51
|
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
|
52
|
Cavin-1: caveolae-dependent signalling and cardiovascular disease. Biochem Soc Trans 2015; 42:284-8. [PMID: 24646232 DOI: 10.1042/bst20130270] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Caveolae are curved lipid raft regions rich in cholesterol and sphingolipids found abundantly in vascular endothelial cells, adipocytes, smooth muscle cells and fibroblasts. They are multifunctional organelles with roles in clathrin-independent endocytosis, cholesterol transport, mechanosensing and signal transduction. Caveolae provide an environment where multiple receptor signalling components are sequestered, clustered and compartmentalized for efficient signal transduction. Many of these receptors, including cytokine signal transducer gp130 (glycoprotein 130), are mediators of chronic inflammation during atherogenesis. Subsequently, disruption of these organelles is associated with a broad range of disease states including cardiovascular disease and cancer. Cavin-1 is an essential peripheral component of caveolae that stabilizes caveolin-1, the main structural/integral membrane protein of caveolae. Caveolin-1 is an essential regulator of eNOS (endothelial nitric oxide synthase) and its disruption leads to endothelial dysfunction which initiates a range of cardiovascular and pulmonary disorders. Although dysfunctional cytokine signalling is also a hallmark of cardiovascular disease, knowledge of caveolae-dependent cytokine signalling is lacking as is the role of cavin-1 independent of caveolae. The present review introduces caveolae, their structural components, the caveolins and cavins, their regulation by cAMP, and their potential role in cardiovascular disease.
Collapse
|
53
|
Delivery of nucleic acids and nanomaterials by cell-penetrating peptides: opportunities and challenges. BIOMED RESEARCH INTERNATIONAL 2015; 2015:834079. [PMID: 25883975 PMCID: PMC4391616 DOI: 10.1155/2015/834079] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 09/18/2014] [Accepted: 09/23/2014] [Indexed: 12/20/2022]
Abstract
Many viral and nonviral systems have been developed to aid delivery of biologically active molecules into cells. Among these, cell-penetrating peptides (CPPs) have received increasing attention in the past two decades for biomedical applications. In this review, we focus on opportunities and challenges associated with CPP delivery of nucleic acids and nanomaterials. We first describe the nature of versatile CPPs and their interactions with various types of cargoes. We then discuss in vivo and in vitro delivery of nucleic acids and nanomaterials by CPPs. Studies on the mechanisms of cellular entry and limitations in the methods used are detailed.
Collapse
|
54
|
Hofmeister LH, Lee SH, Norlander AE, Montaniel KRC, Chen W, Harrison DG, Sung HJ. Phage-display-guided nanocarrier targeting to atheroprone vasculature. ACS NANO 2015; 9:4435-46. [PMID: 25768046 PMCID: PMC4654777 DOI: 10.1021/acsnano.5b01048] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
In regions of the circulation where vessels are straight and unbranched, blood flow is laminar and unidirectional. In contrast, at sites of curvature, branch points, and regions distal to stenoses, blood flow becomes disturbed. Atherosclerosis preferentially develops in these regions of disturbed blood flow. Current therapies for atherosclerosis are systemic and may not sufficiently target these atheroprone regions. In this study, we sought to leverage the alterations on the luminal surface of endothelial cells caused by this atheroprone flow for nanocarrier targeting. In vivo phage display was used to discover unique peptides that selectively bind to atheroprone regions in the mouse partial carotid artery ligation model. The peptide GSPREYTSYMPH (PREY) was found to bind 4.5-fold more avidly to the region of disturbed flow and was used to form targeted liposomes. When administered intravenously, PREY-targeted liposomes preferentially accumulated in endothelial cells in the partially occluded carotid artery and other areas of disturbed flow. Proteomic analysis and immunoblotting indicated that fibronectin and Filamin-A were preferentially bound by PREY nanocarriers in vessels with disturbed flow. In additional experiments, PREY nanocarriers were used therapeutically to deliver the nitric oxide synthase cofactor tetrahydrobiopterin (BH4), which we have previously shown to be deficient in regions of disturbed flow. This intervention increased vascular BH4 and reduced vascular superoxide in the partially ligated artery in wild-type mice and reduced plaque burden in the partially ligated left carotid artery of fat fed atheroprone mice (ApoE(-/-)). Targeting atheroprone sites of the circulation with functionalized nanocarriers provides a promising approach for prevention of early atherosclerotic lesion formation.
Collapse
Affiliation(s)
- Lucas H. Hofmeister
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235
| | - Sue H. Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235
| | | | | | - Wei Chen
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, 37235
| | - David G. Harrison
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235
- Division of Clinical Pharmacology, Vanderbilt University, Nashville, TN, 37235
- Center for Vascular Biology, Vanderbilt University, Nashville, TN, 37235
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, 37235
| | - Hak-Joon Sung
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235
- Division of Cardiovascular Medicine, Vanderbilt University, Nashville, TN, 37235
| |
Collapse
|
55
|
Sanon VP, Sawaki D, Mjaatvedt CH, Jourdan‐Le Saux C. Myocardial Tissue Caveolae. Compr Physiol 2015; 5:871-86. [DOI: 10.1002/cphy.c140050] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
56
|
Trincone A, Schwegmann-Weßels C. Looking for a needle in a haystack: Cellular proteins that may interact with the tyrosine-based sorting signal of the TGEV S protein. Virus Res 2014; 202:3-11. [PMID: 25481285 PMCID: PMC7114463 DOI: 10.1016/j.virusres.2014.11.029] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2014] [Revised: 11/23/2014] [Accepted: 11/26/2014] [Indexed: 11/24/2022]
Abstract
The spike protein S of transmissible gastroenteritis virus, an Alphacoronavirus, contains a tyrosine-based sorting signal that is responsible for ERGIC retention and may be important for a correct viral assembly process. To find out whether the S protein interacts with cellular proteins via this sorting signal, a pulldown assay with GST fusion proteins was performed. Filamin A has been identified as a putative interaction candidate. Immunofluorescence assays confirmed a co-localization between the TGEV S protein and filamin A. Further experiments have to be performed to prove a significant impact of filamin A on TGEV infection. Different approaches of several researchers for the identification of cellular interaction candidates relevant for coronavirus replication are summarized. These results may help in the future to identify the role of cellular proteins during coronavirus assembly at the ER-Golgi intermediate compartment.
Collapse
Affiliation(s)
- Anna Trincone
- Institute for Virology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany
| | - Christel Schwegmann-Weßels
- Institute for Virology, Department of Infectious Diseases, University of Veterinary Medicine Hannover, Bünteweg 17, 30559 Hannover, Germany.
| |
Collapse
|
57
|
Abstract
Intracellular trafficking of viruses and proteins commonly occurs via the early endosome in a process involving Rab5. The RNA Import Complex (RIC)-RNA complex is taken up by mammalian cells and targeted to mitochondria. Through RNA interference, it was shown that mito-targeting of the ribonucleoprotein (RNP) was dependent on caveolin 1 (Cav1), dynamin 2, Filamin A and NSF. Although a minor fraction of the RNP was transported to endosomes in a Rab5-dependent manner, mito-targeting was independent of Rab5 or other endosomal proteins, suggesting that endosomal uptake and mito-targeting occur independently. Sequential immunoprecipitation of the cytosolic vesicles showed the sorting of the RNP away from Cav1 in a process that was independent of the endosomal effector EEA1 but sensitive to nocodazole. However, the RNP was in two types of vesicle with or without Cav1, with membrane-bound, asymmetrically orientated RIC and entrapped RNA, but no endosomal components, suggesting vesicular sorting rather than escape of free RNP from endosomes. In vitro, RNP was directly transferred from the Type 2 vesicles to mitochondria. Live-cell imaging captured spherical Cav1− RNP vesicles emerging from the fission of large Cav+ particles. Thus, RNP appears to traffic by a different route than the classical Rab5-dependent pathway of viral transport.
Collapse
Affiliation(s)
- Joyita Mukherjee
- Genetic Engineering Laboratory, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Calcutta 700032, India Present address: Penn Institute for Regenerative Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Biraj Mahato
- Present address: Penn Institute for Regenerative Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA
| | - Samit Adhya
- Genetic Engineering Laboratory, CSIR-Indian Institute of Chemical Biology, 4 Raja S. C. Mullick Road, Calcutta 700032, India Present address: Penn Institute for Regenerative Medicine, University of Pennsylvania, 3800 Spruce Street, Philadelphia, PA 19104, USA.
| |
Collapse
|
58
|
Adachi-Hayama M, Adachi A, Shinozaki N, Matsutani T, Hiwasa T, Takiguchi M, Saeki N, Iwadate Y. Circulating anti-filamin C autoantibody as a potential serum biomarker for low-grade gliomas. BMC Cancer 2014; 14:452. [PMID: 24946857 PMCID: PMC4094678 DOI: 10.1186/1471-2407-14-452] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2013] [Accepted: 06/05/2014] [Indexed: 11/21/2022] Open
Abstract
Background Glioma is the most common primary malignant central nervous system tumor in adult, and is usually not curable due to its invasive nature. Establishment of serum biomarkers for glioma would be beneficial both for early diagnosis and adequate therapeutic intervention. Filamins are an actin cross-linker and filamin C (FLNC), normally restricted in muscle tissues, offers many signaling molecules an essential communication fields. Recently, filamins have been considered important for tumorigenesis in cancers. Methods We searched for novel glioma-associated antigens by serological identification of antigens utilizing recombinant cDNA expression cloning (SEREX), and found FLNC as a candidate protein. Tissue expressions of FLNC (both in normal and tumor tissues) were examined by immunohistochemistry and quantitative RT-PCR analyses. Serum anti-FLNC autoantibody level was measured by ELISA in normal volunteers and in the patients with various grade gliomas. Results FLNC was expressed in glioma tissues and its level got higher as tumor grade advanced. Anti-FLNC autoantibody was also detected in the serum of glioma patients, but its levels were inversely correlated with the tissue expression. Serum anti-FLNC autoantibody level was significantly higher in low-grade glioma patients than in high-grade glioma patients or in normal volunteers, which was confirmed in an independent validation set of patients’ sera. The autoantibody levels in the patients with meningioma or cerebral infarction were at the same level of normal volunteers, and they were significantly lower than that of low-grade gliomas. Total IgG and anti-glutatione S-transferase (GST) antibody level were not altered among the patient groups, which suggest that the autoantibody response was specific for FLNC. Conclusions The present results suggest that serum anti-FLNC autoantibody can be a potential serum biomarker for early diagnosis of low-grade gliomas while it needs a large-scale clinical study.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Yasuo Iwadate
- Department of Neurological Surgery, Chiba University, Graduate School of Medicine, 1-8-1, Inohana, Chuo-ku, Chiba 260-8670, Japan.
| |
Collapse
|
59
|
Chaudhary N, Gomez GA, Howes MT, Lo HP, McMahon KA, Rae JA, Schieber NL, Hill MM, Gaus K, Yap AS, Parton RG. Endocytic crosstalk: cavins, caveolins, and caveolae regulate clathrin-independent endocytosis. PLoS Biol 2014; 12:e1001832. [PMID: 24714042 PMCID: PMC3979662 DOI: 10.1371/journal.pbio.1001832] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Accepted: 02/25/2014] [Indexed: 12/18/2022] Open
Abstract
Caveolar proteins and caveolae negatively regulate a second clathrin-independent endocytic CLIC/GEEC pathway; caveolin-1 affects membrane diffusion properties of raft-associated CLIC cargo, and the scaffolding domain of caveolin-1 is required and sufficient for endocytic inhibition. Several studies have suggested crosstalk between different clathrin-independent endocytic pathways. However, the molecular mechanisms and functional relevance of these interactions are unclear. Caveolins and cavins are crucial components of caveolae, specialized microdomains that also constitute an endocytic route. Here we show that specific caveolar proteins are independently acting negative regulators of clathrin-independent endocytosis. Cavin-1 and Cavin-3, but not Cavin-2 or Cavin-4, are potent inhibitors of the clathrin-independent carriers/GPI-AP enriched early endosomal compartment (CLIC/GEEC) endocytic pathway, in a process independent of caveola formation. Caveolin-1 (CAV1) and CAV3 also inhibit the CLIC/GEEC pathway upon over-expression. Expression of caveolar protein leads to reduction in formation of early CLIC/GEEC carriers, as detected by quantitative electron microscopy analysis. Furthermore, the CLIC/GEEC pathway is upregulated in cells lacking CAV1/Cavin-1 or with reduced expression of Cavin-1 and Cavin-3. Inhibition by caveolins can be mimicked by the isolated caveolin scaffolding domain and is associated with perturbed diffusion of lipid microdomain components, as revealed by fluorescence recovery after photobleaching (FRAP) studies. In the absence of cavins (and caveolae) CAV1 is itself endocytosed preferentially through the CLIC/GEEC pathway, but the pathway loses polarization and sorting attributes with consequences for membrane dynamics and endocytic polarization in migrating cells and adult muscle tissue. We also found that noncaveolar Cavin-1 can act as a modulator for the activity of the key regulator of the CLIC/GEEC pathway, Cdc42. This work provides new insights into the regulation of noncaveolar clathrin-independent endocytosis by specific caveolar proteins, illustrating multiple levels of crosstalk between these pathways. We show for the first time a role for specific cavins in regulating the CLIC/GEEC pathway, provide a new tool to study this pathway, identify caveola-independent functions of the cavins and propose a novel mechanism for inhibition of the CLIC/GEEC pathway by caveolin. Endocytosis is the process that allows cells to take up molecules from the environment. Several endocytic pathways exist in mammalian cells. While the best understood endocytic pathway uses clathrin, recent years have seen a great increase in our understanding of clathrin-independent endocytic pathways. Here we characterize the crosstalk between caveolae, flask-shaped specialized microdomains present at the plasma membrane, and a second clathrin-independent pathway, the CLIC/GEEC Cdc42-regulated endocytic pathway. These pathways are segregated in migrating cells with caveolae at the rear and CLIC/GEEC endocytosis at the leading edge. Here we find that specific caveolar proteins, caveolins and cavins, can also negatively regulate the CLIC/GEEC pathway. With the help of several techniques, including quantitative electron microscopy analysis and real-time live-cell imaging, we demonstrate that expression of caveolar proteins affects early carrier formation, causes cellular lipid changes, and changes the activity of the key regulator of the CLIC/GEEC pathway, Cdc42. The functional consequences of loss of caveolar proteins on the CLIC/GEEC pathway included inhibition of polarized cell migration and increased endocytosis in tissue explants.
Collapse
Affiliation(s)
- Natasha Chaudhary
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Guillermo A. Gomez
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Mark T. Howes
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Harriet P. Lo
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Kerrie-Ann McMahon
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - James A. Rae
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Nicole L. Schieber
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Michelle M. Hill
- The University of Queensland, Diamantina Institute, Queensland, Australia
| | - Katharina Gaus
- The University of New South Wales, Centre for Vascular Research and Australian Centre for Nanomedicine, New South Wales, Australia
| | - Alpha S. Yap
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
| | - Robert G. Parton
- The University of Queensland, Institute for Molecular Bioscience, Queensland, Australia
- Centre for Microscopy and Microanalysis, Queensland, Australia
- * E-mail:
| |
Collapse
|
60
|
Abstract
Neural proliferation, migration and differentiation require reorganization of the actin cytoskeleton and regulation of vesicle trafficking to provide stability in maintaining cell adhesions, allow for changes in cell shape, and establishing cell polarity. Human disorders involving the actin-binding Filamin A (FLNA) and vesicle trafficking Brefeldin-associated guanine exchange factor 2 (BIG2 is encoded by the ARFGEF2 gene) proteins are implicated in these various developmental processes, resulting in a malformation of cortical development called periventricular heterotopia (nodules along the ventricular lining) and microcephaly (small brain). Here we discuss several recent reports from our laboratory that demonstrate a shared role for both proteins in actin-associated vesicle trafficking, which is required to maintain the expression and stability of cell adhesion and cell cycle associated molecules during cortical development. While changes in FLNA and BIG2 have first been linked to disorders involving the central nervous system, increasing reports suggest they are associated with aberrant development of various other organ systems in the body. These studies suggest that vesicle trafficking defects in FLN-GEF dependent pathways may contribute to a much broader phenotype than previously realized.
Collapse
Affiliation(s)
- Volney L Sheen
- Department of Neurology; Beth Israel Deaconess Medical Center and Harvard Medical School; Boston, MA USA
| |
Collapse
|
61
|
Hackett TL, de Bruin HG, Shaheen F, van den Berge M, van Oosterhout AJ, Postma DS, Heijink IH. Caveolin-1 controls airway epithelial barrier function. Implications for asthma. Am J Respir Cell Mol Biol 2014; 49:662-71. [PMID: 23742006 DOI: 10.1165/rcmb.2013-0124oc] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The molecular basis for airway epithelial fragility in asthma has remained unclear. We investigated whether the loss of caveolin-1, the major component of caveolae and a known stabilizer of adherens junctions, contributes to epithelial barrier dysfunction in asthma. We studied the expression of caveolin-1 and adhesion molecules E-cadherin and β-catenin in airway sections, and we cultured bronchial epithelial cells from patients with asthma and from healthy control subjects. To determine the functional role of caveolin-1, we investigated the effects of caveolin-1 up-regulation and down-regulation on E-cadherin expression, barrier function, and proallergic activity in the human bronchial epithelial cell lines 16HBE and BEAS-2B. The membrane expression of caveolin-1 was significantly lower in airway epithelia from patients with asthma than from subjects without asthma, and this lower expression was maintained in vitro upon air-liquid interface and submerged culturing. Importantly, reduced caveolin-1 expression was accompanied by a loss of junctional E-cadherin and β-catenin expression, disrupted epithelial barrier function, and increased levels of the proallergic cytokine thymic stromal lymphopoietin (TSLP). Furthermore, E-cadherin redistribution upon exposure to epidermal growth factor or house dust mite was paralleled by the internalization of caveolin-1 in 16HBE cells. These effects appear to be causally related, because the short, interfering RNA down-regulation of caveolin-1 resulted in the delocalization of E-cadherin and barrier dysfunction in 16HBE cells. Moreover, caveolin-1 overexpression improved barrier function and reduced TSLP expression in BEAS-2B cells. Together, our data demonstrate a crucial role for caveolin-1 in epithelial cell-cell adhesion, with important consequences for epithelial barrier function and the promotion of Th2 responses in asthma.
Collapse
Affiliation(s)
- Tillie-Louise Hackett
- 1 University of British Columbia James Hogg Research Centre, Heart and Lung Institute, St. Paul's Hospital, Vancouver, British Columbia, Canada; and
| | | | | | | | | | | | | |
Collapse
|
62
|
Lei L, Lu S, Wang Y, Kim T, Mehta D, Wang Y. The role of mechanical tension on lipid raft dependent PDGF-induced TRPC6 activation. Biomaterials 2014; 35:2868-77. [PMID: 24397990 DOI: 10.1016/j.biomaterials.2013.12.030] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 12/13/2013] [Indexed: 12/22/2022]
Abstract
Canonical transient receptor potential channel 6 (TRPC6) can play an important role in governing how cells perceive the surrounding material environment and regulate Ca(2+) signaling. We have designed a TRPC6 reporter based on fluorescence resonance energy transfer (FRET) to visualize the TRPC6-mediated calcium entry and hence TRPC6 activity in live cells with high spatiotemporal resolutions. In mouse embryonic fibroblasts (MEFs), platelet-derived growth factor BB (PDGF) can activate the TRPC6 reporter, mediated by phospholipase C (PLC). This TRPC6 activation occurred mainly at lipid rafts regions of the plasma membrane because disruption of lipid raft/caveolae by methyl-β-cyclodextrin (MβCD) or the expression of dominant-negative caveolin-1 inhibited the TRPC6 activity. Culturing cells on soft materials or releasing the intracellular tension by ML-7 reduced this PDGF-induced activation of TRPC6 without affecting the PDGF-regulated Src or inositol 1,4,5-trisphosphate (IP3) receptor function, suggesting a specific role of mechanical tension in regulating TRPC6. We further showed that the release of intracellular tension had similar effect on the diffusion coefficients of TRPC6 and a raft marker, confirming a strong coupling between TRPC6 and lipid rafts. Therefore, our results suggest that the TRPC6 activation mainly occurs at lipid rafts, which is regulated by the mechanical cues of surrounding materials.
Collapse
Affiliation(s)
- Lei Lei
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States; Department of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Shaoying Lu
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States; Department of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, United States
| | - Yi Wang
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States
| | - Taejin Kim
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States
| | - Dolly Mehta
- Department of Pharmacology, College of Medicine, University of Illinois, Chicago, IL 60612, United States
| | - Yingxiao Wang
- Department of Bioengineering & Beckman Institute for Advanced Science and Technology, University of Illinois, Urbana-Champaign, Urbana, IL 61801, United States; Department of Bioengineering & Institute of Engineering in Medicine, University of California, San Diego, La Jolla, CA 92093, United States.
| |
Collapse
|
63
|
Kapus A, Janmey P. Plasma membrane--cortical cytoskeleton interactions: a cell biology approach with biophysical considerations. Compr Physiol 2013; 3:1231-81. [PMID: 23897686 DOI: 10.1002/cphy.c120015] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
From a biophysical standpoint, the interface between the cell membrane and the cytoskeleton is an intriguing site where a "two-dimensional fluid" interacts with an exceedingly complex three-dimensional protein meshwork. The membrane is a key regulator of the cytoskeleton, which not only provides docking sites for cytoskeletal elements through transmembrane proteins, lipid binding-based, and electrostatic interactions, but also serves as the source of the signaling events and molecules that control cytoskeletal organization and remolding. Conversely, the cytoskeleton is a key determinant of the biophysical and biochemical properties of the membrane, including its shape, tension, movement, composition, as well as the mobility, partitioning, and recycling of its constituents. From a cell biological standpoint, the membrane-cytoskeleton interplay underlies--as a central executor and/or regulator--a multitude of complex processes including chemical and mechanical signal transduction, motility/migration, endo-/exo-/phagocytosis, and other forms of membrane traffic, cell-cell, and cell-matrix adhesion. The aim of this article is to provide an overview of the tight structural and functional coupling between the membrane and the cytoskeleton. As biophysical approaches, both theoretical and experimental, proved to be instrumental for our understanding of the membrane/cytoskeleton interplay, this review will "oscillate" between the cell biological phenomena and the corresponding biophysical principles and considerations. After describing the types of connections between the membrane and the cytoskeleton, we will focus on a few key physical parameters and processes (force generation, curvature, tension, and surface charge) and will discuss how these contribute to a variety of fundamental cell biological functions.
Collapse
Affiliation(s)
- András Kapus
- Keenan Research Center, Li Ka Shing Knowledge Institute, St. Michael's Hospital and Department of Surgery, University of Toronto, Ontario, Canada.
| | | |
Collapse
|
64
|
High-fat diet alters protein composition of detergent-resistant membrane microdomains. Cell Tissue Res 2013; 354:771-81. [DOI: 10.1007/s00441-013-1697-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/03/2013] [Indexed: 12/31/2022]
|
65
|
Senetta R, Stella G, Pozzi E, Sturli N, Massi D, Cassoni P. Caveolin-1 as a promoter of tumour spreading: when, how, where and why. J Cell Mol Med 2013; 17:325-36. [PMID: 23521716 PMCID: PMC3823014 DOI: 10.1111/jcmm.12030] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2012] [Accepted: 01/15/2013] [Indexed: 11/29/2022] Open
Abstract
Caveolae are non-clathrin invaginations of the plasma membrane in most cell types; they are involved in signalling functions and molecule trafficking, thus modulating several biological functions, including cell growth, apoptosis and angiogenesis. The major structural protein in caveolae is caveolin-1, which is known to act as a key regulator in cancer onset and progression through its role as a tumour suppressor. Caveolin-1 can also promote cell proliferation, survival and metastasis as well as chemo- and radioresistance. Here, we discuss recent findings and novel concepts that support a role for caveolin-1 in cancer development and its distant spreading. We also address the potential application of caveolin-1 in tumour therapy and diagnosis.
Collapse
Affiliation(s)
- Rebecca Senetta
- Department of Medical Sciences, University of Turin, Turin, Italy
| | | | | | | | | | | |
Collapse
|
66
|
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: 664] [Impact Index Per Article: 55.3] [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
|
67
|
Yue J, Huhn S, Shen Z. Complex roles of filamin-A mediated cytoskeleton network in cancer progression. Cell Biosci 2013; 3:7. [PMID: 23388158 PMCID: PMC3573937 DOI: 10.1186/2045-3701-3-7] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2012] [Accepted: 01/10/2013] [Indexed: 01/08/2023] Open
Abstract
Filamin-A (FLNA), also called actin-binding protein 280 (ABP-280), was originally identified as a non-muscle actin binding protein, which organizes filamentous actin into orthogonal networks and stress fibers. Filamin-A also anchors various transmembrane proteins to the actin cytoskeleton and provides a scaffold for a wide range of cytoplasmic and nuclear signaling proteins. Intriguingly, several studies have revealed that filamin-A associates with multiple non-cytoskeletal proteins of diverse function and is involved in several unrelated pathways. Mutations and aberrant expression of filamin-A have been reported in human genetic diseases and several types of cancer. In this review, we discuss the implications of filamin-A in cancer progression, including metastasis and DNA damage response.
Collapse
Affiliation(s)
- Jingyin Yue
- Department of Radiation Oncology, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ, 08903, USA.
| | | | | |
Collapse
|
68
|
Lam BD, Hordijk PL. The Rac1 hypervariable region in targeting and signaling: a tail of many stories. Small GTPases 2013; 4:78-89. [PMID: 23354415 DOI: 10.4161/sgtp.23310] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cellular signaling by small GTPases is critically dependent on proper spatio-temporal orchestration of activation and output. In addition to their core G (guanine nucleotide binding)-domain, small GTPases comprise a hypervariable region (HVR) and a lipid anchor that are generally accepted to control subcellullar localization. The HVR encodes in many small GTPases a polybasic region (PBR) that permits charge-mediated association to the inner leaflet of the plasma membrane or to intracellular organelles. Over the past 15-20 years, evidence has accumulated for specific protein-protein interactions, mediated by the HVR, that control both targeting and signaling specificity of small GTPases. Using the RhoGTPase Rac1 as a paradigm we here review a series of protein partners that require the Rac1 HVR for association and that control various aspects of localized Rac1 signaling. Some of these proteins represent Rac1 activators, whereas others mediate Rac1 inactivation and degradation and yet others potentiate Rac1 downstream signaling. Finally, evidence is discussed which shows that the HVR of Rac1 also contributes to effector interactions, co-operating with the N-terminal effector domain. The complexity of localized Rac1 signaling, reviewed here, is most likely exemplary for many other small GTPases as well, representing a challenge to identify and define similar mechanisms controlling the specific signaling induced by small GTPases.
Collapse
Affiliation(s)
- B Daniel Lam
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | | |
Collapse
|
69
|
Cis-9,trans-11-conjugated linoleic acid affects lipid raft composition and sensitizes human colorectal adenocarcinoma HT-29 cells to X-radiation. Biochim Biophys Acta Gen Subj 2012; 1830:2233-42. [PMID: 23116821 DOI: 10.1016/j.bbagen.2012.10.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2012] [Revised: 09/29/2012] [Accepted: 10/18/2012] [Indexed: 11/21/2022]
Abstract
BACKGROUND Investigations concerned the mechanism of HT-29 cells radiosensitization by cis-9,trans-11-conjugated linoleic acid (c9,t11-CLA), a natural component of human diet with proven antitumor activity. METHODS The cells were incubated for 24h with 70μM c9,t11-CLA and then X-irradiated. The following methods were used: gas chromatography (incorporation of the CLA isomer), flow cytometry (cell cycle), cloning (survival), Western blotting (protein distribution in membrane fractions), and pulse-field gel electrophoresis (rejoining of DNA double-strand breaks). In parallel, DNA-PK activity, γ-H2AX foci numbers and chromatid fragmentation were estimated. Gene expression was analysed by RT-PCR and chromosomal aberrations by the mFISH method. Nuclear accumulation of the EGF receptor (EGFR) was monitored by ELISA. RESULTS AND CONCLUSIONS C9,t11-CLA sensitized HT-29 cells to X-radiation. This effect was not due to changes in cell cycle progression or DNA-repair-related gene expression. Post-irradiation DSB rejoining was delayed, corresponding with the insufficient DNA-PK activation, although chromosomal aberration frequencies did not increase. Distributions of cholesterol and caveolin-1 in cellular membrane fractions changed. The nuclear EGFR translocation, necessary to increase the DNA-PK activity in response to oxidative stress, was blocked. We suppose that c9,t11-CLA modified the membrane structure, thus disturbing the intracellular EGFR transport and the EGFR-dependent pro-survival signalling, both functionally associated with lipid raft properties. GENERAL SIGNIFICANCE The results point to the importance of the cell membrane interactions with the nucleus after injury inflicted by X -rays. Compounds like c9,t11-CLA, that specifically alter membrane properties, could be used to develop new anticancer strategies.
Collapse
|
70
|
Reglero-Real N, Marcos-Ramiro B, Millán J. Endothelial membrane reorganization during leukocyte extravasation. Cell Mol Life Sci 2012; 69:3079-99. [PMID: 22573182 PMCID: PMC11114893 DOI: 10.1007/s00018-012-0987-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 03/22/2012] [Accepted: 03/29/2012] [Indexed: 12/30/2022]
Abstract
Leukocyte trafficking from the bloodstream to inflamed tissues across the endothelial barrier is an essential response in innate immunity. Leukocyte adhesion, locomotion, and diapedesis induce signaling in endothelial cells and this is accompanied by a profound reorganization of the endothelial cell surfaces that is only starting to be unveiled. Here we review the current knowledge on the leukocyte-mediated alterations of endothelial membrane dynamics and their role in promoting leukocyte extravasation. The formation of protein- and lipid-mediated cell adhesion nanodomains at the endothelial apical surface, the extension of micrometric apical membrane docking structures, which are derived from microvilli and embrace adhered leukocytes, as well as the vesicle-trafficking pathways that are required for efficient leukocyte diapedesis, are discussed. The coordination between these different endothelial membrane-remodeling events probably provides the road map for transmigrating leukocytes to find exit points in the vessel wall, in a context of severe mechanical and inflammatory stress. A better understanding of how vascular endothelial cells respond to immune cell adhesion should enable new therapeutic strategies to be developed that can abrogate uncontrolled leukocyte extravasation in inflammatory diseases.
Collapse
Affiliation(s)
- Natalia Reglero-Real
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Beatriz Marcos-Ramiro
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| | - Jaime Millán
- Centro de Biología Molecular Severo Ochoa, CSIC-Universidad Autónoma de Madrid, C/Nicolás Cabrera 1, Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
71
|
Baltiérrez-Hoyos R, Roa-Espitia AL, Hernández-González EO. The association between CDC42 and caveolin-1 is involved in the regulation of capacitation and acrosome reaction of guinea pig and mouse sperm. Reproduction 2012; 144:123-34. [DOI: 10.1530/rep-11-0433] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In the mammalian sperm, the acrosome reaction (AR) is considered to be a regulated secretion that is an essential requirement for physiological fertilization. The AR is the all-or-nothing secretion system that allows for multiple membrane fusion events. It is a Ca2+-regulated exocytosis reaction that has also been shown to be regulated by several signaling pathways. CDC42 has a central role in the regulated exocytosis through the activation of SNARE proteins and actin polymerization. Furthermore, the lipid raft protein caveolin-1 (CAV1) functions as a scaffold and guanine nucleotide dissociation inhibitor protein for CDC42, which is inactivated when associated with CAV1. CDC42 and other RHO proteins have been shown to localize in the acrosome region of mammalian sperm; however, their relationship with the AR is unknown. Here, we present the first evidence that CDC42 and CAV1 could be involved in the regulation of capacitation and the AR. Our findings show that CDC42 is activated early during capacitation, reaching an activation maximum after 20 min of capacitation. Spontaneous and progesterone-induced ARs were inhibited when sperm were capacitated in presence of secramine A, a specific CDC42 inhibitor. CAV1 and CDC42 were co-immunoprecipitated from the membranes of noncapacitated sperm; this association was reduced in capacitated sperm, and our data suggest that the phosphorylation (Tyr14) of CAV1 by c-Src is involved in such reductions. We suggest that CDC42 activation is favored by the disruption of the CAV1–CDC42 interaction, allowing for its participation in the regulation of capacitation and the AR.
Collapse
|
72
|
Schmitz M, Signore SC, Zerr I, Althaus HH. Oligodendroglial process formation is differentially affected by modulating the intra- and extracellular cholesterol content. J Mol Neurosci 2012; 49:457-69. [PMID: 22740150 PMCID: PMC3566395 DOI: 10.1007/s12031-012-9833-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2012] [Accepted: 06/04/2012] [Indexed: 12/12/2022]
Abstract
Cholesterol is an essential component of eukaryotic plasma membranes and plays an important role in membrane organization and signaling processes. It is the major lipid component of detergent resistant caveolin-1 containing rafts which previously had been reported as a platform for nerve growth factor (NGF) signaling in oligodendrocytes (OL). Surprisingly, a knockdown of caveolin-1 attenuated the process formation of OL (Schmitz et al. J Neurosci Res 88:572–588, 2010), for which a loss of cholesterol could be responsible. In the present report, we could show that a caveolin-1 knockdown resulted in an elevation of cellular cholesterol level; it may indicate an important role of caveolin-1 in cholesterol trafficking to the plasma membrane. Treatment with exogenous PEG cholesterol, which was incorporated to the plasma membrane, supported oligodendroglial process formation, in particular when OL were stimulated by NGF. In this context we have found that OL express NPC1L1 (Niemann–Pick disease type C1-Like 1) which could modulate cholesterol uptake. In contrast, depletion of membrane-bound cholesterol diminished NGF-induced process formation concomitant with a reduced activity of p42/44 mitogen-activated protein kinases.
Collapse
Affiliation(s)
- Matthias Schmitz
- Max-Planck Institute of Experimental Medicine, RU Neural Regeneration, Hermann-Rein-Straße 3, 37075, Goettingen, Germany.
| | | | | | | |
Collapse
|
73
|
Abstract
Caveolins (Cavs) are integrated plasma membrane proteins that are complex signaling regulators with numerous partners and whose activity is highly dependent on cellular context. Cavs are both positive and negative regulators of cell signaling in and/or out of caveolae, invaginated lipid raft domains whose formation is caveolin expression dependent. Caveolins and rafts have been implicated in membrane compartmentalization; proteins and lipids accumulate in these membrane microdomains where they transmit fast, amplified and specific signaling cascades. The concept of plasma membrane organization within functional rafts is still in exploration and sometimes questioned. In this chapter, we discuss the opposing functions of caveolin in cell signaling regulation focusing on the role of caveolin both as a promoter and inhibitor of different signaling pathways and on the impact of membrane domain localization on caveolin functionality in cell proliferation, survival, apoptosis and migration.
Collapse
|
74
|
Stoeber M, Stoeck IK, Hänni C, Bleck CKE, Balistreri G, Helenius A. Oligomers of the ATPase EHD2 confine caveolae to the plasma membrane through association with actin. EMBO J 2012; 31:2350-64. [PMID: 22505029 PMCID: PMC3364743 DOI: 10.1038/emboj.2012.98] [Citation(s) in RCA: 122] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Accepted: 03/23/2012] [Indexed: 12/15/2022] Open
Abstract
Caveolae are plasma membrane microdomains that play important roles in signalling and endocytosis. The ATPase EHD2 shuttles on and off the static population of caveolae in an ATPase cycledependent manner and links caveolae to actin filaments confining them to the plasma membrane.
Caveolae are specialized domains present in the plasma membrane (PM) of most
mammalian cell types. They function in signalling, membrane regulation, and
endocytosis. We found that the Eps-15 homology domain-containing protein 2 (EHD2, an
ATPase) associated with the static population of PM caveolae. Recruitment to the PM
involved ATP binding, interaction with anionic lipids, and oligomerization into
large complexes (60–75S) via interaction of the EH domains with intrinsic
NPF/KPF motifs. Hydrolysis of ATP was essential for binding of EHD2 complexes to
caveolae. EHD2 was found to undergo dynamic exchange at caveolae, a process that
depended on a functional ATPase cycle. Depletion of EHD2 by siRNA or expression of a
dominant-negative mutant dramatically increased the fraction of mobile caveolar
vesicles coming from the PM. Overexpression of EHD2, in turn, caused confinement of
cholera toxin B in caveolae. The confining role of EHD2 relied on its capacity to
link caveolae to actin filaments. Thus, EHD2 likely plays a key role in adjusting
the balance between PM functions of stationary caveolae and the role of caveolae as
vesicular carriers.
Collapse
Affiliation(s)
- Miriam Stoeber
- Institute of Biochemistry, ETH Zurich, Zurich, Switzerland
| | | | | | | | | | | |
Collapse
|
75
|
Echarri A, Muriel O, Pavón DM, Azegrouz H, Escolar F, Terrón MC, Sanchez-Cabo F, Martínez F, Montoya MC, Llorca O, Del Pozo MA. Caveolar domain organization and trafficking is regulated by Abl kinases and mDia1. J Cell Sci 2012; 125:3097-113. [PMID: 22454521 DOI: 10.1242/jcs.090134] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The biology of caveolin-1 (Cav1)/caveolae is intimately linked to actin dynamics and adhesion receptors. Caveolar domains are organized in hierarchical levels of complexity from curved or flattened caveolae to large, higher-order caveolar rosettes. We report that stress fibers controlled by Abl kinases and mDia1 determine the level of caveolar domain organization, which conditions the subsequent inward trafficking of caveolar domains induced upon loss of cell adhesion from the extracellular matrix. Abl-deficient cells have fewer stress fibers, a smaller pool of stress-fiber co-aligned Cav1 and increased clustering of Cav1/caveolae at the cell surface. Defective caveolar linkage to stress fibers prevents the formation of big caveolar rosettes upon loss of cell adhesion, correlating with a lack of inward trafficking. Live imaging of stress fibers and Cav1 showed that the actin-linked Cav1 pool loses its spatial organization in the absence of actin polymerization and is dragged and clustered by depolymerizing filaments. We identified mDia1 as the actin polymerization regulator downstream of Abl kinases that controls the stress-fiber-linked Cav1 pool. mDia1 knockdown results in Cav1/caveolae clustering and defective inward trafficking upon loss of cell adhesion. By contrast, cell elongation imposed by the excess of stress fibers induced by active mDia1 flattens caveolae. Furthermore, active mDia1 rescues the actin co-aligned Cav1 pool and Cav1 inward trafficking upon loss of adhesion in Abl-deficient cells. Thus, caveolar domain organization and trafficking are tightly coupled to adhesive and stress fiber regulatory pathways.
Collapse
Affiliation(s)
- Asier Echarri
- Integrin Signaling Laboratory, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro, 3, 28029, [corrected] Madrid, Spain.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Sigismund S, Confalonieri S, Ciliberto A, Polo S, Scita G, Di Fiore PP. Endocytosis and signaling: cell logistics shape the eukaryotic cell plan. Physiol Rev 2012; 92:273-366. [PMID: 22298658 DOI: 10.1152/physrev.00005.2011] [Citation(s) in RCA: 234] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Our understanding of endocytosis has evolved remarkably in little more than a decade. This is the result not only of advances in our knowledge of its molecular and biological workings, but also of a true paradigm shift in our understanding of what really constitutes endocytosis and of its role in homeostasis. Although endocytosis was initially discovered and studied as a relatively simple process to transport molecules across the plasma membrane, it was subsequently found to be inextricably linked with almost all aspects of cellular signaling. This led to the notion that endocytosis is actually the master organizer of cellular signaling, providing the cell with understandable messages that have been resolved in space and time. In essence, endocytosis provides the communications and supply routes (the logistics) of the cell. Although this may seem revolutionary, it is still likely to be only a small part of the entire story. A wealth of new evidence is uncovering the surprisingly pervasive nature of endocytosis in essentially all aspects of cellular regulation. In addition, many newly discovered functions of endocytic proteins are not immediately interpretable within the classical view of endocytosis. A possible framework, to rationalize all this new knowledge, requires us to "upgrade" our vision of endocytosis. By combining the analysis of biochemical, biological, and evolutionary evidence, we propose herein that endocytosis constitutes one of the major enabling conditions that in the history of life permitted the development of a higher level of organization, leading to the actuation of the eukaryotic cell plan.
Collapse
Affiliation(s)
- Sara Sigismund
- IFOM, Fondazione Istituto FIRC di Oncologia Molecolare, Milan, Italy
| | | | | | | | | | | |
Collapse
|
77
|
Song Y, Xue L, Du S, Sun M, Hu J, Hao L, Gong L, Yeh D, Xiong H, Shao S. Caveolin-1 knockdown is associated with the metastasis and proliferation of human lung cancer cell line NCI-H460. Biomed Pharmacother 2012; 66:439-47. [PMID: 22898083 DOI: 10.1016/j.biopha.2012.03.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2012] [Accepted: 03/01/2012] [Indexed: 12/30/2022] Open
Abstract
Caveolin-1 (CAV-1), one component of caveolae, involves in multiple cellular processes and signal transductions. We previously showed that the expression of CAV-1 gene in NCI-H446 cells inhibited cell proliferation and promoted cell metastasis. Here we explore the function of CAV-1 on tumor growth and metastasis by using NCI-H460 in vitro. First, we established NCI-H460 cell line, which CAV-1 was stably knockdown. Then we investigated the effects of CAV-1 on the morphology, proliferation, cell cycle and metastasis potential for NCI-H460 cell by crystal violet stains, CCK-8, colony formation, flow cytometry, scratch-wound assay and transwell assay. Western blot was used to examine the expression changes of cyclin D1, PCNA, E-cadherin and β-catenin. Our results showed stable knockdown of CAV-1 inhibited the proliferation of NCI-H460 cells. Cell cycle of the transfected cells was arrested in G1/S phase and the expressions of cyclin D1 and PCNA protein were downregulated. Downregulation of CAV-1 promoted the migration and invasion abilities of NCI-H460 cells in vitro. The expression of β-catenin increased and the level of E-cadherin decreased. In summary, our findings provide experimental evidence that CAV-1 may function as a proproliferative and antimetastatic gene in NCI-H460 cell line.
Collapse
Affiliation(s)
- Yang Song
- Department of Histology and Embryology, Dalian Medical University, Dalian, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
78
|
Ayling LJ, Briddon SJ, Halls ML, Hammond GRV, Vaca L, Pacheco J, Hill SJ, Cooper DMF. Adenylyl cyclase AC8 directly controls its micro-environment by recruiting the actin cytoskeleton in a cholesterol-rich milieu. J Cell Sci 2012; 125:869-86. [PMID: 22399809 DOI: 10.1242/jcs.091090] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The central and pervasive influence of cAMP on cellular functions underscores the value of stringent control of the organization of adenylyl cyclases (ACs) in the plasma membrane. Biochemical data suggest that ACs reside in membrane rafts and could compartmentalize intermediary scaffolding proteins and associated regulatory elements. However, little is known about the organization or regulation of the dynamic behaviour of ACs in a cellular context. The present study examines these issues, using confocal image analysis of various AC8 constructs, combined with fluorescence recovery after photobleaching and fluorescence correlation spectroscopy. These studies reveal that AC8, through its N-terminus, enhances the cortical actin signal at the plasma membrane; an interaction that was confirmed by GST pull-down and immunoprecipitation experiments. AC8 also associates dynamically with lipid rafts; the direct association of AC8 with sterols was confirmed in Förster resonance energy transfer experiments. Disruption of the actin cytoskeleton and lipid rafts indicates that AC8 tracks along the cytoskeleton in a cholesterol-enriched domain, and the cAMP that it produces contributes to sculpting the actin cytoskeleton. Thus, an adenylyl cyclase is shown not just to act as a scaffold, but also to actively orchestrate its own micro-environment, by associating with the cytoskeleton and controlling the association by producing cAMP, to yield a highly organized signalling hub.
Collapse
Affiliation(s)
- Laura J Ayling
- Department of Pharmacology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1PD, UK
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Yue J, Lan S, Yuan C, Shen Z. Prognostic values of filamin-A status for topoisomerase II poison chemotherapy. Int J Biol Sci 2012; 8:442-50. [PMID: 22419889 PMCID: PMC3303170 DOI: 10.7150/ijbs.4155] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Accepted: 02/15/2012] [Indexed: 01/04/2023] Open
Abstract
Filamin-A, also called Actin Binding Protein-280, is not only an essential component of the cytoskeleton networks, but also serves as the scaffold in various signaling networks. It has been shown that filamin-A facilitates DNA repair and filamin-A proficient cells are more resistant to ionizing radiation, bleomycin, and cisplatin. In this study, we assessed the role of filamin-A in modulating cancer cell sensitivity to Topo II poisons, including etoposide and doxorubicin. Intriguingly, we found that cells with filamin-A expression are more sensitive to Topo II poisons than those with defective filamin-A, and filamin-A proficient xenograft melanomas have better response to etoposide treatment than the filamin-A deficient tumors. This is associated with more potent induction of DNA double strand breaks (DSBs) by Topo II poisons in filamin-A proficient cells than the deficient cells. Although the expression of filamin-A enables cells a slightly stronger capability to repair DSB, the net outcome is that filamin-A proficient cells bear more DSBs due to the significantly enhanced DSB induction by Topo II poisons in these cells. We further found that filamin-A proficient cells have increased drug influx and decreased drug efflux, suggesting that filamin-A modulates the intra-cellular drug kinetics of Topo II poisons to facilitate the generation of DSB after Topo II poison exposure. These data suggest a novel function of filamin-A in regulating the pharmacokinetics of Topo II poisons, and that the status of filamin-A may be used as a prognostic marker for Topo II poisons based cancer treatments.
Collapse
Affiliation(s)
- Jingyin Yue
- Department of Radiation Oncology, The Cancer Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, NJ 08903, USA
| | | | | | | |
Collapse
|
80
|
Lipid raft/caveolae signaling is required for Cryptococcus neoformans invasion into human brain microvascular endothelial cells. J Biomed Sci 2012; 19:19. [PMID: 22316086 PMCID: PMC3308189 DOI: 10.1186/1423-0127-19-19] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Accepted: 02/08/2012] [Indexed: 12/30/2022] Open
Abstract
Background Cryptococcus neoformans has a predilection for central nervous system infection. C. neoformans traversal of the blood brain barrier, composed of human brain microvascular endothelial cells (HBMEC), is the crucial step in brain infection. However, the molecular mechanism of the interaction between Cryptococcus neoformans and HBMEC, relevant to its brain invasion, is still largely unknown. Methods In this report, we explored several cellular and molecular events involving the membrane lipid rafts and caveolin-1 (Cav1) of HBMEC during C. neoformans infection. Immunofluorescence microscopy was used to examine the roles of Cav1. The knockdown of Cav1 by the siRNA treatment was performed. Phosphorylation of Cav1 relevant to its invasion functions was investigated. Results We found that the host receptor CD44 colocalized with Cav1 on the plasma membrane, and knockdown of Cav1 significantly reduced the fungal ability to invade HBMEC. Although the CD44 molecules were still present, HBMEC membrane organization was distorted by Cav1 knockdown. Concomitantly, knockdown of Cav1 significantly reduced the fungal crossing of the HBMEC monolayer in vitro. Upon C. neoformans engagement, host Cav1 was phosphorylated in a CD44-dependent manner. This phosphorylation was diminished by filipin, a disrupter of lipid raft structure. Furthermore, the phosphorylated Cav1 at the lipid raft migrated inward to the perinuclear localization. Interestingly, the phospho-Cav1 formed a thread-like structure and colocalized with actin filaments but not with the microtubule network. Conclusion These data support that C. neoformans internalization into HBMEC is a lipid raft/caveolae-dependent endocytic process where the actin cytoskeleton is involved, and the Cav1 plays an essential role in C. neoformans traversal of the blood-brain barrier.
Collapse
|
81
|
Inder KL, Zheng YZ, Davis MJ, Moon H, Loo D, Nguyen H, Clements JA, Parton RG, Foster LJ, Hill MM. Expression of PTRF in PC-3 Cells modulates cholesterol dynamics and the actin cytoskeleton impacting secretion pathways. Mol Cell Proteomics 2012; 11:M111.012245. [PMID: 22030351 PMCID: PMC3277761 DOI: 10.1074/mcp.m111.012245] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2011] [Revised: 10/23/2011] [Indexed: 01/08/2023] Open
Abstract
Expression of caveolin-1 is up-regulated in prostate cancer metastasis and is associated with aggressive recurrence of the disease. Intriguingly, caveolin-1 is also secreted from prostate cancer cell lines and has been identified in secreted prostasomes. Caveolin-1 is the major structural component of the plasma membrane invaginations called caveolae. Co-expression of the coat protein Polymerase I and transcript release factor (PTRF) is required for caveolae formation. We recently found that expression of caveolin-1 in the aggressive prostate cancer cell line PC-3 is not accompanied by PTRF, leading to noncaveolar caveolin-1 lipid rafts. Moreover, ectopic expression of PTRF in PC-3 cells sequesters caveolin-1 into caveolae. Here we quantitatively analyzed the effect of PTRF expression on the PC-3 proteome using stable isotope labeling by amino acids in culture and subcellular proteomics. We show that PTRF reduced the secretion of a subset of proteins including secreted proteases, cytokines, and growth regulatory proteins, partly via a reduction in prostasome secretion. To determine the cellular mechanism accounting for the observed reduction in secreted proteins we analyzed total membrane and the detergent-resistant membrane fractions. Our data show that PTRF expression selectively impaired the recruitment of actin cytoskeletal proteins to the detergent-resistant membrane, which correlated with altered cholesterol distribution in PC-3 cells expressing PTRF. Consistent with this, modulating cellular cholesterol altered the actin cytoskeleton and protein secretion in PC-3 cells. Intriguingly, several proteins that function in ER to Golgi trafficking were reduced by PTRF expression. Taken together, these results suggest that the noncaveolar caveolin-1 found in prostate cancer cells generates a lipid raft microenvironment that accentuates secretion pathways, possibly at the step of ER sorting/exit. Importantly, these effects could be modulated by PTRF expression.
Collapse
Affiliation(s)
- Kerry L. Inder
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Yu Zi Zheng
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
- §Centre for High-Throughput Biology and Department of Biochemistry and Molecular Biology, 2125 East Mall, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Melissa J. Davis
- ¶Queensland Facility for Advanced Bioinformatics, Brisbane, Queensland 4072, Australia
- **The University of Queensland Institute for Molecular Bioscience, Brisbane, Queensland 4072, Australia
| | - Hyeongsun Moon
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Dorothy Loo
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Hien Nguyen
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| | - Judith A. Clements
- ‖Australian Prostate Cancer Research Centre –Queensland, Institute for Molecular Bioscience, The University of Queensland, University of Technology, Brisbane, Queensland 4059, Australia
| | - Robert G. Parton
- **The University of Queensland Institute for Molecular Bioscience, Brisbane, Queensland 4072, Australia
| | - Leonard J. Foster
- §Centre for High-Throughput Biology and Department of Biochemistry and Molecular Biology, 2125 East Mall, University of British Columbia, Vancouver, BC, Canada, V6T 1Z4
| | - Michelle M. Hill
- From the ‡The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia
| |
Collapse
|
82
|
Caveolae and the regulation of endocytosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 729:14-28. [PMID: 22411311 DOI: 10.1007/978-1-4614-1222-9_2] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Although clathrin-mediated endocytosis constitutes the main pathway for internalization of extracellular ligands and plasma membrane components it has generally been accepted that other uptake mechanisms-caveolae-mediated and noncaveolar raft-dependent endocytosis-also exist. During the last 20 years many papers have been published about caveolar endocytosis. These studies have fundamentally changed our view about the endocytotic role of caveolae. Views that caveolae are permanently static structures1 have been extensively considered and rejected. Although the initial steps leading to the pinching off of caveolae from the plasma membrane have been studied in details, there are still contradictory data about the intracellular trafficking of caveolae. It is still not entirely clear whether caveolar endocytosis represents an uptake pathway with distinct cellular compartments to avoid lysosomal degradation or ligands taken up by caveolae can also be targeted to late endosomes/lysosomes.In this chapter, we summarize the data available about caveolar endocytosis focusing on the intracellular route of caveolae and we provide data supporting that caveolar endocytosis can join the classical endocytotic pathway.
Collapse
|
83
|
Muriel O, Echarri A, Hellriegel C, Pavón DM, Beccari L, Del Pozo MA. Phosphorylated filamin A regulates actin-linked caveolae dynamics. J Cell Sci 2011; 124:2763-76. [PMID: 21807941 DOI: 10.1242/jcs.080804] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Caveolae are relatively stable membrane invaginations that compartmentalize signaling, regulate lipid metabolism and mediate viral entry. Caveolae are closely associated with actin fibers and internalize in response to diverse stimuli. Loss of cell adhesion is known to induce rapid and robust caveolae internalization and trafficking toward a Rab11-positive recycling endosome; however, pathways governing this process are poorly understood. Here, we report that filamin A is required to maintain the F-actin-dependent linear distribution of caveolin-1. High spatiotemporal resolution particle tracking of caveolin-1-GFP vesicles by total internal reflection fluorescence (TIRF) microscopy revealed that FLNa is required for the F-actin-dependent arrest of caveolin-1 vesicles in a confined area and their stable anchorage to the plasma membrane. The linear distribution and anchorage of caveolin-1 vesicles are both required for proper caveolin-1 inwards trafficking. De-adhesion-triggered caveolae inward trafficking towards a recycling endosome is impaired in FLNa-depleted HeLa and FLNa-deficient M2-melanoma cells. Inwards trafficking of caveolin-1 requires both the ability of FLNa to bind actin and cycling PKCα-dependent phosphorylation of FLNa on Ser2152 after cell detachment.
Collapse
Affiliation(s)
- Olivia Muriel
- Integrin Signaling Laboratory, Department of Vascular Biology, Centro Nacional de Investigaciones Cardiovasculares (CNIC), Melchor Fernández Almagro 3, 28029 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
84
|
Gervásio OL, Phillips WD, Cole L, Allen DG. Caveolae respond to cell stretch and contribute to stretch-induced signaling. J Cell Sci 2011; 124:3581-90. [PMID: 22045729 DOI: 10.1242/jcs.084376] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Caveolae are invaginations of the plasma membrane that are formed by caveolins. Caveolar membranes are also enriched in cholesterol, glycosphingolipids and signaling enzymes such as Src kinase. Here we investigate the effect of cell stretch upon caveolar dynamics and signaling. Transfection of C2 myoblasts with caveolin-3-YFP led to the formation of caveolae-like membrane pits 50-100 nm in diameter. Glycosphingolipids became immobilized and tightly packed together within caveolin-rich regions of the plasma membrane. Fluorescence resonance energy transfer (FRET) was used to assess the degree of glycosphingolipid packing. Myoblasts were subjected to a brief (1 minute) stretch on an elastic substratum. Stretch caused a reduction in glycosphingolipid FRET, consistent with a reversible unfolding of caveolar pits in response to membrane tension. Cells expressing caveolin-3-YFP also displayed an enhanced stretch-induced activation of Src kinase, as assessed by immunofluorescence. Repeated stretches resulted in the trafficking and remodeling of caveolin-3-rich membrane domains and accelerated turnover of membrane glycosphingolipids. The stretch-induced unfolding of caveolae, activation of Src and redistribution of caveolin and glycosphingolipids might reflect mechanisms of the cellular adaptation to mechanical stresses.
Collapse
Affiliation(s)
- Othon L Gervásio
- School of Medical Sciences, Discipline of Physiology (F13), Bosch Institute, University of Sydney, NSW 2006, Australia
| | | | | | | |
Collapse
|
85
|
Kronstein R, Seebach J, Grossklaus S, Minten C, Engelhardt B, Drab M, Liebner S, Arsenijevic Y, Taha AA, Afanasieva T, Schnittler HJ. Caveolin-1 opens endothelial cell junctions by targeting catenins. Cardiovasc Res 2011; 93:130-40. [PMID: 21960684 DOI: 10.1093/cvr/cvr256] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
AIMS A fundamental phenomenon in inflammation is the loss of endothelial barrier function, in which the opening of endothelial cell junctions plays a central role. However, the molecular mechanisms that ultimately open the cell junctions are largely unknown. METHODS AND RESULTS Impedance spectroscopy, biochemistry, and morphology were used to investigate the role of caveolin-1 in the regulation of thrombin-induced opening of cell junctions in cultured human and mouse endothelial cells. Here, we demonstrate that the vascular endothelial (VE) cadherin/catenin complex targets caveolin-1 to endothelial cell junctions. Association of caveolin-1 with VE-cadherin/catenin complexes is essential for the barrier function decrease in response to the pro-inflammatory mediator thrombin, which causes a reorganization of the complex in a rope ladder-like pattern accompanied by a loss of junction-associated actin filaments. Mechanistically, we show that in response to thrombin stimulation the protease-activated receptor 1 (PAR-1) causes phosphorylation of caveolin-1, which increasingly associates with β- and γ-catenin. Consequently, the association of β- and γ-catenin with VE-cadherin is weakened, thus allowing junction reorganization and a decrease in barrier function. Thrombin-induced opening of cell junctions is lost in caveolin-1-knockout endothelial cells and after expression of a Y/F-caveolin-1 mutant but is completely reconstituted after expression of wild-type caveolin-1. CONCLUSION Our results highlight the pivotal role of caveolin-1 in VE-cadherin-mediated cell adhesion via catenins and, in turn, in barrier function regulation.
Collapse
Affiliation(s)
- Romy Kronstein
- Institute of Physiology, Medical Faculty of the TU-Dresden, Germany
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Liu J, Youn H, Yang J, Du N, Liu J, Liu H, Li B. G-protein alpha-s and -12 subunits are involved in androgen-stimulated PI3K activation and androgen receptor transactivation in prostate cancer cells. Prostate 2011; 71:1276-86. [PMID: 21308712 PMCID: PMC3143312 DOI: 10.1002/pros.21345] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2010] [Accepted: 12/21/2010] [Indexed: 11/10/2022]
Abstract
BACKGROUND The androgen receptor (AR) is a ligand-dependent transcription factor that mediates androgenic hormone action in cells. We recently demonstrated the involvement of phosphoinositide 3-OH kinase (PI3K) p110beta in AR transactivation and gene expression. In this study, we determined the upstream signals that lead to PI3K/p110beta activation and AR transactivation after androgen stimulation. METHODS Human prostate cancer LAPC-4 and 22Rv1 cell lines were used for the experiments. AR transactivation was assessed using an androgen responsive element-driven luciferase (ARE-LUC) assay. Cell proliferation was examined using BrdU incorporation and MTT assays. Target genes were silenced using small interfering RNA (siRNA) approach. Gene expression was evaluated at the mRNA level (real-time RT-PCR) and protein level (Western blot). PI3K kinase activities were measured using immunoprecipitation-based in vitro kinase assay. The AR-DNA-binding activity was determined using chromatin-immunoprecipitation (ChIP) assay. RESULTS First, at the cellular plasma membrane, disrupting the integrity of caveolae microdomain with methyl-β-cyclodextrin (M-β-CD) abolished androgen-induced AR transactivation and gene expression. Then, knocking down caveolae structural proteins caveolin-1 or -2 with the gene-specific siRNAs significantly reduced androgen-induced AR transactivation. Next, silencing Gα(s) and Gα(12) genes but not other G-proteins blocked androgen-induced AR transactivation and cell proliferation. Consistently, overexpression of Gα(s) or Gα(12) active mutants enhanced androgen-induced AR transactivation, of which Gα(s) active mutant sensitized the AR to castration-level of androgen (R1881). Most interestingly, knocking down Gα(s) but not Gα(12) subunit significantly suppressed androgen-stimulated PI3K p110beta activation. However, ChIP analysis revealed that both Gα(s) or Gα(12) subunits are involved in androgen-induced AR interaction with the AR target gene PSA promoter region. CONCLUSION These data suggest that caveolae-associated G-protein alpha subunits are involved in AR transactivation by modulating the activities of different PI3K isoforms.
Collapse
Affiliation(s)
- Jianjun Liu
- Department of Urology, the Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Hyewon Youn
- Institute of Radiation Medicine and Department of Nuclear Medicine, Seoul National University College of Medicine, Seoul 110-799, Korea
- Departments of Urology, the University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Jun Yang
- Departments of Urology, the University of Kansas Medical Center, Kansas City, Kansas 66160
- Department of Urology, Tongji Hospital, Huazhong University of Science & Technology, Wuhan, Hubei 430030, China
| | - Ningchao Du
- Department of Urology, the Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Jihong Liu
- Departments of Urology, the University of Kansas Medical Center, Kansas City, Kansas 66160
| | - Hongwei Liu
- Department of Urology, the Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
| | - Benyi Li
- Department of Urology, the Affiliated Hospital of Guangdong Medical College, Zhanjiang, Guangdong 524001, China
- Departments of Urology, the University of Kansas Medical Center, Kansas City, Kansas 66160
- Corresponding Author: Benyi Li, MD/PhD, KUMC Urology, 3901 Rainbow Blvd, Kansas City, KS 66160. Tel: 913-588-4773; Fax: 913-588-4756;
| |
Collapse
|
87
|
Tomassian T, Humphries LA, Liu SD, Silva O, Brooks DG, Miceli MC. Caveolin-1 orchestrates TCR synaptic polarity, signal specificity, and function in CD8 T cells. THE JOURNAL OF IMMUNOLOGY 2011; 187:2993-3002. [PMID: 21849673 DOI: 10.4049/jimmunol.1101447] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
TCR engagement triggers the polarized recruitment of membrane, actin, and transducer assemblies within the T cell-APC contact that amplify and specify signaling cascades and T effector activity. We report that caveolin-1, a scaffold that regulates polarity and signaling in nonlymphoid cells, is required for optimal TCR-induced actin polymerization, synaptic membrane raft polarity, and function in CD8, but not CD4, T cells. In CD8(+) T cells, caveolin-1 ablation selectively impaired TCR-induced NFAT-dependent NFATc1 and cytokine gene expression, whereas caveolin-1 re-expression promoted NFATc1 gene expression. Alternatively, caveolin-1 ablation did not affect TCR-induced NF-κB-dependent Iκbα expression. Cav-1(-/-) mice did not efficiently promote CD8 immunity to lymphocytic choriomeningitis virus, nor did cav-1(-/-) OT-1(+) CD8(+) T cells efficiently respond to Listeria monocytogenes-OVA after transfer into wild-type hosts. Therefore, caveolin-1 is a T cell-intrinsic orchestrator of TCR-mediated membrane polarity and signal specificity selectively employed by CD8 T cells to customize TCR responsiveness.
Collapse
Affiliation(s)
- Tamar Tomassian
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine and College of Letters and Sciences, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | |
Collapse
|
88
|
Majumdar S, Hajduczki A, Vithayathil R, Olsen TJ, Spitler RM, Mendez AS, Thompson TD, Weiss GA. In vitro evolution of ligands to the membrane protein caveolin. J Am Chem Soc 2011; 133:9855-62. [PMID: 21615158 DOI: 10.1021/ja201792q] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Membrane proteins comprise a third of the human genome, yet present challenging targets for reverse chemical genetics. For example, although implicated in numerous diseases including multiple myeloma, the membrane protein caveolin-1 appears to offer a poor target for the discovery of synthetic ligands due to its largely unknown structure and insolubility. To break this impasse and identify new classes of caveolae controlling lead compounds, we applied phage-based, reverse chemical genetics for the discovery of caveolin-1 ligands derived from the anti-HIV therapeutic T20. Substitution of homologous residues into the T20 sequence used a process analogous to medicinal chemistry for the affinity maturation to bind caveolin. The resultant caveolin-1 ligands bound with >1000-fold higher affinity than wild-type T20. Two types of ELISAs and isothermal titration calorimetry (ITC) measurements demonstrated high affinity binding to caveolin by the T20 variants with K(d) values in the 150 nM range. Microscopy experiments with the highest affinity caveolin ligands confirmed colocalization of the ligands with endogenous caveolin in NIH 3T3 cells. The results establish the foundation for targeting caveolin and caveolae formation in living cells.
Collapse
Affiliation(s)
- Sudipta Majumdar
- Department of Chemistry, University of California, Irvine, California 92697, USA
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Fecchi K, Travaglione S, Spadaro F, Quattrini A, Parolini I, Piccaro G, Raggi C, Fabbri A, Felicetti F, Carè A, Fiorentini C, Sargiacomo M. Human melanoma cells express FGFR/Src/Rho signaling that entails an adhesion-independent caveolin-1 membrane association. Int J Cancer 2011; 130:1273-83. [PMID: 21445970 DOI: 10.1002/ijc.26092] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2010] [Accepted: 03/07/2011] [Indexed: 11/08/2022]
Abstract
Caveolae have been indicated as a center of cytoskeleton regulation for Src kinase/Rho GTPase signaling. In addition, Src recruitment on intact cortical actin cytoskeleton appears to be required for bFGF/FGFR signal activation. Recently, we established a relationship between caveolin-1 (Cav-1) expression and cell migration in human malignant melanoma, constitutively activated by a bFGF autoregulatory loop. This work intends to investigate whether caveolae's asset, through bFGF/FGFR/c-Src/Rho signaling, could be related to melanoma cell anchorage. Accordingly, we revealed the existence of a FGFR/Src kinase pathway in Cav-1 enriched detergent-resistant membranes (DRMs) of Me665/1 metastatic melanoma cells, as confirmed by FGFR silencing. Moreover, we determined the expression and phosphorylation levels of Cav-1/Src/Erk signal pathway as a function of FGFR activation and cell density. A sucrose density gradient ultracentrifugation was employed to monitor Cav-1 membrane association and buoyancy in Me665/1 cells treated for actin fragmentation or for altered phosphorylation signals. As a result, melanoma cells show remarkable resistance to Cav-1 disassembly, together with persisting cell signal activity, being Src and Cav-1 crucial modulators of Rho GTPases. In conclusion, our study primarily highlights, in a metastatic melanoma cell line expressing caveolin, the circumstances whereby caveola structural and functional endurance enables the FGFR/Src/Rho GTPases pathway to keep on cell progression.
Collapse
Affiliation(s)
- Katia Fecchi
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, 299-00161-Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Sayner SL, Balczon R, Frank DW, Cooper DMF, Stevens T. Filamin A is a phosphorylation target of membrane but not cytosolic adenylyl cyclase activity. Am J Physiol Lung Cell Mol Physiol 2011; 301:L117-24. [PMID: 21478251 DOI: 10.1152/ajplung.00417.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Transmembrane adenylyl cyclase (AC) generates a cAMP pool within the subplasma membrane compartment that strengthens the endothelial cell barrier. This cAMP signal is steered toward effectors that promote junctional integrity and is inactivated before it accesses microtubules, where the cAMP signal causes phosphorylation of tau, leading to microtubule disassembly and barrier disruption. During infection, Pseudomonas aeruginosa uses a type III secretion system to inject a soluble AC, ExoY, into the cytosol of pulmonary microvascular endothelial cells. ExoY generates a cAMP signal that disrupts the endothelial cell barrier. We tested the hypothesis that this ExoY-dependent cAMP signal causes phosphorylation of tau, without inducing phosphorylation of membrane effectors that strengthen endothelial barrier function. To approach this hypothesis, we first discerned the membrane compartment in which endogenous transmembrane AC6 resides. AC6 was resolved in caveolin-rich lipid raft fractions with calcium channel proteins and the cell adhesion molecules N-cadherin, E-cadherin, and activated leukocyte adhesion molecule. VE-cadherin was excluded from the caveolin-rich fractions and was detected in the bulk plasma membrane fractions. The actin binding protein, filamin A, was detected in all membrane fractions. Isoproterenol activation of ACs promoted filamin phosphorylation, whereas thrombin inhibition of AC6 reduced filamin phosphorylation within the membrane fraction. In contrast, ExoY produced a cAMP signal that did not cause filamin phosphorylation yet induced tau phosphorylation. Hence, our data indicate that cAMP signals are strictly compartmentalized; whereas cAMP emanating from transmembrane ACs activates barrier-enhancing targets, such as filamin, cAMP emanating from soluble ACs activates barrier-disrupting targets, such as tau.
Collapse
Affiliation(s)
- Sarah L Sayner
- Department of Cell Biology and Neuroscience, University of South Alabama, Mobile, Alabama 36688, USA.
| | | | | | | | | |
Collapse
|
91
|
Black AT, Hayden PJ, Casillas RP, Heck DE, Gerecke DR, Sinko PJ, Laskin DL, Laskin JD. Regulation of Hsp27 and Hsp70 expression in human and mouse skin construct models by caveolae following exposure to the model sulfur mustard vesicant, 2-chloroethyl ethyl sulfide. Toxicol Appl Pharmacol 2011; 253:112-20. [PMID: 21457723 DOI: 10.1016/j.taap.2011.03.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Revised: 03/16/2011] [Accepted: 03/22/2011] [Indexed: 01/14/2023]
Abstract
Dermal exposure to the vesicant sulfur mustard causes marked inflammation and tissue damage. Basal keratinocytes appear to be a major target of sulfur mustard. In the present studies, mechanisms mediating skin toxicity were examined using a mouse skin construct model and a full-thickness human skin equivalent (EpiDerm-FT™). In both systems, administration of the model sulfur mustard vesicant, 2-chloroethyl ethyl sulfide (CEES, 100-1000μM) at the air surface induced mRNA and protein expression of heat shock proteins 27 and 70 (Hsp27 and Hsp70). CEES treatment also resulted in increased expression of caveolin-1, the major structural component of caveolae. Immunohistochemistry revealed that Hsp27, Hsp70 and caveolin-1 were localized in basal and suprabasal layers of the epidermis. Caveolin-1 was also detected in fibroblasts in the dermal component of the full thickness human skin equivalent. Western blot analysis of caveolar membrane fractions isolated by sucrose density centrifugation demonstrated that Hsp27 and Hsp70 were localized in caveolae. Treatment of mouse keratinocytes with filipin III or methyl-β-cyclodextrin, which disrupt caveolar structure, markedly suppressed CEES-induced Hsp27 and Hsp70 mRNA and protein expression. CEES treatment is known to activate JNK and p38 MAP kinases; in mouse keratinocytes, inhibition of these enzymes suppressed CEES-induced expression of Hsp27 and Hsp70. These data suggest that MAP kinases regulate Hsp 27 and Hsp70; moreover, caveolae-mediated regulation of heat shock protein expression may be important in the pathophysiology of vesicant-induced skin toxicity.
Collapse
Affiliation(s)
- Adrienne T Black
- Pharmacology and Toxicology, Rutgers University, Piscataway, NJ, USA
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Nethe M, Hordijk PL. A model for phospho-caveolin-1-driven turnover of focal adhesions. Cell Adh Migr 2011; 5:59-64. [PMID: 20948305 DOI: 10.4161/cam.5.1.13702] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The regulation of Focal Adhesion (FA) dynamics is a key aspect of cellular motility. FAs concentrate integrins and associated cytoskeletal elements as well as a large number of regulatory proteins, including adapters, kinases and small GTPases of the Rho Family. We have recently shown that activated Rac1 can localize to FAs and can initiate the accumulation of the adapter protein Caveolin1 (Cav1) at FAs. As reported by several groups including ours, this translocation requires Cav1 phosphorylation at Tyr14, presumably by Src. Here we provide additional data regarding this process and briefly review recent literature. Finally, we incorporated the different pieces of available information into a mechanistic model. This model proposes that local Rac1 activation initiates a series of events that involve endosomal traffic of Cav1 and Src, targeting these proteins to or near FAs. Next, within specific membrane domains, Src can mediate the phosphorylation of Cav1 at Tyr 14, which is important for the stable FA localization of Cav1. Finally, dephosphorylation of Cav1 may represent a key step required for internalization, FA turnover and cell motility.
Collapse
Affiliation(s)
- Micha Nethe
- Department of Molecular Cell Biology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| | | |
Collapse
|
93
|
Kalwa H, Michel T. The MARCKS protein plays a critical role in phosphatidylinositol 4,5-bisphosphate metabolism and directed cell movement in vascular endothelial cells. J Biol Chem 2010; 286:2320-30. [PMID: 21097841 DOI: 10.1074/jbc.m110.196022] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The MARCKS protein (myristoylated alanine-rich C kinase substrate) is an actin- and calmodulin-binding protein that is expressed in many mammalian tissues. The role of MARCKS in endothelial signaling responses is incompletely understood. We found that siRNA-mediated knockdown of MARCKS in cultured endothelial cells abrogated directed cell movement in a wound healing assay. We used biochemical and cell imaging approaches to explore the role of MARCKS in endothelial signal transduction pathways activated by insulin. Insulin treatment of vascular endothelial cells promoted the dose- and time-dependent phosphorylation of MARCKS. Cell imaging and hydrodynamic approaches revealed that MARCKS is targeted to plasmalemmal caveolae and undergoes subcellular translocation in response to insulin. Insulin treatment promoted an increase in levels of the signaling phospholipid phosphatidylinositol 4,5-bisphosphate (PIP(2)) in plasmalemmal caveolae. The insulin-stimulated increase in caveolar PIP(2) was blocked by siRNA-mediated knockdown of MARCKS, as determined using both biochemical assays and imaging studies using FRET-based PIP(2) biosensors. The critical role of PIP(2) in MARCKS responses was explored by examining the PIP(2)- and actin-binding proteins Arp2/3 and N-WASP. Insulin promoted the rapid and robust phosphorylation of both N-WASP and Arp2/3, but these phosphorylation responses were markedly attenuated by siRNA-mediated MARCKS knockdown. Moreover, MARCKS knockdown effectively abrogated N-WASP activation in response to insulin, as determined using a FRET-based N-WASP activity biosensor. Taken together, these studies show that MARCKS plays a key role in insulin-dependent endothelial signaling to PIP(2) and is a critical determinant of actin assembly and directed cell movement in the vascular endothelium.
Collapse
Affiliation(s)
- Hermann Kalwa
- Cardiovascular Medicine Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | |
Collapse
|
94
|
Affiliation(s)
- Michele Bastiani
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Robert G. Parton
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland 4072, Australia
- Centre for Microscopy and Microanalysis, The University of Queensland, Brisbane, Queensland 4072, Australia
| |
Collapse
|
95
|
Boettcher JP, Kirchner M, Churin Y, Kaushansky A, Pompaiah M, Thorn H, Brinkmann V, MacBeath G, Meyer TF. Tyrosine-phosphorylated caveolin-1 blocks bacterial uptake by inducing Vav2-RhoA-mediated cytoskeletal rearrangements. PLoS Biol 2010; 8. [PMID: 20808760 PMCID: PMC2927421 DOI: 10.1371/journal.pbio.1000457] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2009] [Accepted: 07/07/2010] [Indexed: 12/11/2022] Open
Abstract
During the early stages of infection, Neisseria gonorrhoeae triggers a phosphotyrosine-dependent Cav1-Vav2-RhoA signaling cascade that promotes the pathogen's extracellular state. Certain bacterial adhesins appear to promote a pathogen's extracellular lifestyle rather than its entry into host cells. However, little is known about the stimuli elicited upon such pathogen host-cell interactions. Here, we report that type IV pili (Tfp)-producing Neisseria gonorrhoeae (P+GC) induces an immediate recruitment of caveolin-1 (Cav1) in the host cell, which subsequently prevents bacterial internalization by triggering cytoskeletal rearrangements via downstream phosphotyrosine signaling. A broad and unbiased analysis of potential interaction partners for tyrosine-phosphorylated Cav1 revealed a direct interaction with the Rho-family guanine nucleotide exchange factor Vav2. Both Vav2 and its substrate, the small GTPase RhoA, were found to play a direct role in the Cav1-mediated prevention of bacterial uptake. Our findings, which have been extended to enteropathogenic Escherichia coli, highlight how Tfp-producing bacteria avoid host cell uptake. Further, our data establish a mechanistic link between Cav1 phosphorylation and pathogen-induced cytoskeleton reorganization and advance our understanding of caveolin function. Like many bacterial pathogens, successful attachment of Neisseria gonorrhoeae—the causative agent of the sexually transmitted disease gonorrhoea—to its host cells depends on specialized structures on the bacterial surface called type IV pili (Tfp). Pathogen attachment induces changes within host cells that may facilitate and promote infection. In this study, we identify some of the earliest cellular signals elicited by N. gonorrhoeae during infection, which, in this case, prevent the organism from entering the cell precociously. After attachment to host cells the bacteria form microcolonies on the cell surface. Underneath these microcolonies, so-called cortical plaques form within the host cell—these contain the cytoskeleton protein actin and a range of signaling proteins. We show that N. gonorrhoeae recruits a host cell protein called caveolin-1 to the cell membrane where the bacteria are attached; here, caveloin-1 effectively impedes uptake of the bacteria by activating a signaling cascade that involves its phosphorylation on a tyrosine residue and subsequent interactions with proteins that regulate the cytoskeleton. Thus, these proteins play a pivotal role in maintaining N. gonorrhoeae in the extracellular milieu. By extrapolating our findings to another Tfp-producing bacterium, the enteropathogenic Escherichia coli, we argue that the establishment and maintenance of this extracellular state benefits certain pathogens by giving them time to express proteins required for subsequent steps of infection.
Collapse
Affiliation(s)
- Jan Peter Boettcher
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Marieluise Kirchner
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Yuri Churin
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Alexis Kaushansky
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Malvika Pompaiah
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Hans Thorn
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Volker Brinkmann
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Gavin MacBeath
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, United States of America
| | - Thomas F. Meyer
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
- * E-mail:
| |
Collapse
|
96
|
Radhakrishnan A, Yeo D, Brown G, Myaing MZ, Iyer LR, Fleck R, Tan BH, Aitken J, Sanmun D, Tang K, Yarwood A, Brink J, Sugrue RJ. Protein analysis of purified respiratory syncytial virus particles reveals an important role for heat shock protein 90 in virus particle assembly. Mol Cell Proteomics 2010; 9:1829-48. [PMID: 20530633 DOI: 10.1074/mcp.m110.001651] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
In this study, we used imaging and proteomics to identify the presence of virus-associated cellular proteins that may play a role in respiratory syncytial virus (RSV) maturation. Fluorescence microscopy of virus-infected cells revealed the presence of virus-induced cytoplasmic inclusion bodies and mature virus particles, the latter appearing as virus filaments. In situ electron tomography suggested that the virus filaments were complex structures that were able to package multiple copies of the virus genome. The virus particles were purified, and the protein content was analyzed by one-dimensional nano-LC MS/MS. In addition to all the major virus structural proteins, 25 cellular proteins were also detected, including proteins associated with the cortical actin network, energy pathways, and heat shock proteins (HSP70, HSC70, and HSP90). Representative actin-associated proteins, HSC70, and HSP90 were selected for further biological validation. The presence of beta-actin, filamin-1, cofilin-1, HSC70, and HSP90 in the virus preparation was confirmed by immunoblotting using relevant antibodies. Immunofluorescence microscopy of infected cells stained with antibodies against relevant virus and cellular proteins confirmed the presence of these cellular proteins in the virus filaments and inclusion bodies. The relevance of HSP90 to virus infection was examined using the specific inhibitors 17-N-Allylamino-17-demethoxygeldanamycin. Although virus protein expression was largely unaffected by these drugs, we noted that the formation of virus particles was inhibited, and virus transmission was impaired, suggesting an important role for HSP90 in virus maturation. This study highlights the utility of proteomics in facilitating both our understanding of the role that cellular proteins play during RSV maturation and, by extrapolation, the identification of new potential targets for antiviral therapy.
Collapse
Affiliation(s)
- Anuradha Radhakrishnan
- Division of Molecular and Cell Biology, School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
97
|
Vieira FS, Corrêa G, Einicker-Lamas M, Coutinho-Silva R. Host-cell lipid rafts: a safe door for micro-organisms? Biol Cell 2010; 102:391-407. [PMID: 20377525 PMCID: PMC7161784 DOI: 10.1042/bc20090138] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2010] [Accepted: 02/10/2010] [Indexed: 12/20/2022]
Abstract
The lipid raft hypothesis proposed that these microdomains are small (10-200 nM), highly dynamic and enriched in cholesterol, glycosphingolipids and signalling phospholipids, which compartmentalize cellular processes. These membrane regions play crucial roles in signal transduction, phagocytosis and secretion, as well as pathogen adhesion/interaction. Throughout evolution, many pathogens have developed mechanisms to escape from the host immune system, some of which are based on the host membrane microdomain machinery. Thus lipid rafts might be exploited by pathogens as signalling and entry platforms. In this review, we summarize the role of lipid rafts as players in the overall invasion process used by different pathogens to escape from the host immune system.
Collapse
Affiliation(s)
- Flávia Sarmento Vieira
- Laboratório de Imunofisiologia, Universidade Federal do Rio de Janeiro, Instituto de Biofísica Carlos Chagas Filho, CCS, Rio de Janeiro, RJ, Brazil
| | | | | | | |
Collapse
|
98
|
Hansen CG, Nichols BJ. Exploring the caves: cavins, caveolins and caveolae. Trends Cell Biol 2010; 20:177-86. [PMID: 20153650 DOI: 10.1016/j.tcb.2010.01.005] [Citation(s) in RCA: 225] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 01/07/2010] [Accepted: 01/11/2010] [Indexed: 01/29/2023]
Abstract
Caveolae are ampullate (flask-shaped) invaginations that are abundant in the plasma membrane of many mammalian cell types. Although caveolae are implicated in a wide range of processes including endothelial transcytosis, lipid homeostasis and cellular signalling, a detailed molecular picture of many aspects of their function has been elusive. Until recently, the only extensively characterised protein components of caveolae were the caveolins. Recently, data from several laboratories have demonstrated that a family of four related proteins, termed cavins 1-4, plays key roles in caveolar biogenesis and function. Salient properties of the cavin family include their propensity to form complexes with each other and their different but overlapping tissue distribution. This review summarises recent data on the cavins, and sets them in the context of open questions on the construction and function of caveolae. The discovery of cavins implies that caveolae might have unexpectedly diverse structural properties, in accord with the wide range of functions attributed to these 'little caves'.
Collapse
|
99
|
Lajoie P, Nabi IR. Lipid Rafts, Caveolae, and Their Endocytosis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2010; 282:135-63. [DOI: 10.1016/s1937-6448(10)82003-9] [Citation(s) in RCA: 266] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
|
100
|
Norambuena A, Palma F, Poblete MI, Donoso MV, Pardo E, González A, Huidobro-Toro JP. UTP controls cell surface distribution and vasomotor activity of the human P2Y2 receptor through an epidermal growth factor receptor-transregulated mechanism. J Biol Chem 2009; 285:2940-50. [PMID: 19996104 DOI: 10.1074/jbc.m109.081166] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Extracellular nucleotides transmit signals into the cells through the P2 family of cell surface receptors. These receptors are amply expressed in human blood vessels and participate in vascular tone control; however, their signaling mechanisms remain unknown. Here we show that in smooth muscle cells of isolated human chorionic arteries, the activation of the P2Y(2) receptor (P2Y(2)R) induces not only its partition into membrane rafts but also its rapid internalization. Cholesterol depletion with methyl-beta-cyclodextrin reduced the association of the agonist-activated receptor into membrane rafts but did not affect either the UTP-mediated vasoconstrictions or the vasomotor responses elicited by both serotonin and KCl. Ex vivo perfusion of human chorionic artery segments with 1-10 mum UTP, a selective P2Y(2)R agonist, displaced the P2Y(2)R localization into membrane rafts within 1 min, a process preceded by the activation of both RhoA and Rac1 GTPases. AG1478, a selective and potent inhibitor of the epidermal growth factor receptor tyrosine kinase activity, not only blocked the UTP-induced vasomotor activity but also abrogated both RhoA and Rac1 activation, the P2Y(2)R association with membrane rafts, and its internalization. Altogether, these results show for the first time that the plasma membrane distribution of the P2Y(2)R is transregulated by the epidermal growth factor receptor, revealing an unsuspected functional interplay that controls both the membrane distribution and the vasomotor activity of the P2Y(2)R in intact human blood vessels.
Collapse
Affiliation(s)
- Andrés Norambuena
- Departamento de Ciencias Fisiológicas, Facultad de Ciencias Biológicas, Centro de Regulación Celular y Patología JV Luco, Instituto Milenio de Biología Fundamental y Aplicada, Santiago, Chile.
| | | | | | | | | | | | | |
Collapse
|