Rab7 and Rab11 GTPases. However, in contrast to dominant-negative mutant of Rab7 (T22N), overexpression of dominant-negative mutant Rab11 (S25N) did not affect the virus infectivity. Altogether, our study revealed that MPyV cytoplasmic trafficking leading to productive infection bypasses recycling endosomes, does not require the function of kinesin-1 and kinesin-2, but depends on functional dynein-mediated transport along microtubules for translocation of the virions from peripheral, often caveolin-positive compartments to late endosomes and ER – a prerequisite for efficient delivery of the viral genome to the nucleus.
Collapse
|
40
|
Cambi A, Lakadamyali M, Lidke DS, Garcia-Parajo MF. Meeting report--Visualizing signaling nanoplatforms at a higher spatiotemporal resolution. J Cell Sci 2014; 126:3817-21. [PMID: 23995382 DOI: 10.1242/jcs.137901] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The International Symposium entitled ‘Visualizing signaling nanoplatforms at a higher spatiotemporal resolution’ sponsored by the Institució Catalana de Recerca i Estudis Avançats (ICREA) was held on 29–31 May 2013 at the ICFO-Institute of Photonic Sciences, in Barcelona, Spain. The meeting brought together a multidisciplinary group of international leaders in the fields of super-resolution imaging (nanoscopy) and cell membrane biology, and served as a forum to further our understanding of the fundamental mechanisms that govern nanostructures and protein–function relationships at the cell membrane.
Collapse
Affiliation(s)
- Alessandra Cambi
- Department of Tumor Immunology, Nijmegen Centre for Molecular Life Sciences, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands
| | | | | | | |
Collapse
|
41
|
Bogdan S, Schultz J, Grosshans J. Formin' cellular structures: Physiological roles of Diaphanous (Dia) in actin dynamics. Commun Integr Biol 2014; 6:e27634. [PMID: 24719676 PMCID: PMC3977921 DOI: 10.4161/cib.27634] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Revised: 12/21/2013] [Accepted: 12/23/2013] [Indexed: 01/06/2023] Open
Abstract
Members of the Diaphanous (Dia) protein family are key regulators of fundamental actin driven cellular processes, which are conserved from yeast to humans. Researchers have uncovered diverse physiological roles in cell morphology, cell motility, cell polarity, and cell division, which are involved in shaping cells into tissues and organs. The identification of numerous binding partners led to substantial progress in our understanding of the differential functions of Dia proteins. Genetic approaches and new microscopy techniques allow important new insights into their localization, activity, and molecular principles of regulation.
Collapse
Affiliation(s)
- Sven Bogdan
- Institut für Neurobiologie; Universität Münster; Münster, Germany
| | - Jörg Schultz
- Bioinformatik, Biozentrum; Universität Würzburg; Würzburg, Germany
| | - Jörg Grosshans
- Institut für Biochemie; Universitätsmedizin; Universität Göttingen; Göttingen, Germany
| |
Collapse
|
42
|
Abstract
Morphogenesis is the remarkable process by which cells self-assemble into complex tissues and organs that exhibit specialized form and function during embryological development. Many of the genes and chemical cues that mediate tissue and organ formation have been identified; however, these signals alone are not sufficient to explain how tissues and organs are constructed that exhibit their unique material properties and three-dimensional forms. Here, we review work that has revealed the central role that physical forces and extracellular matrix mechanics play in the control of cell fate switching, pattern formation, and tissue development in the embryo and how these same mechanical signals contribute to tissue homeostasis and developmental control throughout adult life.
Collapse
Affiliation(s)
- Tadanori Mammoto
- Vascular Biology Program, Boston Children's Hospital and Harvard Medical School, Boston, Massachusetts 02115;
| | | | | |
Collapse
|
43
|
Schroeter MR, Leifheit-Nestler M, Hubert A, Schumann B, Glückermann R, Eschholz N, Krüger N, Lutz S, Hasenfuss G, Konstantinides S, Schäfer K. Leptin promotes neointima formation and smooth muscle cell proliferation via NADPH oxidase activation and signalling in caveolin-rich microdomains. Cardiovasc Res 2013; 99:555-65. [PMID: 23723060 DOI: 10.1093/cvr/cvt126] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
AIMS Apolipoprotein E (apoE) may act as a vasculoprotective factor by promoting plasma lipid clearance and cholesterol efflux. Moreover, apoE accumulates at sites of vascular injury and modulates the effect of growth factors on smooth muscle cells (SMCs). Experimental data suggested that hypothalamic apoE expression is reduced in obesity and associated with leptin resistance. In this study, we examined the role of apoE in mediating the effects of leptin on vascular lesion formation. METHODS AND RESULTS Leptin was administered to apoE knockout (apoE-/-) mice via osmotic pumps to increase its circulating levels. Morphometric analysis revealed that leptin did not alter neointima formation and failed to increase α-actin- or PCNA-immunopositive SMCs after vascular injury. Similar findings were obtained after analysis of atherosclerotic lesions. Comparison of apoE-/-, wild-type, or LDL receptor-/- mice and functional analyses in aortic SMCs from WT or apoE-/- mice or human arterial SMCs after treatment with small interfering (si)RNA or heparinase revealed that leptin requires the presence of apoE, expressed, secreted and bound to the cell surface, to fully activate leptin receptor signalling and to promote SMC proliferation and neointima formation. Mechanistically, leptin induced the phosphorylation and membrane translocation of caveolin (cav)-1, and apoE down-regulation or caveolae disruption inhibited the leptin-induced p47phox activation, ROS formation and SMC proliferation. Finally, leptin failed to increase neointima formation in mice lacking cav-1. CONCLUSION Our findings suggest that apoE mediates the effects of leptin on vascular lesion formation by stabilizing cav-1-enriched cell membrane microdomains in SMCs, thus allowing NADPH oxidase assembly and ROS-mediated mitogenic signalling.
Collapse
Affiliation(s)
- Marco R Schroeter
- Department of Cardiology and Pulmonary Medicine, University Medical Center Göttingen, Robert Koch Strasse 40, Göttingen D-37075, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ben-Dov N, Korenstein R. Actin-cytoskeleton rearrangement modulates proton-induced uptake. Exp Cell Res 2013; 319:946-54. [DOI: 10.1016/j.yexcr.2013.01.017] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2012] [Revised: 01/21/2013] [Accepted: 01/22/2013] [Indexed: 10/27/2022]
|
45
|
Parton RG, del Pozo MA. Caveolae as plasma membrane sensors, protectors and organizers. Nat Rev Mol Cell Biol 2013; 14:98-112. [PMID: 23340574 DOI: 10.1038/nrm3512] [Citation(s) in RCA: 676] [Impact Index Per Article: 56.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Caveolae are submicroscopic, plasma membrane pits that are abundant in many mammalian cell types. The past few years have seen a quantum leap in our understanding of the formation, dynamics and functions of these enigmatic structures. Caveolae have now emerged as vital plasma membrane sensors that can respond to plasma membrane stresses and remodel the extracellular environment. Caveolae at the plasma membrane can be removed by endocytosis to regulate their surface density or can be disassembled and their structural components degraded. Coat proteins, called cavins, work together with caveolins to regulate the formation of caveolae but also have the potential to dynamically transmit signals that originate in caveolae to various cellular destinations. The importance of caveolae as protective elements in the plasma membrane, and as membrane organizers and sensors, is highlighted by links between caveolae dysfunction and human diseases, including muscular dystrophies and cancer.
Collapse
Affiliation(s)
- Robert G Parton
- Institute for Molecular Bioscience and Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, QLD 4072, Australia.
| | | |
Collapse
|
46
|
Nassoy P, Lamaze C. Stressing caveolae new role in cell mechanics. Trends Cell Biol 2012; 22:381-9. [PMID: 22613354 DOI: 10.1016/j.tcb.2012.04.007] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2012] [Revised: 04/16/2012] [Accepted: 04/18/2012] [Indexed: 01/22/2023]
Abstract
It has been almost 60 years since caveolae were first visualized by Eichi Yamada and George Palade. Nevertheless, these specialized invaginations of the plasma membrane remain without clear and recognized physiological function. The recent identification of new caveolar components and the ability to probe cell mechanics with sophisticated opticophysical devices have shed new light on this fascinating organelle. Early studies from the 1970s suggested that caveolae could participate in the regulation of membrane dynamics. Recent data have established caveolae as mechanosensors that respond immediately to mechanical stress by flattening into the plasma membrane. Here, we focus on the molecular consequences that result from the caveolar disassembly/reassembly cycle induced by membrane tension variations at the surface of the cell under physiological and pathological conditions.
Collapse
Affiliation(s)
- Pierre Nassoy
- Université P. et M. Curie/CNRS UMR168, 26 rue d'Ulm, 75248 Paris Cedex 05, France
| | | |
Collapse
|
47
|
de Curtis I, Meldolesi J. Cell surface dynamics – how Rho GTPases orchestrate the interplay between the plasma membrane and the cortical cytoskeleton. J Cell Sci 2012; 125:4435-44. [DOI: 10.1242/jcs.108266] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Small GTPases are known to regulate hundreds of cell functions. In particular, Rho family GTPases are master regulators of the cytoskeleton. By regulating actin nucleation complexes, Rho GTPases control changes in cell shape, including the extension and/or retraction of surface protrusions and invaginations. Protrusion and invagination of the plasma membrane also involves the interaction between the plasma membrane and the cortical cytoskeleton. This interplay between membranes and the cytoskeleton can lead to an increase or decrease in the plasma membrane surface area and its tension as a result of the fusion (exocytosis) or internalization (endocytosis) of membranous compartments, respectively. For a long time, the cytoskeleton and plasma membrane dynamics were investigated separately. However, studies from many laboratories have now revealed that Rho GTPases, their modulation of the cytoskeleton, and membrane traffic are closely connected during the dynamic remodeling of the cell surface. Arf- and Rab-dependent exocytosis of specific vesicles contributes to the targeting of Rho GTPases and their regulatory factors to discrete sites of the plasma membrane. Rho GTPases regulate the tethering of exocytic vesicles and modulate their subsequent fusion. They also have crucial roles in the different forms of endocytosis, where they participate in the sorting of membrane domains as well as the sculpting and sealing of membrane flasks and cups. Here, we discuss how cell surface dynamics depend on the orchestration of the cytoskeleton and the plasma membrane by Rho GTPases.
Collapse
|