151
|
Cristofaro V, Peters CA, Yalla SV, Sullivan MP. Smooth muscle caveolae differentially regulate specific agonist induced bladder contractions. Neurourol Urodyn 2007; 26:71-80. [PMID: 17123298 DOI: 10.1002/nau.20361] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
AIMS Caveolae are cholesterol-rich plasmalemmal microdomains that serve as sites for sequestration of signaling proteins and thus may facilitate, organize, and integrate responses to extracellular stimuli. While previous studies in the bladder have demonstrated alterations in caveolae with particular physiologic or pathologic conditions, little attention has been focused on the functional significance of these organelles. Therefore, the purpose of this study was to investigate the role of caveolae in the modulation of receptor-mediated signal transduction and determine the presence and localization of caveolin proteins in bladder tissue. METHODS Contractile responses to physiologic agonists were measured in rat bladder tissue before and after disruption of caveolae achieved by depleting membrane cholesterol with methyl-beta-cyclodextrin. Stimulation with agonists was repeated after caveolae were restored as a result of cholesterol replenishment. RT-PCR, immmunohistochemistry, and Western blotting were used to determine the expression and localization of caveolin mRNA and proteins. RESULTS Following caveolae disruption, contractile responses to angiotensin II and serotonin were attenuated, whereas responses to bradykinin and phenylephrine were augmented. Cholesterol replenishment restored responses towards baseline. Carbachol and KCl induced contractions were not affected by caveolae disruption. Ultrastructure analysis confirmed loss of caveolae following cholesterol depletion with cyclodextrin and caveolae restoration following cholesterol replacement. Gene and protein expression of caveolin-1, -2, and -3 was detected in bladder tissue. Immunoreactivity for all three caveolins was observed in smooth muscle cells throughout the bladder. CONCLUSIONS The functional effects of cholesterol depletion on specific agonist-induced contractile events and the expression of all three caveolins in bladder smooth muscle support a central role for caveolae in regulation of selective G-protein-coupled receptor signaling pathways in bladder smooth muscle. Thus, caveolae serve to differentially regulate bladder smooth muscle by a stimulus-dependent potentiation or inhibition of bladder contraction.
Collapse
Affiliation(s)
- V Cristofaro
- Division of Urology, VA Boston Healthcare System, Brigham & Women's Hospital, Boston, Massachusetts 02132, USA
| | | | | | | |
Collapse
|
152
|
Shmuel M, Nodel-Berner E, Hyman T, Rouvinski A, Altschuler Y. Caveolin 2 regulates endocytosis and trafficking of the M1 muscarinic receptor in MDCK epithelial cells. Mol Biol Cell 2007; 18:1570-85. [PMID: 17314410 PMCID: PMC1855036 DOI: 10.1091/mbc.e06-07-0618] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Clathrin and caveolins are known for their involvement in the internalization of numerous receptors. Here we show that in polarized epithelial Madin-Darby canine kidney cells, both the clathrin machinery and caveolins are involved in the endocytosis and delivery to the plasma membrane (PM) of the M1 muscarinic acetylcholine receptor (mAChR). We initially localized this receptor to the lateral membrane, where it accumulates proximal to the tight junctions. From there it is internalized through the clathrin-mediated pathway. In addition, the receptor may associate on the PM with caveolin (cav) 2 or in intracellular compartments with either cav 2, or monomeric or oligomeric cav 1. Association of the PM M1 mAChR with cav 2 inhibits receptor endocytosis through the clathrin-mediated pathway or retains the receptor in an intracellular compartment. This intracellular association attenuates receptor trafficking. Expression of cav 1 with cav 2 rescues the latter's inhibitory effect. The caveolins stimulate M1 mAChR oligomerization thus maintaining a constant amount of monomeric receptor. These results provide evidence that caveolins play a role in the attenuation of the M1 muscarinic receptor's intracellular trafficking to and from the PM.
Collapse
Affiliation(s)
- Miriam Shmuel
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Efrat Nodel-Berner
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Tehila Hyman
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Alexander Rouvinski
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| | - Yoram Altschuler
- Department of Pharmacology, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
| |
Collapse
|
153
|
Sanna E, Miotti S, Mazzi M, De Santis G, Canevari S, Tomassetti A. Binding of nuclear caveolin-1 to promoter elements of growth-associated genes in ovarian carcinoma cells. Exp Cell Res 2007; 313:1307-17. [PMID: 17359972 DOI: 10.1016/j.yexcr.2007.02.005] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2006] [Revised: 01/18/2007] [Accepted: 02/05/2007] [Indexed: 11/26/2022]
Abstract
Caveolin-1 (cav-1), a member of a protein family associated mainly with cell membrane microdomains in many cell types, acts as a tumor suppressor in ovarian carcinoma cells. Biochemical analyses demonstrated that cav-1 was also localized in the nuclei of ovarian carcinoma cells, endogenously (SKOV3) or ectopically (IGtC3) expressing cav-1. By confocal analyses, the same cell lines as well as IGROV1 and SKOV3 cells transiently transfected with green fluorescent protein-cav-1 fusion protein showed nuclear punctate speckled pattern. Subnuclear distribution analysis revealed cav-1 mainly associated with the nuclear matrix, but also slightly with chromatin. Cav-1 was found in nuclear high-molecular weight complexes and by confocal analysis was found to co-localized with the inner nuclear membrane protein emerin. Cyclin D1 and folate receptor promoters were modulated by cav-1 in SKOV3 cells as demonstrated by transient transfection with or silencing of cav-1. Chromatin immunoprecipitation and supershift assays indicated that nuclear cav-1 can bind in vitro and in vivo to promoter sequences of both cyclin D1 and folate receptor genes. These data suggest that in ovarian carcinoma cells cav-1, localized in transcriptionally inactive chromatin, exerts a functional activity mediated, at least in part, by directly binding to sequences of genes involved in proliferation.
Collapse
Affiliation(s)
- Elena Sanna
- Unit of Molecular Therapies, Department of Experimental Oncology, Istituto Nazionale Tumori, 20133, Milan, Italy
| | | | | | | | | | | |
Collapse
|
154
|
Howell GJ, Holloway ZG, Cobbold C, Monaco AP, Ponnambalam S. Cell biology of membrane trafficking in human disease. ACTA ACUST UNITED AC 2007; 252:1-69. [PMID: 16984815 PMCID: PMC7112332 DOI: 10.1016/s0074-7696(06)52005-4] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding the molecular and cellular mechanisms underlying membrane traffic pathways is crucial to the treatment and cure of human disease. Various human diseases caused by changes in cellular homeostasis arise through a single gene mutation(s) resulting in compromised membrane trafficking. Many pathogenic agents such as viruses, bacteria, or parasites have evolved mechanisms to subvert the host cell response to infection, or have hijacked cellular mechanisms to proliferate and ensure pathogen survival. Understanding the consequence of genetic mutations or pathogenic infection on membrane traffic has also enabled greater understanding of the interactions between organisms and the surrounding environment. This review focuses on human genetic defects and molecular mechanisms that underlie eukaryote exocytosis and endocytosis and current and future prospects for alleviation of a variety of human diseases.
Collapse
Affiliation(s)
- Gareth J Howell
- Endothelial Cell Biology Unit, Faculty of Biological Sciences, University of Leeds, Leeds LS2 9JT, United Kingdom
| | | | | | | | | |
Collapse
|
155
|
Sun XH, Flynn DC, Castranova V, Millecchia LL, Beardsley AR, Liu J. Identification of a novel domain at the N terminus of caveolin-1 that controls rear polarization of the protein and caveolae formation. J Biol Chem 2007; 282:7232-41. [PMID: 17213184 DOI: 10.1074/jbc.m607396200] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
When cells are migrating, caveolin-1, the principal protein component of caveolae, is excluded from the leading edge and polarized at the cell rear. The dynamic feature depends on a specific sequence motif that directs intracellular trafficking of the protein. Deletion mutation analysis revealed a putative polarization domain at the N terminus of caveolin-1, between amino acids 32-60. Alanine substitution identified a minimal sequence of 10 residues ((46)TKEIDLVNRD(55)) necessary for caveolin-1 rear polarization. Interestingly, deletion of amino acids 1-60 did not prevent the polarization of caveolin-1 in human umbilical vein endothelial cells or wild-type mouse embryonic fibroblasts because of an interaction of Cav(61-178) mutant with endogenous caveolin-1. Surprisingly, expression of the depolarization mutant in caveolin-1 null cells dramatically impeded caveolae formation. Furthermore, knockdown of caveolae formation by methyl-beta-cyclodextrin failed to prevent wild-type caveolin-1 rear polarization. Importantly, genetic depletion of caveolin-1 led to disoriented migration, which can be rescued by full-length caveolin-1 but not the depolarization mutant, indicating a role of caveolin-1 polarity in chemotaxis. Thus, we have identified a sequence motif that is essential for caveolin-1 rear polarization and caveolae formation.
Collapse
Affiliation(s)
- Xing-Hui Sun
- Department of Physiology and Pharmacology, West Virginia University, Morgantown, West Virginia 26506, USA
| | | | | | | | | | | |
Collapse
|
156
|
Costa MJ, Senou M, Van Rode F, Ruf J, Capello M, Dequanter D, Lothaire P, Dessy C, Dumont JE, Many MC, Van Sande J. Reciprocal negative regulation between thyrotropin/3',5'-cyclic adenosine monophosphate-mediated proliferation and caveolin-1 expression in human and murine thyrocytes. Mol Endocrinol 2007; 21:921-32. [PMID: 17202321 DOI: 10.1210/me.2006-0328] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
The expression of caveolins is down-regulated in tissue samples of human thyroid autonomous adenomas and in the animal model of this disease. Because several cell types present in thyroid express caveolins, it remained unclear if this down-regulation occurs in thyrocytes and which are the mechanism and role of this down-regulation in the tumor context. Here we show that prolonged stimulation of isolated human thyrocytes by TSH/cAMP/cAMP-dependent protein kinase inhibits caveolins' expression. The expression of caveolins is not down-regulated by activators of other signaling pathways relevant to thyroid growth/function. Therefore, the down-regulation of caveolins' expression in autonomous adenomas is a direct consequence of the chronic activation of the TSH/cAMP pathway in thyrocytes. The down-regulation of caveolin-1 occurs at the mRNA level, with a consequent protein decrease. TSH/cAMP induces a transcription-dependent, translation-independent destabilization of the caveolin-1 mRNA. This effect is correlated to the known proliferative role of that cascade in thyrocytes. In vivo, thyrocytes of caveolin-1 knockout mice display enhanced proliferation. This demonstrates, for the first time, the in vivo significance of the specific caveolin-1 down-regulation by one mitogenic cascade and its relation to a human disease.
Collapse
Affiliation(s)
- Maria José Costa
- Institut de Recherche Interdisciplinaire, Campus Erasme, Université Libre de Bruxelles, 808 Route de Lennik, Building C, 1070 Brussels, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Abstract
Isolation of detergent-resistant membranes (DRMs; also known as detergent-insoluble glycolipid-enriched membranes [DIGs] or glycolipid-enriched membranes [GEMs]) that are enriched in proteins and lipids with a high affinity for rafts is one of the simplest and most widely used methods for studying rafts. However, it is essential to understand the limitations as well as the advantages of this method. DRMs do not correspond precisely to rafts in living cells. For this reason, finding a protein enriched in DRMs does not prove that it was in rafts in the living cell. Furthermore, the fraction of a protein found in DRMs provides no quantitative information about the fraction of the protein originally in rafts. In fact, DRMs may be isolated from membranes that did not even contain rafts before detergent extraction. DRM-association is useful because it reflects a high-inherent affinity of a protein for the ordered membrane state found in rafts. Treatments that affect the DRM-association of a protein can thus be inferred to affect its raft affinity. Current models suggest that rafts may form in a regulated manner, often associated with clustering of membrane proteins or lipids, during processes such as signal transduction. DRM-association is a read-out of whether a protein is likely to associate with rafts that form under these conditions.
Collapse
Affiliation(s)
- Deborah A Brown
- Dept. of Biochemistry & Cell Biology, Stony Brook University, NY 11794-4215, USA
| |
Collapse
|
158
|
Guirland C, Zheng JQ. Membrane lipid rafts and their role in axon guidance. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 621:144-55. [PMID: 18269217 DOI: 10.1007/978-0-387-76715-4_11] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The plasma membrane of cells contains a variety of lipid and protein molecules that are often segregated and heterogeneously distributed in microdomains. Lipid rafts represent a generalized concept of membrane microdomains that are enriched in cholesterol and sphingolipids and, characteristically, resistant to cold detergent extraction. Lipid rafts have recently received considerable attention because they are thought to be involved in many cellular functions, in particular, signal transduction for extracellular stimuli. Many of these functions are also intimately related to the processes involved in neural development, including neurotrophic factor signaling and synaptic plasticity. Recent studies from our lab and others have indicated an important role for lipid rafts in axonal growth and guidance. Specifically, our data show that lipid rafts on the plasma membrane provide platforms for spatial and temporal control of guidance signaling by extracellular cues. In addition, lipid rafts may also function in other aspects of axonal growth and guidance, including spatial and temporal regulation of adhesion, cytoskeletal dynamics, and growth cone motility. Further elucidating how membrane rafts are involved in guided axonal growth would provide important insights into the intricate signaling mechanisms underlying neuronal wiring, which is fundamental for normal brain development and functional recovery after injury and diseases.
Collapse
Affiliation(s)
- Carmine Guirland
- Department of Neuroscience and Cell Biology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway 08854, USA
| | | |
Collapse
|
159
|
Sprenger R, Fontijn R, van Marle J, Pannekoek H, Horrevoets A. Spatial segregation of transport and signalling functions between human endothelial caveolae and lipid raft proteomes. Biochem J 2006; 400:401-10. [PMID: 16886909 PMCID: PMC1698592 DOI: 10.1042/bj20060355] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Lipid rafts and caveolae are biochemically similar, specialized domains of the PM (plasma membrane) that cluster specific proteins. However, they are morphologically distinct, implying different, possibly complementary functions. Two-dimensional gel electrophoresis preceding identification of proteins by MS was used to compare the relative abundance of proteins in DRMs (detergent-resistant membranes) isolated from HUVEC (human umbilical-vein endothelial cells), and caveolae immunopurified from DRM fractions. Various signalling and transport proteins were identified and additional cell-surface biotinylation revealed the majority to be exposed, demonstrating their presence at the PM. In resting endothelial cells, the scaffold of immunoisolated caveolae consists of only few resident proteins, related to structure [CAV1 (caveolin-1), vimentin] and transport (V-ATPase), as well as the GPI (glycosylphosphatidylinositol)-linked, surface-exposed protein CD59. Further quantitative characterization by immunoblotting and confocal microscopy of well-known [eNOS (endothelial nitric oxide synthase) and CAV1], less known [SNAP-23 (23 kDa synaptosome-associated protein) and BASP1 (brain acid soluble protein 1)] and novel [C8ORF2 (chromosome 8 open reading frame 2)] proteins showed different subcellular distributions with none of these proteins being exclusive to either caveolae or DRM. However, the DRM-associated fraction of the novel protein C8ORF2 (approximately 5% of total protein) associated with immunoseparated caveolae, in contrast with the raft protein SNAP-23. The segregation of caveolae from lipid rafts was visually confirmed in proliferating cells, where CAV1 was spatially separated from eNOS, SNAP-23 and BASP1. These results provide direct evidence for the previously suggested segregation of transport and signalling functions between specialized domains of the endothelial plasma membrane.
Collapse
Affiliation(s)
- Richard R. Sprenger
- *Department of Medical Biochemistry, Academic Medical Center K1-114, Meibergdreef 15, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Ruud D. Fontijn
- *Department of Medical Biochemistry, Academic Medical Center K1-114, Meibergdreef 15, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Jan van Marle
- †Department of Cell Biology, Academic Medical Centre, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Hans Pannekoek
- *Department of Medical Biochemistry, Academic Medical Center K1-114, Meibergdreef 15, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
| | - Anton J. G. Horrevoets
- *Department of Medical Biochemistry, Academic Medical Center K1-114, Meibergdreef 15, University of Amsterdam, 1105 AZ, Amsterdam, The Netherlands
- To whom correspondence should be addressed (email )
| |
Collapse
|
160
|
Hunter I, Nixon GF. Spatial compartmentalization of tumor necrosis factor (TNF) receptor 1-dependent signaling pathways in human airway smooth muscle cells. Lipid rafts are essential for TNF-alpha-mediated activation of RhoA but dispensable for the activation of the NF-kappaB and MAPK pathways. J Biol Chem 2006; 281:34705-15. [PMID: 16982613 PMCID: PMC2653078 DOI: 10.1074/jbc.m605738200] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor (TNF)-alpha-induced activation of RhoA, mediated by TNF receptor 1 (TNFR1), is a prerequisite step in a pathway that leads to increased 20-kDa light chain of myosin (MLC20) phosphorylation and airway smooth muscle contraction. In this study, we have investigated the proximal events in TNF-alpha-induced RhoA activation. TNFR1 is localized to both lipid raft and nonraft regions of the plasma membrane in primary human airway smooth muscle cells. TNF-alpha engagement of TNFR1 recruited the adaptor proteins TRADD, TRAF-2, and RIP into lipid rafts and activated RhoA, NF-kappaB, and MAPK pathways. Depletion of cholesterol from rafts with methyl-beta-cyclodextrin caused a redistribution of TNFR1 to nonraft plasma membrane and prevented ligand-induced RhoA activation. By contrast, TNF-alpha-induced activation of NF-kappaB and MAPKs was unaffected by methyl-beta-cyclodextrin indicating that, in airway smooth muscle cells, activation of these pathways occurred independently of lipid rafts. Targeted knockdown of caveolin-1 completely abrogated TNF-alpha-induced RhoA activation, identifying this raft-resident protein as a positive regulator of the activation process. The signaling adaptors TRADD and RIP were also found to be necessary for ligand-induced RhoA activation. Taken together, our results suggest that in airway smooth muscle cells, spatial compartmentalization of TNFR1 provides a mechanism for generating distinct signaling outcomes in response to ligand engagement and define a mechanistic role for lipid rafts and caveolin-1 in TNF-alpha-induced activation of RhoA.
Collapse
Affiliation(s)
- Irene Hunter
- School of Medical Sciences, University of Aberdeen, IMS Building, Foresterhill, Aberdeen AB25 2ZD, United Kingdom.
| | | |
Collapse
|
161
|
Kai M, Sakane F, Jia YJ, Imai SI, Yasuda S, Kanoh H. Lipid Phosphate Phosphatases 1 and 3 Are Localized in Distinct Lipid Rafts. ACTA ACUST UNITED AC 2006; 140:677-86. [PMID: 17005594 DOI: 10.1093/jb/mvj195] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Lipid phosphate phosphatases (LPPs), integral membrane proteins with six transmembrane domains, dephosphorylate a variety of extracellular lipid phosphates. Although LPP3 is already known to bind to Triton X-100-insoluble rafts, we here report that LPP1 is also associated with lipid rafts distinct from those harboring LPP3. We found that LPP1 was Triton X-100-soluble, but CHAPS-insoluble in LNCaP cells endogenously expressing LPP1 and several LPP1 cDNA-transfected cells including NIH3T3 fibroblasts. In addition to the non-ionic detergent insolubility, LPP1 further possessed several properties formulated for raft-localizing proteins as follows: first, the CHAPS-insolubility was resistant to the actin-disrupting drug cytochalasin D; second, the CHAPS-insoluble LPP1 floated in an Optiprep density gradient; third, the CHAPS insolubility of LPP1 was lost by cholesterol depletion; and finally, the subcellular distribution pattern of LPP1 exclusively overlapped with that of a raft marker, cholera toxin B subunit. Interestingly, confocal microscopic analysis showed that LPP1 was distributed to membrane compartments distinct from those of LPP3. Analysis using various LPP1/LPP3 chimeras revealed that their first extracellular regions determine the different Triton X-100 solubilities. These results indicate that LPP1 and LPP3 are distributed in distinct lipid rafts that may provide unique microenvironments defining their non-redundant physiological functions.
Collapse
Affiliation(s)
- Masahiro Kai
- Department of Biochemistry, Sapporo Medical University School of Medicine, West-17, South-1, Sapporo 060-8556
| | | | | | | | | | | |
Collapse
|
162
|
Wang Y, Du D, Fang L, Yang G, Zhang C, Zeng R, Ullrich A, Lottspeich F, Chen Z. Tyrosine phosphorylated Par3 regulates epithelial tight junction assembly promoted by EGFR signaling. EMBO J 2006; 25:5058-70. [PMID: 17053785 PMCID: PMC1630420 DOI: 10.1038/sj.emboj.7601384] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2006] [Accepted: 09/12/2006] [Indexed: 11/08/2022] Open
Abstract
The conserved polarity complex, comprising the partitioning-defective (Par) proteins Par3 and Par6, and the atypical protein kinase C, functions in various cell-polarization events and asymmetric cell divisions. However, little is known about whether and how external stimuli-induced signals may regulate Par3 function in epithelial cell polarity. Here, we found that Par3 was tyrosine phosphorylated through phosphoproteomic profiling of pervanadate-induced phosphotyrosine proteins. We also demonstrated that the tyrosine phosphorylation event induced by multiple growth factors including epidermal growth factor (EGF) was dependent on activation of Src family kinase (SFK) members c-Src and c-Yes. The tyrosine residue 1127 (Y1127) of Par3 was identified as the major EGF-induced phosphorylation site. Moreover, we found that Y1127 phosphorylation reduced the association of Par3 with LIM kinase 2 (LIMK2), thus enabling LIMK2 to regulate cofilin phosphorylation dynamics. Substitution of Y1127 for phenylalanine impaired the EGF-induced Par3 and LIMK2 dissociation and delayed epithelial tight junction (TJ) assembly considerably. Collectively, these data suggest a novel, phosphotyrosine-dependent fine-tuning mechanism of Par3 in epithelial TJ assembly controlled by the EGF receptor-SFK signaling pathway.
Collapse
Affiliation(s)
- Yiguo Wang
- Key Laboratory of Proteomics and Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Graduate School of the Chinese Academy of Sciences, Beijing, China
| | - Dan Du
- Key Laboratory of Proteomics and Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Graduate School of the Chinese Academy of Sciences, Beijing, China
| | - Longhou Fang
- Key Laboratory of Proteomics and Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Graduate School of the Chinese Academy of Sciences, Beijing, China
| | - Guang Yang
- Key Laboratory of Proteomics and Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- Graduate School of the Chinese Academy of Sciences, Beijing, China
| | - Chenyi Zhang
- Key Laboratory of Proteomics and Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Rong Zeng
- Key Laboratory of Proteomics and Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Axel Ullrich
- Department of Molecular Biology, Max-Planck-Institute of Biochemistry, Martinsried, Germany
| | | | - Zhengjun Chen
- Key Laboratory of Proteomics and Laboratory of Molecular Cell Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
- SHARF Laboratory, Shanghai, China
- Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai 200031, China. Tel.: 86 21 54921081; Fax: 86 21 54921081; E-mail:
| |
Collapse
|
163
|
Eldstrom J, Van Wagoner DR, Moore ED, Fedida D. Localization of Kv1.5 channels in rat and canine myocyte sarcolemma. FEBS Lett 2006; 580:6039-46. [PMID: 17054951 DOI: 10.1016/j.febslet.2006.09.069] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2006] [Accepted: 09/27/2006] [Indexed: 10/24/2022]
Abstract
Voltage-gated potassium (Kv) channel subtypes localize to the plasma membrane of a number of cell types, and the sarcolemma in myocytes. Because many signaling molecules concentrate in subdomains of the plasma membrane, the localization of Kv channels to these sites may have important implications for channel function and regulation. In this study, the association of the voltage-gated potassium channel Kv1.5 with a specific subtype of lipid rafts, caveolae, in rat and canine cardiac myocytes has been investigated. Interactions between caveolin-3 and beta-dystroglycan or eNOS, as well as between Kv1.5 and alpha-actinin were readily detected in co-immunoprecipitation experiments, whereas no association between Kv1.5 and caveolin-3 was evident. Wide-field microscopy and deconvolution techniques revealed that the percent co-localization of Kv1.5 with caveolin-3 was extremely low in atrial myocytes from rat and canine hearts (8+/-1% and 12.2+/-2%, respectively), and limited in ventricular myocytes (11+/-4% and 20+/-3% in rat and canine, respectively). Immunoelectron microscopic imaging of rat atrial and ventricular tissues showed that Kv1.5 and caveolin-3 labeling generally did not overlap. In HEK293 cells stably expressing the channel, Kv1.5 did not target to the low buoyant density raft fraction along with flotillin but instead fractionated along with the non-raft associated transferrin receptor. Taken together, these results suggest that Kv1.5 is not present in caveolae of rat and canine heart.
Collapse
Affiliation(s)
- Jodene Eldstrom
- Department of Anesthesiology, Pharmacology and Therapeutics, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC, Canada V6T 1Z3.
| | | | | | | |
Collapse
|
164
|
Hartung A, Bitton-Worms K, Rechtman MM, Wenzel V, Boergermann JH, Hassel S, Henis YI, Knaus P. Different routes of bone morphogenic protein (BMP) receptor endocytosis influence BMP signaling. Mol Cell Biol 2006; 26:7791-805. [PMID: 16923969 PMCID: PMC1636853 DOI: 10.1128/mcb.00022-06] [Citation(s) in RCA: 203] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Endocytosis is important for a variety of functions in eukaryotic cells, including the regulation of signaling cascades via transmembrane receptors. The internalization of bone morphogenetic protein (BMP) receptor type I (BRI) and type II (BRII) and its relation to signaling were largely unexplored. Here, we demonstrate that both receptor types undergo constitutive endocytosis via clathrin-coated pits (CCPs) but that only BRII undergoes also caveola-like internalization. Using several complementary approaches, we could show that (i) BMP-2-mediated Smad1/5 phosphorylation occurs at the plasma membrane in nonraft regions, (ii) continuation of Smad signaling resulting in a transcriptional response requires endocytosis via the clathrin-mediated route, and (iii) BMP signaling leading to alkaline phosphatase induction initiates from receptors that fractionate into cholesterol-enriched, detergent-resistant membranes. Furthermore, we show that BRII interacts with Eps15R, a constitutive component of CCPs, and with caveolin-1, the marker protein of caveolae. Taken together, the localization of BMP receptors in distinct membrane domains is prerequisite to their taking different endocytosis routes with specific impacts on Smad-dependent and Smad-independent signaling cascades.
Collapse
Affiliation(s)
- Anke Hartung
- Department of Physiological Chemistry II, University of Wuerzburg, Wuerzburg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
165
|
Ishii M, Iwai K, Koike M, Ohshima S, Kudo-Tanaka E, Ishii T, Mima T, Katada Y, Miyatake K, Uchiyama Y, Saeki Y. RANKL-induced expression of tetraspanin CD9 in lipid raft membrane microdomain is essential for cell fusion during osteoclastogenesis. J Bone Miner Res 2006; 21:965-76. [PMID: 16753027 DOI: 10.1359/jbmr.060308] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
UNLABELLED We showed that CD9, a member of tetraspanin superfamily proteins, is expressed in a specific membrane microdomain, called "lipid raft," and is crucial for cell fusion during osteoclastogenesis after activation of the RANK/RANKL system. INTRODUCTION Osteoclasts are bone-resorbing multinuclear polykaryons that are essential for bone remodeling and are formed through cell fusion of mononuclear macrophage/monocyte lineage precursors. Although osteoclastogenesis has been shown to be critically regulated by the RANK/RANKL system, the mechanism how precursor cells fuse with each other remains unclear. We examined the function of CD9, a member of tetraspanin superfamily, which has previously been shown to form macromolecular membrane microdomains and to regulate cell-cell fusion in various cell types. MATERIALS AND METHODS We used RAW264.7, a macrophage/monocyte lineage cell line, which can differentiate into osteoclast-like polykaryons on the application of RANKL. Expression and distribution of CD9 was assessed by Western blotting, fluorescence-assorted cell sorting (FACS) and immunohistochemistry with light and electron microscopy. A specific neutralizing antibody and RNA interference were used to inhibit the function of CD9, and green fluorescent protein (GFP)-CD9 was exogenously expressed to enhance the effect of CD9. The distribution of CD9 in lipid microdomain was examined by biochemical (sucrose density gradient) isolation and imaging technique. RESULTS CD9 is expressed on cell surfaces of RAW264.7, which is enhanced by RANKL. Targeted inhibition of CD9 decreases the number of osteoclast-like cells. On the other hand, overexpression of CD9 promotes spontaneous cell fusion even in the absence of RANKL. CD9 is localized in detergent-insoluble "lipid raft" microdomain in RANKL stimulation, and disruption of lipid rafts markedly reduces the formation of osteoclast-like polykaryons. Immunohistochemical studies of bone tissues revealed the expression of CD9 in osteoclasts in vivo. CONCLUSIONS These data suggest that function of tetraspanin CD9 and its expression in lipid rafts are crucial for cell fusion during osteoclastogenesis.
Collapse
Affiliation(s)
- Masaru Ishii
- Department of Clinical Research, Osaka Minami Medical Center, Osaka, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
166
|
Sonnino S, Prinetti A, Mauri L, Chigorno V, Tettamanti G. Dynamic and Structural Properties of Sphingolipids as Driving Forces for the Formation of Membrane Domains. Chem Rev 2006; 106:2111-25. [PMID: 16771445 DOI: 10.1021/cr0100446] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Sandro Sonnino
- Center of Excellence on Neurodegenerative Diseases, Department of Medical Chemistry, Biochemistry and Biotechnology, University of Milan, 20090 Segrate (MI), Italy.
| | | | | | | | | |
Collapse
|
167
|
Cheng ZJ, Singh RD, Marks DL, Pagano RE. Membrane microdomains, caveolae, and caveolar endocytosis of sphingolipids. Mol Membr Biol 2006; 23:101-10. [PMID: 16611585 DOI: 10.1080/09687860500460041] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Caveolae are flask-shape membrane invaginations of the plasma membrane that have been implicated in endocytosis, transcytosis, and cell signaling. Recent years have witnessed the resurgence of studies on caveolae because they have been found to be involved in the uptake of some membrane components such as glycosphingolipids and integrins, as well as viruses, bacteria, and bacterial toxins. Accumulating evidence shows that endocytosis mediated by caveolae requires unique structural and signaling machinery (caveolin-1, src kinase), which indicates that caveolar endocytosis occurs through a mechanism which is distinct from other forms of lipid microdomain-associated, clathrin-independent endocytosis. Furthermore, a balance of glycosphingolipids, cholesterol, and caveolin-1 has been shown to be important in regulating caveolae endocytosis.
Collapse
Affiliation(s)
- Zhi-Jie Cheng
- Department of Biochemistry and Molecular Biology, Thoracic Diseases Research Unit, Mayo Clinic and Foundation, Rochester, Minnesota 55905, USA
| | | | | | | |
Collapse
|
168
|
Macdonald J, Li Z, Su W, Pike LJ. The membrane proximal disulfides of the EGF receptor extracellular domain are required for high affinity binding and signal transduction but do not play a role in the localization of the receptor to lipid rafts. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2006; 1763:870-8. [PMID: 16842869 PMCID: PMC2771566 DOI: 10.1016/j.bbamcr.2006.05.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/02/2006] [Revised: 04/23/2006] [Accepted: 05/02/2006] [Indexed: 11/28/2022]
Abstract
The EGF receptor is a transmembrane receptor tyrosine kinase that is enriched in lipid rafts. Subdomains I, II and III of the extracellular domain of the EGF receptor participate in ligand binding and dimer formation. However, the function of the cysteine-rich subdomain IV has not been elucidated. In this study, we analyzed the role of the membrane-proximal portion of subdomain IV in EGF binding and signal transduction. A double Cys-->Ala mutation that breaks the most membrane-proximal disulfide bond (Cys600 to Cys612), ablated high affinity ligand binding and substantially reduced signal transduction. A similar mutation that breaks the overlapping Cys596 to Cys604 disulfide had little effect on receptor function. Mutation of residues within the Cys600 to Cys612 disulfide loop did not alter the ligand binding or signal transducing activities of the receptor. Despite the fact that the C600,612A EGF receptor was significantly impaired functionally, this receptor as well as all of the other receptors with mutations in the region of residues 596 to 612 localized normally to lipid rafts. These data suggest that the disulfide-bonded structure of the membrane-proximal portion of the EGF receptor, rather than its primary sequence, is important for EGF binding and signaling but is not involved in localizing the receptor to lipid rafts.
Collapse
Affiliation(s)
| | | | | | - Linda J. Pike
- Author to whom correspondence should be addressed, Telephone: 314) 362-9502 FAX: (314) 362-7183,
| |
Collapse
|
169
|
Kasai A, Shima T, Okada M. Role of Src family tyrosine kinases in the down-regulation of epidermal growth factor signaling in PC12 cells. Genes Cells 2006; 10:1175-87. [PMID: 16324154 DOI: 10.1111/j.1365-2443.2005.00909.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Src family tyrosine kinases (SFKs) play pivotal roles as molecular switches for various intracellular signaling pathways. SFKs have been implicated in epidermal growth factor (EGF) signaling, although their precise mechanisms of action in this pathway remain elusive. To address this issue, we focused on a membrane microdomain, lipid rafts, where SFKs are enriched. In PC12 cells, the EGF receptor (EGFR) is constitutively concentrated in lipid rafts, and further accumulation takes place upon EGF stimulation, followed by activation of SFKs, especially Src and Yes. Inhibition of SFK or disruption of lipid raft function causes EGF-induced neurite extension of PC12 cells. These effects are accompanied by an extended duration of Erk1/2 activation and are suppressed by a MEK inhibitor. In Csk(-/-) fibroblasts, suppression of SFK results in prolonged EGF-induced activation of Erk1/2, with concomitant suppression of EGFR degradation. Furthermore, analysis of the behavior of labeled EGF in PC12 cells reveals that suppression of SFK activity attenuates the rate of clustering of activated EGFR on the membrane. These results suggest that SFK activity in lipid rafts is required to facilitate the down-regulation of EGF signaling, by regulating the clustering of activated EGFR on the membrane in PC12 cells.
Collapse
Affiliation(s)
- Atsuko Kasai
- Department of Oncogene Research, Research Institute for Microbial Diseases, Osaka University, Suita, Japan
| | | | | |
Collapse
|
170
|
Meye C, Schumann J, Wagner A, Gross P. Effects of homocysteine on the levels of caveolin-1 and eNOS in caveolae of human coronary artery endothelial cells. Atherosclerosis 2006; 190:256-63. [PMID: 16616146 DOI: 10.1016/j.atherosclerosis.2006.03.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2005] [Revised: 02/20/2006] [Accepted: 03/09/2006] [Indexed: 11/24/2022]
Abstract
One aspect of homocysteine (Hcy) action is the impairment of endothelial cell function due to an impairment of endothelial nitric oxide (NO) production. The activity of the endothelial isoform of NO synthase (eNOS) is regulated by its interaction with caveolin-1 (Cav-1). The aim of this study was to determine whether Hcy may alter the levels of Cav-1 and eNOS in endothelial caveolae. We isolated caveolae-enriched membrane fractions from Hcy-treated human coronary artery endothelial cells. We found that treatment with 500 microM Hcy for 6h significantly reduced the levels of Cav-1 and eNOS in caveolae compared to untreated control by 47+/-7% and by 38+/-14%, respectively. Similarly, long-term incubation (96h) of HCAEC with 100 microM Hcy led to a comparable effect. The decreased Cav-1 abundance in endothelial caveolae in response to Hcy resulted from a decrease in Cav-1 expression at the transcriptional level. The reduced levels of eNOS in caveolae were caused by a translocation of eNOS from the caveolar fractions to noncaveolar fractions. The effects of Hcy were associated with an impairment of stimulated release of NO. These results suggest that Hcy induced impairment of NO production through a modulation of Cav-1 expression associated with a loss of eNOS in caveolae.
Collapse
Affiliation(s)
- Constanze Meye
- Division of Nephrology, Department of Internal Medicine III, Technical University of Dresden, Dresden, Germany.
| | | | | | | |
Collapse
|
171
|
Abstract
Many biological functions of heme oxygenase (HO), such as cytoprotection against oxidative stress, vasodilation, neurotransmission in the central or peripheral nervous systems, and anti-inflammatory, anti-apoptotic, or anti-proliferative potential, have been attributed to its enzymatic byproduct carbon monoxide (CO), although roles for biliverdin/bilirubin and iron have also been proposed. In addition to these well-characterized effects, recent findings reveal that HO-derived CO may act as an oxygen sensor and circadian modulator of heme biosynthesis. In lymphocytes, CO may participate in regulatory T cell function. A number of the known signaling effects of CO depend on stimulation of soluble guanylate cyclase and/or activation of mitogen-activated protein kinases (MAPK). Furthermore, modulation of caveolin-1 status may serve as an essential component of certain aspects of CO action, such as growth control. In this review, we summarize recent findings of the beneficial or detrimental effects of endogenous CO with an emphasis on the signaling pathways and downstream targets that trigger the action of this gas.
Collapse
Affiliation(s)
- Hong Pyo Kim
- Department of Medicine, Division of Pulmonary, Allergy and Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| | | | | |
Collapse
|
172
|
Wu L, Gonias SL. The low-density lipoprotein receptor-related protein-1 associates transiently with lipid rafts. J Cell Biochem 2006; 96:1021-33. [PMID: 16149055 DOI: 10.1002/jcb.20596] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The low-density lipoprotein receptor-related protein-1 (LRP-1) is a multifunctional receptor that undergoes constitutive endocytosis and recycling. To identify LRP-1 in lipid rafts, we biotin-labeled cells using a membrane-impermeable reagent and prepared Triton X-100 fractions. Raft-associated proteins were identified in streptavidin affinity-precipitates of the Triton X-100-insoluble fraction. PDGF beta-receptor was identified exclusively in lipid rafts, whereas transferrin receptor was excluded. LRP-1 distributed partially into rafts in murine embryonic fibroblasts (MEFs) and HT 1080 cells, but not in smooth muscle cells and CHO cells. LRP-1 partitioning into rafts was not altered by ligands, including alpha2-macroglobulin, platelet-derived growth factor-BB, and receptor-associated protein (RAP). To examine LRP-1 trafficking between membrane microdomains, we developed a novel method based on biotinylation and detergent fractionation. Association of LRP-1 with rafts was transient; by 15 min, nearly all of the LRP-1 that was initially raft-associated exited this compartment. LRP-1 in the Triton X-100-soluble fraction, which excludes lipid rafts, demonstrated complex kinetics, with phases reflecting import from rafts, endocytosis, and recycling. Potassium depletion blocked LRP-1 endocytosis but did not inhibit trafficking of LRP-1 from rafts into detergent-soluble microdomains. Our data support a model in which LRP-1 transiently associates with rafts but does not form a stable pool. Fluid movement of LRP-1 between microdomains may facilitate its function in promoting the endocytosis of other plasma membrane proteins, such as the urokinase receptor, which localizes in lipid rafts.
Collapse
Affiliation(s)
- Lihua Wu
- Department of Pathology, University of California San Diego, La Jolla, California 92093, USA
| | | |
Collapse
|
173
|
Eisensamer B, Uhr M, Meyr S, Gimpl G, Deiml T, Rammes G, Lambert JJ, Zieglgänsberger W, Holsboer F, Rupprecht R. Antidepressants and antipsychotic drugs colocalize with 5-HT3 receptors in raft-like domains. J Neurosci 2006; 25:10198-206. [PMID: 16267227 PMCID: PMC6725799 DOI: 10.1523/jneurosci.2460-05.2005] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Despite different chemical structure and pharmacodynamic signaling pathways, a variety of antidepressants and antipsychotics inhibit ion fluxes through 5-HT3 receptors in a noncompetitive manner with the exception of the known competitive antagonists mirtazapine and clozapine. To further investigate the mechanisms underlying the noncompetitive inhibition of the serotonin-evoked cation current, we quantified the concentrations of different types of antidepressants and antipsychotics in fractions of sucrose flotation gradients isolated from HEK293 (human embryonic kidney 293) cells stably transfected with the 5-HT3A receptor and of N1E-115 neuroblastoma cells in relation to the localization of the 5-HT3 receptor protein within the cell membrane. Western blots revealed a localization of the 5-HT3 receptor protein exclusively in the low buoyant density (LBD) fractions compatible with a localization within raft-like domains. Also, the antidepressants desipramine, fluoxetine, and reboxetine and the antipsychotics fluphenazine, haloperidol, and clozapine were markedly enriched in LBD fractions, whereas no accumulation occurs for mirtazapine, carbamazepine, moclobemide, and risperidone. The concentrations of psychopharmacological drugs within LBD fractions was strongly associated with their inhibitory potency against serotonin-induced cation currents. The noncompetitive antagonism of antidepressants at the 5-HT3 receptor was not conferred by an enhancement of receptor internalization as shown by immunofluorescence studies, assessment of receptor density in clathrin-coated vesicles, and electrophysiological recordings after coexpression of a dominant-negative mutant of dynamin I, which inhibits receptor internalization. In conclusion, enrichment of antidepressants and antipsychotics in raft-like domains within the cell membrane appears to be crucial for their antagonistic effects at ligand-gated ion channels such as 5-HT3 receptors.
Collapse
|
174
|
Carro E, Spuch C, Trejo JL, Antequera D, Torres-Aleman I. Choroid plexus megalin is involved in neuroprotection by serum insulin-like growth factor I. J Neurosci 2006; 25:10884-93. [PMID: 16306401 PMCID: PMC6725866 DOI: 10.1523/jneurosci.2909-05.2005] [Citation(s) in RCA: 162] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The involvement of circulating insulin-like growth factor I (IGF-I) in the beneficial effects of physical exercise on the brain makes this abundant serum growth factor a physiologically relevant neuroprotective signal. However, the mechanisms underlying neuroprotection by serum IGF-I remain primarily unknown. Among many other neuroprotective actions, IGF-I enhances clearance of brain amyloid beta (Abeta) by modulating transport/production of Abeta carriers at the blood-brain interface in the choroid plexus. We found that physical exercise increases the levels of the choroid plexus endocytic receptor megalin/low-density lipoprotein receptor-related protein-2 (LRP2), a multicargo transporter known to participate in brain uptake of Abeta carriers. By manipulating choroid plexus megalin levels through viral-directed overexpression and RNA interference, we observed that megalin mediates IGF-I-induced clearance of Abeta and is involved in IGF-I transport into the brain. Through this dual role, megalin participates in the neuroprotective actions of IGF-I including prevention of tau hyperphosphorylation and maintenance of cognitive function in a variety of animal models of cognitive loss. Because we found that in normal aged animals, choroid plexus megalin/LRP2 is decreased, an attenuated IGF-I/megalin input may contribute to increased risk of neurodegeneration, including late-onset Alzheimer's disease.
Collapse
Affiliation(s)
- Eva Carro
- Laboratory of Neuroendocrinology, Cajal Institute, Consejo Superior de Investigaciones Científicas, 28002 Madrid, Spain
| | | | | | | | | |
Collapse
|
175
|
Abstract
Despite many endeavors, no satisfactory strategy has emerged for modulating the aging process, most probably because they were based on faulty rationales. In an extension of the "gate theory of aging" that we proposed recently, we propose here that caveolin, an essential component of caveolae structure, may offer a potential target for modulating the aging process. According to the gate theory, certain biomolecules such as caveolins, amphiphysins, G proteins, and integrins play decisive roles in determining the senescent phenotype and thus provide targets for modulating the aging process. Among these molecules, we chose caveolin, because it can associate with a variety of regulatory and structural molecules via their scaffolding domains and thereby influence a broad spectrum of biological phenomena including both the physiology and morphology of the senescent cells. This is an attempt to review the vast body of evidence available in the literature, both direct and indirect, supporting the accord of this pivotal role to the caveolin in the background of the gate theory for the aging process.
Collapse
Affiliation(s)
- Sang Chul Park
- Department of Biochemistry and Molecular Biology, Aging and Apoptosis Research Center, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
176
|
Parr RD, Storey SM, Mitchell DM, McIntosh AL, Zhou M, Mir KD, Ball JM. The rotavirus enterotoxin NSP4 directly interacts with the caveolar structural protein caveolin-1. J Virol 2006; 80:2842-54. [PMID: 16501093 PMCID: PMC1395425 DOI: 10.1128/jvi.80.6.2842-2854.2006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2005] [Accepted: 12/13/2005] [Indexed: 11/20/2022] Open
Abstract
Rotavirus nonstructural protein 4 (NSP4) is known to function as an intracellular receptor at the endoplasmic reticulum (ER) critical to viral morphogenesis and is the first characterized viral enterotoxin. Exogenously added NSP4 induces diarrhea in rodent pups and stimulates secretory chloride currents across intestinal segments as measured in Ussing chambers. Circular dichroism studies further reveal that intact NSP4 and the enterotoxic peptide (NSP4(114-135)) that is located within the extended, C-terminal amphipathic helix preferentially interact with caveola-like model membranes. We now show colocalization of NSP4 and caveolin-1 in NSP4-transfected and rotavirus-infected mammalian cells in reticular structures surrounding the nucleus (likely ER), in the cytosol, and at the cell periphery by laser scanning confocal microscopy. A direct interaction between NSP4 residues 112 to 140 and caveolin-1 was determined by the Pro-Quest yeast two-hybrid system with full-length NSP4 and seven overlapping deletion mutants as bait, caveolin-1 as prey, and vice versa. Coimmunoprecipitation of NSP4-caveolin-1 complexes from rotavirus-infected mammalian cells demonstrated that the interaction occurs during viral infection. Finally, binding of caveolin-1 from mammalian cell lysates to Sepharose-bound, NSP4-specific synthetic peptides confirmed the yeast two-hybrid data and further delineated the binding domain to amino acids 114 to 135. We propose that the association of NSP4 and caveolin-1 contributes to NSP4 intracellular trafficking from the ER to the cell surface and speculate that exogenously added NSP4 stimulates signaling molecules located in caveola microdomains.
Collapse
Affiliation(s)
- Rebecca D Parr
- Department of Pathobiology, Texas A&M University 4467, College Station, Texas 77843, USA
| | | | | | | | | | | | | |
Collapse
|
177
|
Bush WS, Ihrke G, Robinson JM, Kenworthy AK. Antibody-specific detection of caveolin-1 in subapical compartments of MDCK cells. Histochem Cell Biol 2006; 126:27-34. [PMID: 16770576 DOI: 10.1007/s00418-006-0144-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2005] [Indexed: 10/25/2022]
Abstract
Caveolin-1 is the major structural component of caveolae and is also found in the Golgi complex of many cell types. Occasionally, caveolin-1 has been observed in additional intracellular compartments, including recycling endosomes. Why caveolin-1 expression is detected at these sites only infrequently is not clear. In this study, we test the hypothesis that non-caveolar, non-Golgi pools of caveolin-1 display unique and/or fixation-dependent epitopes. We compared the ability of a panel of antibodies raised against various domains of caveolin-1 to detect distinct subcellular pools of the protein by immunofluorescence microscopy in Madin-Darby canine kidney (MDCK) cells, a cell line where the subcellular localization of caveolin-1 has been extensively characterized. We show that three antibodies directed to the N-terminus of caveolin-1 recognize a previously undetected pool of caveolin-1 in the subapical region of MDCK cells, a localization characteristic of endosomal recycling compartments. The antibodies vary in their ability to label caveolin-1 at the cell surface, and the epitopes detected by each are highly fixation dependent. Our findings suggest that no single caveolin antibody or staining condition is capable of detecting all the caveolin-1 in a cell simultaneously. Consequently, the subcellular distribution of caveolin-1 may be much broader than currently believed.
Collapse
Affiliation(s)
- William S Bush
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, 718 Light Hall, Nashville, TN 37232, USA
| | | | | | | |
Collapse
|
178
|
Zhao H, Loh HH, Law PY. Adenylyl cyclase superactivation induced by long-term treatment with opioid agonist is dependent on receptor localized within lipid rafts and is independent of receptor internalization. Mol Pharmacol 2006; 69:1421-32. [PMID: 16415176 DOI: 10.1124/mol.105.020024] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Long-term opioid agonist treatment results in adenylyl cyclase superactivation. A recent "RAVE" theory implicates a direct correlation between the ability of agonist to induce receptor internalization and the magnitude of adenylyl cyclase superactivation. We decided to test such a theory by examining the adenylyl cyclase superactivation after long-term activation of mu-opioid receptor (MOR) in an EcR293 cell model. We examined the magnitudes of adenylyl cyclase superactivation in the presence of naloxone after long-term treatment with morphine, etorphine, and methadone, three agonists reported to have differential activities in promoting MOR internalization. It can be shown that the magnitudes of adenylyl cyclase superactivation after treating with these three agonists, although different, were dependent on MOR density. Blunting MOR internalization with the dominant-negative mutant of dynamin, K44E, did not alter the magnitude of either morphine- or etorphine-induced adenylyl cyclase superactivation. In the presence of diprenorphine, the magnitude of adenylyl cyclase superactivation after etorphine treatment was identical to that observed with morphine. It could be demonstrated further that adenylyl cyclase superactivation is dependent on the cell surface-located MOR. Sucrose gradient fractionation demonstrated the colocalization of MOR and adenylyl cyclase V/VI with caveolin-1, a marker for lipid rafts. After long-term agonist treatment, the majority of MOR remained at the lipid rafts. Methyl-beta-cyclodextrin (MbetaCD) completely blunted the adenylyl cyclase superactivation and agonist-induced receptor internalization. These MbetaCD actions were reversed by incubating the cells with cholesterol. Thus, the adenylyl cyclase superactivation is not dependent on agonist-induced receptor internalization. Rather, the location of MOR at lipid rafts is an absolute requirement for the observed adenylyl cyclase superactivation.
Collapse
Affiliation(s)
- Hui Zhao
- Department of Pharmacology, 6-120 Jackson Hall, Medical School, University of Minnesota, 321 Church St. S.E., Minneapolis, MN 55455-0217, USA
| | | | | |
Collapse
|
179
|
Boyanapalli M, Kottis V, Lahoud O, Bamri-Ezzine S, Braun PE, Mikol DD. Oligodendrocyte-myelin glycoprotein is present in lipid rafts and caveolin-1-enriched membranes. Glia 2006; 52:219-27. [PMID: 15968633 DOI: 10.1002/glia.20237] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The oligodendrocyte-myelin glycoprotein is a ligand of the neuronal Nogo receptor and a potent inhibitor of neurite outgrowth, but its physiological function remains to be elucidated. The oligodendrocyte-myelin glycoprotein is anchored solely in the outer leaflet of the plasma membrane via its glycosylphosphatidylinositol anchor, and through its leucine-rich repeat domain, it likely interacts with other proteins. In the present study, we compare its buoyancy and detergent solubility characteristics with those of other myelin proteins. Based on its detergent solubility profile and membrane fractionation using established ultracentrifugation procedures, we conclude that the oligodendrocyte-myelin glycoprotein is a lipid raft component that is closely associated with the axolemma. Moreover, it associates with caveolin-1 and caveolin-1-enriched membranes. We postulate that, by virtue of its concentration in lipid rafts and perhaps through interactions with caveolin-1, the oligodendrocyte-myelin glycoprotein may influence signaling pathways.
Collapse
|
180
|
Fujita T, Otsu K, Oshikawa J, Hori H, Kitamura H, Ito T, Umemura S, Minamisawa S, Ishikawa Y. Caveolin-3 inhibits growth signal in cardiac myoblasts in a Ca2+-dependent manner. J Cell Mol Med 2006; 10:216-24. [PMID: 16563233 PMCID: PMC3933113 DOI: 10.1111/j.1582-4934.2006.tb00302.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2006] [Accepted: 02/01/2006] [Indexed: 01/19/2023] Open
Abstract
Caveolin, a major protein component of caveolae, directly interacts with multiple signaling molecules, such as Ras and growth factor receptors, and inhibits their function. However, the role of the second messenger system in mediating this inhibition by caveolin remains poorly understood. We examined the role of Ca2+-dependent signal in caveolin- mediated growth inhibition using a rat cardiac myoblast cell line (H9C2), in which the expression of caveolin- 3, the muscle specific subtype, can be induced using the LacSwitch system. Upon induction with IPTG and serum-starvation, the expression of caveolin-3 was increased by 3.3-fold relative to that of mock-induced cells. The recombinant caveolin-3 was localized to the same subcellular fraction as endogenous caveolin-3 after sucrose gradient purification. Angiotensin II enhanced ERK phosphorylation, but this enhancement was significantly decreased in caveolin-3-induced cells in comparison to that in mock-induced cells. Similarly, when cells were stimulated with fetal calf serum, DNA synthesis, as determined by [3H]-thymidine incorporation, was significantly decreased in caveolin- 3-induced cells. When cells were treated with Ca2+ chelator (BAPTA and EGTA), however, this attenuation was blunted. Calphostin (PKC inhibitor), but not cyclosporine A treatment (calcineurin inhibitor), blunted this attenuation in caveolin-3 induced cells. Our findings suggest that caveolin exhibits growth inhibition in a Ca2+-dependent manner, most likely through PKC, in cardiac myoblasts.
Collapse
Affiliation(s)
- Takayuki Fujita
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
| | - Kouji Otsu
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
| | - Jin Oshikawa
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
| | - Hideaki Hori
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
| | - Hitoshi Kitamura
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
| | - Takaaki Ito
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
| | - Satoshi Umemura
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
| | - Susumu Minamisawa
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
| | - Yoshihiro Ishikawa
- Cardiovascular Research Institute, Department of Pathology and Department of Medicine, Yokohama City University Graduate School of Medical ScienceYokohama, Japan
- Cardiovascular Research Institute, Department of Cell Biology & Molecular Medicine and Medicine (Cardiology), New Jersey Medical SchoolNewark, NJ, USA
| |
Collapse
|
181
|
Masserini M, Pitto M, Raimondo F, Cazzaniga E, Sesana S, Bellini T. Methyl-beta-cyclodextrin treatment affects the thermotropic behaviour of membranes and detergent-resistant membrane fractions of cultured A431 cells. Biol Pharm Bull 2005; 28:2185-8. [PMID: 16327146 DOI: 10.1248/bpb.28.2185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Membranes and detergent-resistant membrane fractions isolated from human epidermoid carcinoma A431 cells after treatment with methyl-beta-cyclodextrin, a compound commonly used in pharmaceutical applications and in manipulation of membrane cholesterol content, display thermotropic transitions at about 15 degrees C and above 37 degrees C, respectively, when analyzed by differential scanning calorimetry. The transitions, absent in untreated cells, were reversible upon cycling through heating and cooling scans, and attributable to lipid components of the membranes, possibly sphingolipids. These results suggest that, after treatment with methyl-beta-cyclodextrin, membranes may show thermotropic transitions, an unusual feature for cellular bilayers, which is likely to influence biological functions.
Collapse
Affiliation(s)
- Massimo Masserini
- Department of Experimental, Environmental Medicine and Biotechnology, University of Milano-Bicocca, via Cadore 48, 20052 Monza, Italy.
| | | | | | | | | | | |
Collapse
|
182
|
Tagawa A, Mezzacasa A, Hayer A, Longatti A, Pelkmans L, Helenius A. Assembly and trafficking of caveolar domains in the cell: caveolae as stable, cargo-triggered, vesicular transporters. ACTA ACUST UNITED AC 2005; 170:769-79. [PMID: 16129785 PMCID: PMC2171342 DOI: 10.1083/jcb.200506103] [Citation(s) in RCA: 206] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Using total internal reflection fluorescence microscopy (TIR-FM), fluorescence recovery after photobleaching (FRAP), and other light microscopy techniques, we analyzed the dynamics, the activation, and the assembly of caveolae labeled with fluorescently tagged caveolin-1 (Cav1). We found that when activated by simian virus 40 (SV40), a non-enveloped DNA virus that uses caveolae for cell entry, the fraction of mobile caveolae was dramatically enhanced both in the plasma membrane (PM) and in the caveosome, an intracellular organelle that functions as an intermediate station in caveolar endocytosis. Activation also resulted in increased microtubule (MT)-dependent, long-range movement of caveolar vesicles. We generated heterokaryons that contained GFP- and RFP-tagged caveolae by fusing cells expressing Cav1-GFP and -RFP, respectively, and showed that even when activated, individual caveolar domains underwent little exchange of Cav1. Only when the cells were subjected to transient cholesterol depletion, did the caveolae domain exchange Cav1. Thus, in contrast to clathrin-, or other types of coated transport vesicles, caveolae constitute stable, cholesterol-dependent membrane domains that can serve as fixed containers through vesicle traffic. Finally, we identified the Golgi complex as the site where newly assembled caveolar domains appeared first.
Collapse
Affiliation(s)
- Akiko Tagawa
- Swiss Federal Institute of Technology (ETH) Zürich, ETH-Hönggerberg, 8093 Zürich, Switzerland
| | | | | | | | | | | |
Collapse
|
183
|
Miotti S, Tomassetti A, Facetti I, Sanna E, Berno V, Canevari S. Simultaneous expression of caveolin-1 and E-cadherin in ovarian carcinoma cells stabilizes adherens junctions through inhibition of src-related kinases. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 167:1411-27. [PMID: 16251425 PMCID: PMC1603782 DOI: 10.1016/s0002-9440(10)61228-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/02/2005] [Indexed: 01/01/2023]
Abstract
Cadherin-mediated adhesion plays an important role in maintaining cell-cell contacts and reducing tumor metastasis. However, neo-expression of E-cadherin in ovarian carcinoma does not prevent the release and spread of cells from the primary tumor. Because caveolin-1 is down-regulated concomitantly with E-cad expression, we investigated whether the stability of adherens junctions in ovarian carcinoma was affected by caveolin-1 expression. We used IGROV1 cells transfected with caveolin-1 (IGtC3), mock-transfected control cells (IGtM87), and SKOV3 cells that endogenously express caveolin-1. Simultaneous expression of caveolin-1 and E-cadherin favored membrane distribution of E-cadherin and its associated catenin (p120ctn), even when caveolin-1 was only focally associated with adherens junctions. Silencing of caveolin-1 induced intracellular E-cadherin redistribution in IGtC3 and SKOV3 cells. Treatment with the specific src kinase inhibitor PP1 increased E-cadherin expression in IGtM87 and SKOV3 cells and enhanced membrane localization of both E-cadherin and p120ctn. However, PP1 could not completely reverse the detrimental effects on cell-cell adhesion induced by Ca2+ depletion in IGtM87 cells. Together, our data suggest that caveolin-1 expression indirectly promotes cell-cell adhesion in ovarian carcinoma cells by a mechanism involving inhibition of src-related kinases. Thus, down-regulation or loss of caveolin-1 might contribute significantly to the spread of tumor cells from the primary tumor.
Collapse
Affiliation(s)
- Silvia Miotti
- Unit of Molecular Therapies, Department of Experimental Oncology, Istituto Nazionale Tumori, Via Venezian 1, 20133 Milan, Italy.
| | | | | | | | | | | |
Collapse
|
184
|
Abstract
Advances in our understanding of the complexity of GnRH actions at the pituitary and the various mechanisms involved in mediating differential LH and FSH biosynthesis and secretion at the gonadotrope, are continually emerging. In this review, we summarise recent studies pertaining to GnRH and GnRH receptor phylogeny, the divergent signalling and trafficking pathways initiated and utilised by GnRH and its receptor, and the pathways that mediate gonadotropin secretion from the gonadotrope.
Collapse
Affiliation(s)
- Adam J Pawson
- Human Reproductive Sciences Unit, Medical Research Council, The University of Edinburgh Chancellor's Building, 49 Little France Crescent, Edinburgh EH16 4SB, UK
| | | |
Collapse
|
185
|
Ishii M, Ikushima M, Kurachi Y. In vivo interaction between RGS4 and calmodulin visualized with FRET techniques: possible involvement of lipid raft. Biochem Biophys Res Commun 2005; 338:839-46. [PMID: 16246308 DOI: 10.1016/j.bbrc.2005.10.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2005] [Accepted: 10/05/2005] [Indexed: 10/25/2022]
Abstract
Regulators of G-protein signaling (RGS) are a family of proteins which accelerate intrinsic GTP-hydrolysis on heterotrimeric G-protein-alpha-subunits. Although it has been suggested that the function of RGS4 is reciprocally regulated by competitive binding of the membrane phospholipid, phosphatidylinositol-3,4,5,-trisphosphate(PtdIns(3,4,5)P(3)), and Ca(2+)/calmodulin (CaM), it remains to be shown that these interactions occur in vivo. Here, using fluorescence resonance energy transfer (FRET) techniques, we show that an elevation of intracellular Ca(2+) concentration by ionomycin increased the FRET efficiency from ECFP (a variant of cyan fluorescent protein)-labeled calmodulin to Venus (a variant of yellow fluorescent protein)-labeled RGS4. The increase in FRET efficiency was greatly attenuated by pre-treating the cells with methyl-beta-cyclodextrin, which depletes membrane cholesterol and thus disrupts lipid rafts. These results provide the first demonstration of a Ca(2+)-dependent interaction between RGS4 and CaM in vivo and show that association in lipid rafts of the plasma membrane might be involved in this physiological regulation of RGS proteins.
Collapse
Affiliation(s)
- Masaru Ishii
- Department of Pharmacology, Graduate School of Medicine, Osaka University, Osaka 565-0871, Japan
| | | | | |
Collapse
|
186
|
Maurice P, Waeckel L, Pires V, Sonnet P, Lemesle M, Arbeille B, Vassy J, Rochette J, Legrand C, Fauvel-Lafève F. The platelet receptor for type III collagen (TIIICBP) is present in platelet membrane lipid microdomains (rafts). Histochem Cell Biol 2005; 125:407-17. [PMID: 16205938 DOI: 10.1007/s00418-005-0076-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/08/2005] [Indexed: 02/02/2023]
Abstract
Platelet interactions with collagen are orchestrated by the presence or the migration of platelet receptor(s) for collagen into lipid rafts, which are specialized lipid microdomains from the platelet plasma membrane enriched in signalling proteins. Electron microscopy shows that in resting platelets, TIIICBP, a receptor specific for type III collagen, is present on the platelet membrane and associated with the open canalicular system, and redistributes to the platelet membrane upon platelet activation. After platelet lysis by 1% Triton X-100 and the separation of lipid rafts on a discontinuous sucrose gradient, TIIICBP is recovered in lipid raft-containing fractions and Triton X-100 insoluble fractions enriched in cytoskeleton proteins. Platelet aggregation, induced by type III collagen, was inhibited after disruption of the lipid rafts by cholesterol depletion, whereas platelet adhesion under static conditions did not require lipid raft integrity. These results indicate that TIIICBP, a platelet receptor involved in platelet interaction with type III collagen, is localized within platelet lipid rafts where it could interact with other platelet receptors for collagen (GP VI and alpha2beta1 integrin) for efficient platelet activation.
Collapse
Affiliation(s)
- Pascal Maurice
- INSERM, U 553, IFR 105, Institut d'Hématologie, Université Paris VII Denis Diderot, 75475, Paris, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
187
|
Hirsch AJ, Medigeshi GR, Meyers HL, DeFilippis V, Früh K, Briese T, Lipkin WI, Nelson JA. The Src family kinase c-Yes is required for maturation of West Nile virus particles. J Virol 2005; 79:11943-51. [PMID: 16140770 PMCID: PMC1212629 DOI: 10.1128/jvi.79.18.11943-11951.2005] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2005] [Accepted: 06/17/2005] [Indexed: 11/20/2022] Open
Abstract
The role of cellular genes in West Nile virus (WNV) replication is not well understood. Examination of cellular transcripts upregulated during WNV infection revealed an increase in the expression of the src family kinase (SFK) c-Yes. WNV-infected cell lines treated with the SFK inhibitor PP2 demonstrated a 2- to 4-log decrease in viral titers, suggesting that SFK activity is required for completion of the viral replication cycle. RNA interference mediated knock-down of c-Yes, but not c-Src, and similarly reduced virus yield, specifically implicating c-Yes in WNV production. Interestingly, PP2 treatment did not reduce intracellular levels of either viral RNA or protein, suggesting that the drug does not act on the early stages of replication. However, endoglycosidase H (endoH) digestion of the viral envelope (E) glycoprotein revealed that the acquisition of endoH-resistant glycans by E, but not endogenous major histocompatibility complex class I, was reduced in PP2-treated cells, demonstrating that E specifically does not traffic beyond the endoplasmic reticulum in the absence of SFK activity. Electron microscopy further revealed that PP2-treated WNV-infected cells accumulated an increased number of virions in the ER compared to untreated cells. Therefore, we conclude that inhibition of SFK activity did not interfere with virus assembly but prevented transit of virions through the secretory pathway. These results identify c-Yes as a cellular protein that is involved in WNV assembly and egress.
Collapse
Affiliation(s)
- Alec J Hirsch
- Vaccine and Gene Therapy Institute, Oregon Health & Sciences University, 505 N.W. 185th Avenue, Beaverton, Oregon 97006, USA.
| | | | | | | | | | | | | | | |
Collapse
|
188
|
Veluthakal R, Chvyrkova I, Tannous M, McDonald P, Amin R, Hadden T, Thurmond DC, Quon MJ, Kowluru A. Essential role for membrane lipid rafts in interleukin-1beta-induced nitric oxide release from insulin-secreting cells: potential regulation by caveolin-1+. Diabetes 2005; 54:2576-85. [PMID: 16123345 DOI: 10.2337/diabetes.54.9.2576] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
We recently reported that the activation of H-Ras represents one of the signaling steps underlying the interleukin-1beta (IL-1beta)-mediated metabolic dysfunction of the islet beta-cell. In the present study, we examined potential contributory roles of membrane-associated, cholesterol-enriched lipid rafts/caveolae and their constituent proteins (e.g., caveolin-1 [Cav-1]) as potential sites for IL-1beta-induced nitric oxide (NO) release in the isolated beta-cell. Disruption of lipid rafts (e.g., with cyclodextrin) markedly reduced IL-1beta-induced gene expression of inducible NO synthase (iNOS) and NO release from beta-cells. Immunologic and confocal microscopic evidence also suggested a transient but significant stimulation of tyrosine phosphorylation of Cav-1 in beta-cells briefly (for 15 min) exposed to IL-1beta that was markedly attenuated by three structurally distinct inhibitors of protein tyrosine phosphorylation. Overexpression of an inactive mutant of Cav-1 lacking the tyrosine phosphorylation site (Y14F) or an siRNA-mediated Cav-1 knock down also resulted in marked attenuation of IL-1beta-induced iNOS gene expression and NO release from these cells, thus further implicating Cav-1 in this signaling cascade. IL-1beta treatment also increased (within 20 min) the translocation of H-Ras into lipid rafts. Here we provide the first evidence to suggest that tyrosine phosphorylation of Cav-1 and subsequent interaction among members of the Ras signaling pathway within the membrane lipid microdomains represent early signaling mechanisms of IL-1beta in beta-cells.
Collapse
Affiliation(s)
- Rajakrishnan Veluthakal
- Department of Pharmaceutical Sciences, Wayne State University, 259 Mack Ave., Detroit, MI 48201, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Silva WI, Maldonado HM, Velázquez G, Rubio-Dávila M, Miranda JD, Aquino E, Mayol N, Cruz-Torres A, Jardón J, Salgado-Villanueva IK. Caveolin isoform expression during differentiation of C6 glioma cells. Int J Dev Neurosci 2005; 23:599-612. [PMID: 16135403 DOI: 10.1016/j.ijdevneu.2005.07.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2005] [Revised: 07/18/2005] [Accepted: 07/19/2005] [Indexed: 12/01/2022] Open
Abstract
Caveolae, a specialized form of lipid rafts, are cholesterol- and sphingolipid-rich membrane microdomains implicated in potocytosis, endocytosis, transcytosis, and as platforms for signal transduction. One of the major constituents of caveolae are three highly homologous caveolin isoforms (caveolin-1, caveolin-2, and caveolin-3). The present study expands the analysis of caveolin isoform expression in C6 glioma cells. Three complementary approaches were used to assess their differential expression during the dibutyryl-cyclic AMP-induced differentiation of C6 cells into an astrocyte-like phenotype. Immunoblotting, conventional RT-PCR, and real-time RT-PCR analysis established the expression of the caveolin-3 isoform in C6 cells, in addition to caveolin-1 and caveolin-2. Similar to the other isoforms, caveolin-3 was associated with light-density, detergent-insoluble caveolae membrane fractions obtained using sucrose-density gradient centrifugation. The three caveolin isoforms display different temporal patterns of mRNA/protein expression during the differentiation of C6 cells. Western blot and real-time RT-PCR analysis demonstrate that caveolin-1 and caveolin-2 are up-regulated during the late stages of the differentiation of C6 cells. Meanwhile, caveolin-3 is gradually down-regulated during the differentiation process. Indirect immunofluorescence analysis via laser-scanning confocal microscopy reveals that the three caveolin isoforms display similar subcellular distribution patterns. In addition, co-localization of caveolin-1/caveolin-2 and caveolin-1/caveolin-3 was detected in both C6 glioma phenotypes. The findings reveal a differential temporal pattern of caveolin gene expression during phenotypic differentiation of C6 glioma cells, with potential implications to developmental and degenerative events in the brain.
Collapse
Affiliation(s)
- W I Silva
- Department of Physiology, School of Medicine, University of Puerto Rico, Medical Sciences Campus, P.O. Box 365067, San Juan, PR.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Rudajev V, Novotny J, Hejnova L, Milligan G, Svoboda P. Dominant Portion of Thyrotropin-Releasing Hormone Receptor Is Excluded from Lipid Domains. Detergent-Resistant and Detergent-Sensitive Pools of TRH Receptor and Gqα/G11α Protein. ACTA ACUST UNITED AC 2005; 138:111-25. [PMID: 16091585 DOI: 10.1093/jb/mvi114] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Some G protein-coupled receptors might be spacially targetted to discrete domains within the plasma membrane. Here we assessed the localization in membrane domains of the epitope-tagged, fluorescent version of thyrotropin-releasing hormone receptor (VSV-TRH-R-GFP) expressed in HEK293 cells. Our comparison of three different methods of cell fractionation (detergent extraction, alkaline treatment/sonication and mechanical homogenization) indicated that the dominant portion of plasma membrane pool of the receptor was totally solubilized by Triton X-100 and its distribution was similar to that of transmembrane plasma membrane proteins (glycosylated and non-glycosylated forms of CD147, MHCI, CD29, CD44, transmembrane form of CD58, Tapa1 and Na,K-ATPase). As expected, caveolin and GPI-bound proteins CD55, CD59 and GPI-bound form of CD58 were preferentially localized in detergent-resistant membrane domains (DRMs). Trimeric G proteins G(q)alpha/G(11)alpha, G(i)alpha1/G(i)alpha2, G(s)alphaL/G(s)alphaS and Gbeta were distributed almost equally between detergent-resistant and detergent-solubilized pools. In contrast, VSV-TRH-R-GFP, Galpha, Gbeta and caveolin were localized massively only in low-density membrane fragments of plasma membranes, which were generated by alkaline treatment/sonication or by mechanical homogenization of cells. These data indicate that VSV-TRH-R-GFP as well as other transmembrane markers of plasma membranes are excluded from TX-100-resistant, caveolin-enriched membrane domains. Trimeric G protein G(q)alpha/G(11)alpha occurs in both DRMs and in the bulk of plasma membranes, which is totally solubilized by TX-100.
Collapse
Affiliation(s)
- Vladimir Rudajev
- Institute of Physiology, Academy of Sciences of the Czech Republic, Videnska 1083, 142 20 Prague 4, Czech Republic
| | | | | | | | | |
Collapse
|
191
|
Mayawala K, Vlachos DG, Edwards JS. Heterogeneities in EGF receptor density at the cell surface can lead to concave up scatchard plot of EGF binding. FEBS Lett 2005; 579:3043-7. [PMID: 15896781 DOI: 10.1016/j.febslet.2005.04.059] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2005] [Revised: 03/30/2005] [Accepted: 04/12/2005] [Indexed: 11/18/2022]
Abstract
The mechanism responsible for the concave up nature of the Scatchard plot of epidermal growth factor (EGF) binding on EGF receptor (EGFR) has been a controversial issue for more than a decade. Past efforts to mechanistically simulate the concave up nature of the Scatchard plot of EGF binding have shown that negative cooperativity in EGF binding on an EGFR dimer or inclusion of some external site or binding event can describe this behavior. However, herein we show that heterogeneity in the density of EGFR due to localization in certain regions of the plasma membrane, which has been experimentally reported, can also lead to concave up shape of the Scatchard plot of the EGF binding on EGFR.
Collapse
Affiliation(s)
- Kapil Mayawala
- Department of Chemical Engineering, University of Delaware, 150 Academy Street, Newark, DE 19716, USA
| | | | | |
Collapse
|
192
|
Abstract
While our understanding of lipid microdomains has advanced in recent years, many aspects of their formation and dynamics are still unclear. In particular, the molecular determinants that facilitate the partitioning of integral membrane proteins into lipid raft domains are yet to be clarified. This review focuses on a family of raft-associated integral membrane proteins, termed flotillins, which belongs to a larger class of integral membrane proteins that carry an evolutionarily conserved domain called the prohibitin homology (PHB) domain. A number of studies now suggest that eucaryotic proteins carrying this domain have affinity for lipid raft domains. The PHB domain is carried by a diverse array of proteins including stomatin, podocin, the archetypal PHB protein, prohibitin, lower eucaryotic proteins such as the Dictyostelium discoideum proteins vacuolin A and vacuolin B and the Caenorhabditis elegans proteins unc-1, unc-24 and mec-2. The presence of this domain in some procaryotic proteins suggests that the PHB domain may constitute a primordial lipid recognition motif. Recent work has provided new insights into the trafficking and targeting of flotillin and other PHB domain proteins. While the function of this large family of proteins remains unclear, studies of the C. elegans PHB proteins suggest possible links to a class of volatile anaesthetics raising the possibility that these lipophilic agents could influence lipid raft domains. This review will discuss recent insights into the cell biology of flotillins and the large family of evolutionarily conserved PHB domain proteins.
Collapse
Affiliation(s)
- Isabel C Morrow
- Institute for Molecular Bioscience, Centre for Microscopy and Microanalysis, University of Queensland, Brisbane, Queensland 4072, Australia
| | | |
Collapse
|
193
|
Wang HQ, Altomare DA, Skele KL, Poulikakos PI, Kuhajda FP, Di Cristofano A, Testa JR. Positive feedback regulation between AKT activation and fatty acid synthase expression in ovarian carcinoma cells. Oncogene 2005; 24:3574-82. [PMID: 15806173 DOI: 10.1038/sj.onc.1208463] [Citation(s) in RCA: 146] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Activation of AKT and overexpression of fatty acid synthase (FAS) are frequently observed in human ovarian cancer. To explore a possible connection between AKT and FAS, immunohistochemical analyses were conducted on an ovarian cancer tissue microarray, which revealed a significant correlation between phosphorylated AKT (phospho-AKT) and expression of FAS. To investigate the relationship between phospho-AKT and FAS in vitro, a variety of experiments employing a specific phosphatidylinositol 3-OH kinase (PI3K) inhibitor (LY294002), inducible PTEN expression in PTEN-null cells, or AKT1 siRNA demonstrated that phosphatidylinositol-3 kinase (PI3K)/AKT signaling modulates FAS expression. In contrast, inhibition of FAS activity by the drug C75 resulted in downregulation of phospho-AKT and increased cell death. To explore the functional relationship between phospho-AKT and FAS, we used SKOV3, C200, and OVCAR10 ovarian carcinoma cells, which have constitutively active AKT, and OVCAR5 cells, which have very low basal phospho-AKT levels. Treatment with LY294002 abolished AKT activity and potentiated apoptosis induced by FAS inhibitors cerulenin or C75 only in cells with constitutively active AKT, suggesting that constitutive activation of AKT protects against FAS inhibitor-induced cell death. Furthermore, inhibition of FAS activity by cerulenin or C75 resulted in downregulation of phospho-AKT, which preceded the induction of apoptosis. To investigate the relationship between phospho-AKT and FAS in vivo, severe combined immunodeficient mice injected intraperitoneally with SKOV3 cells were treated with C75. Growth of SKOV3 xenografts was markedly inhibited by C75. Analysis of the levels of phospho-AKT and FAS in C75-treated tumors revealed concordant downregulation of phospho-AKT and FAS. Collectively, our findings are consistent with a working model in which AKT activation regulates FAS expression, at least in part, whereas FAS activity modulates AKT activation.
Collapse
Affiliation(s)
- Hui Qin Wang
- Human Genetics Program, Fox Chase Cancer Center, 333 Cottman Avenue, Philadelphia, PA 19111-2497, USA
| | | | | | | | | | | | | |
Collapse
|
194
|
Cho KA, Park SC. Caveolin-1 as a prime modulator of aging: a new modality for phenotypic restoration? Mech Ageing Dev 2005; 126:105-10. [PMID: 15610768 DOI: 10.1016/j.mad.2004.09.029] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Aging can be characterized by structural changes and functional deterioration during the lifetime, for which hundreds of explanations have been put forward. Recently, we have proposed the gate theory of aging, in which gatekeeper molecules at the membrane level would play the prime role in determining the senescent phenotype. Caveolin-1 would be a prime candidate for such a role as a major determinant of the aging process. Caveolin-1 can associate with a variety of molecules, involved in signal transduction, endocytosis and transcytosis, cytoskeletal arrangement, etc. The level of caveolin-1 is strictly regulated to maintain cellular integrity, leading to cellular transformation if depleted, and to the senescent phenotype if overexpressed. In case of senescent cells, the functional and physiological responses to the mitogenic stimuli can be restored and the morphological shape can be resumed by simple adjustment of caveolin-1 status. Therefore, it is suggested that prime modulator molecules, represented by caveolin-1, play a key role in determining the senescent phenotype, either as a physiological response or altered morphology.
Collapse
Affiliation(s)
- Kyung A Cho
- Department of Biochemistry and Molecular Biology, Aging and Apoptosis Research Center, Seoul National University College of Medicine, 28 Yungon Dong, Chong No Ku, Seoul 110-799, South Korea
| | | |
Collapse
|
195
|
Ciana A, Balduini C, Minetti G. Detergent-resistant membranes in human erythrocytes and their connection to the membrane-skeleton. J Biosci 2005; 30:317-28. [PMID: 16052070 DOI: 10.1007/bf02703669] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
In cell membranes, local inhomogeneity in the lateral distribution of lipids and proteins is thought to exist in vivo in the form of lipid 'rafts', microdomains enriched in cholesterol and sphingolipids, and in specific classes of proteins, that appear to play specialized roles for signal transduction, cell-cell recognition, parasite or virus infection, and vesicular trafficking. These structures are operationally defined as membranes resistant to solubilization by nonionic detergents at 4 degree C (detergent-resistant membranes, DRMs). This definition appears to be necessary and sufficient, although additional manoeuvres, not always described with sufficient detail, may be needed to ensure isolation of DRMs, like mechanical homogenization, and changes in the pH and/or ionic strength of the solubilization medium. We show here for the human erythrocyte that the different conditions adopted may lead to the isolation of qualitatively and quantitatively different DRM fractions, thus contributing to the complexity of the notion itself of lipid raft. A significant portion of erythrocyte DRMs enriched in reported lipid raft markers, such as flotillin-1, flotillin-2 and GM1, is anchored to the spectrin membrane-skeleton via electrostatic interactions that can be disrupted by the simultaneous increase in pH and ionic strength of the solubilization medium.
Collapse
Affiliation(s)
- Annarita Ciana
- Università di Pavia, Dipartimento di Biochimica "A. Castellani", via Bassi, 21, 27100 Pavia, Italy
| | | | | |
Collapse
|
196
|
D'Alessio A, Al-Lamki RS, Bradley JR, Pober JS. Caveolae participate in tumor necrosis factor receptor 1 signaling and internalization in a human endothelial cell line. THE AMERICAN JOURNAL OF PATHOLOGY 2005; 166:1273-82. [PMID: 15793306 PMCID: PMC1602396 DOI: 10.1016/s0002-9440(10)62346-2] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Caveolae are abundant in endothelial cells (ECs) in situ but markedly diminished in cultured cells, making it difficult to assess their role in cytokine signaling. We report here that the human EC line EA.hy926 retains an abundant caveolar system in culture. Tumor necrosis factor (TNF) receptor 1 (TNFR1/CD120a) was enriched in caveolae and co-immunoprecipitated with caveolin-1 from caveolae isolated from these cells. To further investigate the role(s) of caveolae in TNF signaling in ECs, cells were treated with methyl-beta-cyclodextrin to disrupt caveolae. Methyl-beta-cyclodextrin did not alter total cell surface expression of TNFR1 or TNF-induced degradation of IkappaBalpha, a measure of nuclear factor-kappaB activation, but it did inhibit TNF-induced phosphorylation of Akt, a measure of phosphatidylinositol-3 kinase activation. Serum-induced phosphorylation of AKT was unaffected. Treatment with TNF induced disappearance of TNFR1 from caveolae and dissociation from caveolin-1 within 5 minutes. In contrast to transferrin receptor, internalized TNFR1 did not co-localize with clathrin, except possibly in the Golgi, at any time point examined. By 60 minutes of treatment with TNF, TNFR1 appeared in endosomes. We conclude that caveolae function in ECs to allow TNFR1 to activate phosphatidylinositol-3 kinase and Akt, perhaps through receptor cross talk, and that ligand-induced internalization and trafficking of TNFR1 to endosomes may originate directly from this compartment.
Collapse
MESH Headings
- Caveolae/drug effects
- Caveolae/metabolism
- Caveolae/ultrastructure
- Cell Line
- Endothelium, Vascular/cytology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/ultrastructure
- Flow Cytometry
- Humans
- Immunoblotting
- Immunoprecipitation
- Microscopy, Confocal
- Microscopy, Electron, Transmission
- Phosphatidylinositol 3-Kinases/metabolism
- Protein Serine-Threonine Kinases/metabolism
- Protein Transport/physiology
- Proto-Oncogene Proteins/metabolism
- Proto-Oncogene Proteins c-akt
- Receptor Cross-Talk/physiology
- Receptors, Tumor Necrosis Factor, Type I/drug effects
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Signal Transduction/drug effects
- Signal Transduction/physiology
- Tumor Necrosis Factor-alpha/pharmacology
- beta-Cyclodextrins/pharmacology
Collapse
Affiliation(s)
- Alessio D'Alessio
- Interdepartmental Program in Vascular Biology and Transplantation, Boyer Center for Molecular Medicine, Yale University School of Medicine, 295 Congress Ave., New Haven, CT 06536-0812, USA
| | | | | | | |
Collapse
|
197
|
Yu P, Yang Z, Jones JE, Wang Z, Owens SA, Mueller SC, Felder RA, Jose PA. D1 dopamine receptor signaling involves caveolin-2 in HEK-293 cells. Kidney Int 2005; 66:2167-80. [PMID: 15569306 DOI: 10.1111/j.1523-1755.2004.66007.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Dopamine receptors in the kidney, especially those belonging to the D1-like receptor family, are important in the regulation of renal function and blood pressure. Because of increasing evidence that G protein-coupled receptors (GPCRs) are associated with caveolae and lipid rafts, we tested the hypothesis that the D1 dopamine receptor (D1R) and signaling molecules are regulated by caveolin in caveolae or lipid rafts. METHODS Six experimental approaches were used: (1) construction of tagged human D1Rs (hD1Rs) and transfectants; (2) cell culture [human embryonic kidney (HEK)-293 and immortalized rat renal proximal tubule cells] and biotinylation; (3) cell fractionation by sucrose gradient centrifugation; (4) immunoprecipitation and immunoblotting; (5) immunofluorescence and confocal microscopy; and (6) adenylyl cyclase assays. RESULTS hD1Rs, heterologously expressed in HEK-293 cells, formed protein species with molecular mass ranging from 50 to 250 kD, and were localized in lipid rafts and nonraft plasma membranes. The hD1Rs cofractionated with caveolin-2, G protein subunits, and several signaling molecules. Both exogenously expressed hD1Rs and endogenously expressed rat D1Rs colocalized and coimmunoprecipitated with caveolin-2. A D1R agonist (fenoldopam) increased the amount of caveolin-2beta associated with hD1Rs and activated adenylyl cyclase to a greater extent in lipid rafts than in nonraft plasma membranes. Reduction in the expression of caveolin-2 with antisense oligonucleotides attenuated the stimulatory effect of fenoldopam on cyclic adenosine monophosphate (cAMP) accumulation. CONCLUSION The majority of hD1Rs are distributed in lipid rafts. Heterologously and endogenously expressed D1Rs in renal cells are associated with and regulated by caveolin-2.
Collapse
Affiliation(s)
- Peiying Yu
- Department of Pediatrics, Georgetown University School of Medicine, Washington, D.C., USA.
| | | | | | | | | | | | | | | |
Collapse
|
198
|
Williams TM, Lisanti MP. Caveolin-1 in oncogenic transformation, cancer, and metastasis. Am J Physiol Cell Physiol 2005; 288:C494-506. [PMID: 15692148 DOI: 10.1152/ajpcell.00458.2004] [Citation(s) in RCA: 411] [Impact Index Per Article: 21.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
Caveolae are 50- to 100-nm omega-shaped invaginations of the plasma membrane that function as regulators of signal transduction. Caveolins are a class of oligomeric structural proteins that are both necessary and sufficient for caveolae formation. Interestingly, caveolin-1 has been implicated in the pathogenesis of oncogenic cell transformation, tumorigenesis, and metastasis. Here, we review the available experimental evidence (gleaned from cultured cells, animal models, and human tumor samples) that caveolin-1 (Cav-1) functions as a "tumor and/or metastasis modifier gene." Genetic evidence from the study of Cav-1(-/-) null mice and human breast cancer mutations [CAV-1 (P132L)] supports the idea that caveolin-1 normally functions as a negative regulator of cell transformation and mammary tumorigenesis. In contrast, caveolin-1 may function as a tumor promoter in prostate cancers. We discuss possible molecular mechanisms to explain these intriguing, seemingly opposing, findings. More specifically, caveolin-1 phosphorylation (at Tyr14 and Ser80) and mutations (P132L) may override or inactivate the growth inhibitory activity of the caveolin-scaffolding domain (residues 82-101).
Collapse
Affiliation(s)
- Terence M Williams
- Department of Molecular Pharmacology, and The Albert Einstein Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | | |
Collapse
|
199
|
McDonald TP, Pitt AR, Brown G, Rixon HWM, Sugrue RJ. Evidence that the respiratory syncytial virus polymerase complex associates with lipid rafts in virus-infected cells: a proteomic analysis. Virology 2005; 330:147-57. [PMID: 15527841 DOI: 10.1016/j.virol.2004.09.034] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2004] [Revised: 08/31/2004] [Accepted: 09/28/2004] [Indexed: 10/26/2022]
Abstract
The interaction between the respiratory syncytial virus (RSV) polymerase complex and lipid rafts was examined in HEp2 cells. Lipid-raft membranes were prepared from virus-infected cells and their protein content was analysed by Western blotting and mass spectrometry. This analysis revealed the presence of the N, P, L, M2-1 and M proteins. However, these proteins appeared to differ from one another in their association with these structures, with the M2-1 protein showing a greater partitioning into raft membranes compared to that of the N, P or M proteins. Determination of the polymerase activity profile of the gradient fractions revealed that 95% of the detectable viral enzyme activity was associated with lipid-raft membranes. Furthermore, analysis of virus-infected cells by confocal microscopy suggested an association between these proteins and the raft-lipid, GM1. Together, these results provide evidence that the RSV polymerase complex is able to associate with lipid rafts in virus-infected cells.
Collapse
|
200
|
Cavallo-Medved D, Mai J, Dosescu J, Sameni M, Sloane BF. Caveolin-1 mediates the expression and localization of cathepsin B, pro-urokinase plasminogen activator and their cell-surface receptors in human colorectal carcinoma cells. J Cell Sci 2005; 118:1493-503. [PMID: 15769846 DOI: 10.1242/jcs.02278] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Cathepsin B and pro-urokinase plasminogen activator (pro-uPA) localize to the caveolae of HCT 116 human colorectal carcinoma cells, an association mediated by active K-RAS. In this study, we established a stable HCT 116 cell line with a gene encoding antisense caveolin-1 (AS-cav-1) to examine the effects of caveolin-1, the main structural protein of caveolae, on the expression and localization of cathepsin B and pro-uPA, and their cell-surface receptors p11 and uPA receptor (uPAR), respectively. AS-cav-1 HCT 116 cells secreted less procathepsin B than control (empty vector) cells as measured by immunoblotting and pepsin activation of the proenzyme. Expression and secretion of pro-uPA was also downregulated in AS-cav-1 HCT 116 cells. Localization of cathepsin B and pro-uPA to caveolae was reduced in AS-cav-1 HCT 116 cells, and these cells expressed less total and caveolae-associated p11 and uPAR compared with control cells. Previous studies have shown that uPAR forms a complex with caveolin-1 and beta1-integrin, and we here show that downregulation of caveolin-1 also suppressed the localization of beta1-integrin to caveolae of these cells. Finally, downregulation of caveolin-1 in HCT 116 cells inhibited degradation of the extracellular matrix protein collagen IV and the invasion of these cells through Matrigel. Based on these results, we hypothesize that caveolin-1 affects the expression and localization of cathepsin B and pro-uPA, and their receptors, thereby mediating cell-surface proteolytic events associated with invasion of colon cancer cells.
Collapse
Affiliation(s)
- Dora Cavallo-Medved
- Department of Pharmacology, Wayne State University, School of Medicine, Detroit, MI 48201, USA.
| | | | | | | | | |
Collapse
|