1
|
Bai X, Smith HE, Golden A. Identification of genetic suppressors for a BSCL2 lipodystrophy pathogenic variant in Caenorhabditis elegans. Dis Model Mech 2024; 17:dmm050524. [PMID: 38454882 PMCID: PMC11051982 DOI: 10.1242/dmm.050524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 03/04/2024] [Indexed: 03/09/2024] Open
Abstract
Seipin (BSCL2), a conserved endoplasmic reticulum protein, plays a critical role in lipid droplet (LD) biogenesis and in regulating LD morphology, pathogenic variants of which are associated with Berardinelli-Seip congenital generalized lipodystrophy type 2 (BSCL2). To model BSCL2 disease, we generated an orthologous BSCL2 variant, seip-1(A185P), in Caenorhabditis elegans. In this study, we conducted an unbiased chemical mutagenesis screen to identify genetic suppressors that restore embryonic viability in the seip-1(A185P) mutant background. A total of five suppressor lines were isolated and recovered from the screen. The defective phenotypes of seip-1(A185P), including embryonic lethality and impaired eggshell formation, were significantly suppressed in each suppressor line. Two of the five suppressor lines also alleviated the enlarged LDs in the oocytes. We then mapped a suppressor candidate gene, lmbr-1, which is an ortholog of human limb development membrane protein 1 (LMBR1). The CRISPR/Cas9 edited lmbr-1 suppressor alleles, lmbr-1(S647F) and lmbr-1(P314L), both significantly suppressed embryonic lethality and defective eggshell formation in the seip-1(A185P) background. The newly identified suppressor lines offer valuable insights into potential genetic interactors and pathways that may regulate seipin in the lipodystrophy model.
Collapse
Affiliation(s)
- Xiaofei Bai
- Department of Biology, University of Florida, Gainesville, FL 32610, USA
- Genetics Institute, University of Florida, Gainesville, FL 32610, USA
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Harold E. Smith
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Andy Golden
- National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
2
|
Bai X, Smith HE, Golden A. Identification of Genetic Suppressors for a Berardinelli-Seip Congenital Generalized Lipodystrophy Type 2 (BSCL2) Pathogenic Variant in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.09.22.559059. [PMID: 37790539 PMCID: PMC10542546 DOI: 10.1101/2023.09.22.559059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Maintaining the metabolic homeostasis of fatty acids is crucial for human health. Excess fatty acids are stored in lipid droplets (LDs), the primary energy reservoir that helps regulate fat and lipid homeostasis in nearly all cell types. Seipin (BSCL2), a conserved endoplasmic reticulum protein, plays a critical role in LD biogenesis and regulating LD morphology. Pathogenic variants of seipin are associated with multiple human genetic diseases, including Berardinelli-Seip Congenital Generalized Lipodystrophy Type 2 (BSCL2). However, the cellular and molecular mechanisms by which dysfunctional seipin leads to these diseases remain unclear. To model BSCL2 disease, we generated an orthologous BSCL2 pathogenic variant seip-1(A185P) using CRISPR/Cas9 genome editing in Caenorhabditis elegans . This variant led to severe developmental and cellular defects, including embryonic lethality, impaired eggshell formation, and abnormally enlarged LDs. We set out to identify genetic determinants that could suppress these defective phenotypes in the seip-1(A185P) mutant background. To this end, we conducted an unbiased chemical mutagenesis screen to identify genetic suppressors that restore embryonic viability in the seip-1(A185P) mutant background. A total of five suppressor lines were isolated and recovered from the screen. The defective phenotypes of seip-1(A185P) , including embryonic lethality and impaired eggshell formation, were significantly suppressed in each suppressor line. Two of the five suppressor lines also alleviated the enlarged LDs in the oocytes. We then mapped a suppressor candidate gene, R05D3.2 (renamed as lmbr-1 ), which is an ortholog of human LMBR1 (limb development membrane protein 1). The CRISPR/Cas9 edited lmbr-1 suppressor alleles, lmbr-1(Ser647Phe) and lmbr-1(Pro314Leu) , both significantly suppressed embryonic lethality and defective eggshell formation in the seip-1(A185P) background. The newly identified suppressor lines offer valuable insights into potential genetic interactors and pathways that may regulate seipin in the lipodystrophy model.
Collapse
|
3
|
Abstract
Caveolin-1 (CAV1) is commonly considered to function as a cell surface protein, for instance in the genesis of caveolae. Nonetheless, it is also present in many intracellular organelles and compartments. The contributions of these intracellular pools to CAV1 function are generally less well understood, and this is also the case in the context of cancer. This review will summarize literature available on the role of CAV1 in cancer, highlighting particularly our understanding of the canonical (CAV1 in the plasma membrane) and non-canonical pathways (CAV1 in organelles and exosomes) linked to the dual role of the protein as a tumor suppressor and promoter of metastasis. With this in mind, we will focus on recently emerging concepts linking CAV1 function to the regulation of intracellular organelle communication within the same cell where CAV1 is expressed. However, we now know that CAV1 can be released from cells in exosomes and generate systemic effects. Thus, we will also elaborate on how CAV1 participates in intracellular communication between organelles as well as signaling between cells (non-canonical pathways) in cancer.
Collapse
|
4
|
Pol A, Morales-Paytuví F, Bosch M, Parton RG. Non-caveolar caveolins – duties outside the caves. J Cell Sci 2020; 133:133/9/jcs241562. [DOI: 10.1242/jcs.241562] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
ABSTRACT
Caveolae are invaginations of the plasma membrane that are remarkably abundant in adipocytes, endothelial cells and muscle. Caveolae provide cells with resources for mechanoprotection, can undergo fission from the plasma membrane and can regulate a variety of signaling pathways. Caveolins are fundamental components of caveolae, but many cells, such as hepatocytes and many neurons, express caveolins without forming distinguishable caveolae. Thus, the function of caveolins goes beyond their roles as caveolar components. The membrane-organizing and -sculpting capacities of caveolins, in combination with their complex intracellular trafficking, might contribute to these additional roles. Furthermore, non-caveolar caveolins can potentially interact with proteins normally excluded from caveolae. Here, we revisit the non-canonical roles of caveolins in a variety of cellular contexts including liver, brain, lymphocytes, cilia and cancer cells, as well as consider insights from invertebrate systems. Non-caveolar caveolins can determine the intracellular fluxes of active lipids, including cholesterol and sphingolipids. Accordingly, caveolins directly or remotely control a plethora of lipid-dependent processes such as the endocytosis of specific cargoes, sorting and transport in endocytic compartments, or different signaling pathways. Indeed, loss-of-function of non-caveolar caveolins might contribute to the common phenotypes and pathologies of caveolin-deficient cells and animals.
Collapse
Affiliation(s)
- Albert Pol
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010, Barcelona, Spain
| | - Frederic Morales-Paytuví
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
| | - Marta Bosch
- Cell Compartments and Signaling Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Universitat de Barcelona, 08036, Barcelona, Spain
- Department of Biomedical Sciences, Faculty of Medicine, Universitat de Barcelona, 08036, Barcelona, Spain
| | - Robert G. Parton
- Institute for Molecular Bioscience (IMB), The University of Queensland (UQ), Brisbane, Queensland 4072, Australia
- Centre for Microscopy and Microanalysis (CMM) IMB, The University of Queensland (UQ), Brisbane, Queensland 4072, Australia
| |
Collapse
|
5
|
Viau C, Haçariz O, Karimian F, Xia J. Comprehensive phenotyping and transcriptome profiling to study nanotoxicity in C. elegans. PeerJ 2020; 8:e8684. [PMID: 32149031 PMCID: PMC7049462 DOI: 10.7717/peerj.8684] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 02/04/2020] [Indexed: 12/11/2022] Open
Abstract
Engineered nanoparticles are used at an increasing rate in both industry and medicine without fully understanding their impact on health and environment. The nematode Caenorhabditis elegans is a suitable model to study the toxic effects of nanoparticles as it is amenable to comprehensive phenotyping, such as locomotion, growth, neurotoxicity and reproduction. In this study, we systematically evaluated the effects of silver (Ag) and five metal oxide nanoparticles: SiO2, CeO2, CuO, Al2O3 and TiO2. The results showed that Ag and SiO2 exposures had the most toxic effects on locomotion velocity, growth and reproduction, whereas CeO2, Al2O3 and CuO exposures were mostly neurotoxic. We further performed RNAseq to compare the gene expression profiles underlying Ag and SiO2toxicities. Gene set enrichment analyses revealed that exposures to Ag and SiO2consistently downregulated several biological processes (regulations in locomotion, reproductive process and cell growth) and pathways (neuroactive ligand-receptor interaction, wnt and MAPK signaling, etc.), with opposite effects on genes involved in innate immunity. Our results contribute to mechanistic insights into toxicity of Ag and SiO2 nanoparticles and demonstrated that C. elegans as a valuable model for nanotoxicity assessment.
Collapse
Affiliation(s)
- Charles Viau
- Institute of Parasitology, McGill University, Montreal, Canada
| | - Orçun Haçariz
- Institute of Parasitology, McGill University, Montreal, Canada
| | - Farial Karimian
- Institute of Parasitology, McGill University, Montreal, Canada
| | - Jianguo Xia
- Institute of Parasitology, McGill University, Montreal, Canada.,Department of Animal Science, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
6
|
Dubois C, Pophillat M, Audebert S, Fourquet P, Lecomte C, Dubourg N, Galas S, Camoin L, Frelon S. Differential modification of the C. elegans proteome in response to acute and chronic gamma radiation: Link with reproduction decline. THE SCIENCE OF THE TOTAL ENVIRONMENT 2019; 676:767-781. [PMID: 31055208 DOI: 10.1016/j.scitotenv.2019.04.039] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 03/28/2019] [Accepted: 04/03/2019] [Indexed: 06/09/2023]
Abstract
Emission of ionizing radiation (IR) in the environment is a natural phenomenon which can be enhanced by human activities. Ecosystems are then chronically exposed to IR. But environmental risk assessment of chronic exposure suffers from a lack of knowledge. Extrapolation of data from acute to chronic exposure is not always relevant, and can lead to uncertainties as effects could be different between the two irradiation modes, especially regarding reproduction endpoint, which is an ecologically relevant parameter. In the present study, we decided to refine the understanding of the molecular mechanisms involved in response to acute and chronic γ-irradiation by a global proteome label free LC-MS/MS analysis. C. elegans were exposed to 3 common cumulated radiation doses for acute or chronic exposure condition and global modification of the proteome was studied. This analysis of protein expression has demonstrated the modulation of proteins involved in regulatory biological processes such as lipid transport, DNA replication, germ cell development, apoptosis, ion transport, cuticle development, and aging at lower doses than those for which individual effects on reproduction have been previously observed. Thus, these proteins could constitute early and sensitive markers of radio-induced reprotoxicity; more specifically HAT-1, RPS-19 in acute and VIT-3 for chronic conditions that are expressed in a dose-dependent manner. Finally, to focus on reproduction process, this analysis showed either repression or overexpression of 12 common proteins in organisms exposed to acute or chronic irradiation, respectively. These proteins include the vitellogenin cluster notably involved in lipid transport and oocyte maturation and proteins involved in cuticle development and molting i.e. COL-14, GLF-1, NOAH-1, NOAH-2, ACN-1. These results show that protein expression modulation is a sensitive and predictive marker of radio-induced reproductive effects, but also highlight limitation of data extrapolation from acute to chronic exposure for environmental risk assessment.
Collapse
Affiliation(s)
- Cécile Dubois
- IRSN/PSE-ENV/SRTE, Laboratoire d'ecotoxicologie des radionucléides, BP3 - 13115 St Paul lez Durance Cedex, France
| | - Matthieu Pophillat
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Stéphane Audebert
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Patrick Fourquet
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France
| | - Catherine Lecomte
- IRSN/PSE-ENV/SRTE, Laboratoire d'ecotoxicologie des radionucléides, BP3 - 13115 St Paul lez Durance Cedex, France
| | - Nicolas Dubourg
- IRSN/PSE-ENV/SRTE, Laboratoire d'ecotoxicologie des radionucléides, BP3 - 13115 St Paul lez Durance Cedex, France
| | - Simon Galas
- IBMM, University of Montpellier, CNRS, ENSCM, Montpellier, France
| | - Luc Camoin
- Aix-Marseille Univ, Inserm, CNRS, Institut Paoli-Calmettes, CRCM, Marseille Protéomique, Marseille, France.
| | - Sandrine Frelon
- IRSN/PSE-ENV/SRTE, Laboratoire d'ecotoxicologie des radionucléides, BP3 - 13115 St Paul lez Durance Cedex, France.
| |
Collapse
|
7
|
Okada S, Raja SA, Okerblom J, Boddu A, Horikawa Y, Ray S, Okada H, Kawamura I, Murofushi Y, Murray F, Patel HH. Deletion of caveolin scaffolding domain alters cancer cell migration. Cell Cycle 2019; 18:1268-1280. [PMID: 31116089 DOI: 10.1080/15384101.2019.1618118] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Caveolin-1 (Cav-1) is an integral membrane protein that plays an important role in proliferative and terminally differentiated cells. As a structural component of Caveolae, Cav-1 interacts with signaling molecules via a caveolin scaffolding domain (CSD) regulating cell signaling. Recent reports have shown that Cav-1 is a negative regulator in tumor metastasis. Therefore, we hypothesize that Cav-1 inhibits cell migration through its CSD. HeLa cells were engineered to overexpress Cav-1 (Cav-1 OE), Cav-1 without a functional CSD (∆CSD), or enhanced green fluorescent protein (EGFP) as a control. HeLa cell migration was suppressed in Cav-1 OE cells while ∆CSD showed increased migration, which corresponded to a decrease in the tight junction protein, zonula occludens (ZO-1). The migration phenotype was confirmed in multiple cancer cell lines. Phosphorylated STAT-3 was decreased in Cav-1 OE cells compared to control and ∆CSD cells; reducing STAT-3 expression alone decreased cell migration. ∆CSD blunted HeLa proliferation by increasing the number of cells in the G2/M phase of the cell cycle. Overexpressing the CSD peptide alone suppressed HeLa cell migration and inhibited pSTAT3. These findings suggest that Cav-1 CSD may be critical in controlling the dynamic phenotype of cancer cells by facilitating the interaction of specific signal transduction pathways, regulating STAT3 and participating in a G2/M checkpoint. Modulating the CSD and targeting specific proteins may offer potential new therapies in the treatment of cancer metastasis.
Collapse
Affiliation(s)
- Sunaho Okada
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,b Department of Anesthesiology and UCSD School of Medicine , San Diego , CA , USA
| | - Sadaf A Raja
- c Department of Biosciences , COMSATS Institute of Information Technology , Islamabad , Pakistan
| | - Jonathan Okerblom
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,b Department of Anesthesiology and UCSD School of Medicine , San Diego , CA , USA
| | - Aayush Boddu
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,b Department of Anesthesiology and UCSD School of Medicine , San Diego , CA , USA
| | - Yousuke Horikawa
- d Department of Pediatrics , Sharp Rees-Stealy Medical Group , San Diego , CA , USA.,e Department of Anesthesiology , Tokushima University , Tokushima , Japan
| | | | - Hideshi Okada
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,g Department of Anesthesiology and Medicine , UCSD School of Medicine , San Diego , CA , USA.,h Department of Emergency and Disaster Medicine , Gifu University Graduate School of Medicine , Gifu , Japan
| | - Itta Kawamura
- i Department of Cardiovascular Medicine , Gifu Heart Center , Gifu , Japan
| | - Yoshiteru Murofushi
- g Department of Anesthesiology and Medicine , UCSD School of Medicine , San Diego , CA , USA
| | - Fiona Murray
- j Aberdeen Cardiovascular & Diabetes Centre, School of Medicine, Medical Sciences & Nutrition, Institute of Medical Sciences , University of Aberdeen , Aberdeen , Scotland
| | - Hemal H Patel
- a Veterans Administration San Diego Healthcare System , San Diego , CA , USA.,b Department of Anesthesiology and UCSD School of Medicine , San Diego , CA , USA
| |
Collapse
|
8
|
Roitenberg N, Bejerano-Sagie M, Boocholez H, Moll L, Marques FC, Golodetzki L, Nevo Y, Elami T, Cohen E. Modulation of caveolae by insulin/IGF-1 signaling regulates aging of Caenorhabditis elegans. EMBO Rep 2018; 19:embr.201745673. [PMID: 29945933 DOI: 10.15252/embr.201745673] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Revised: 05/27/2018] [Accepted: 05/29/2018] [Indexed: 11/09/2022] Open
Abstract
Reducing insulin/IGF-1 signaling (IIS) extends lifespan, promotes protein homeostasis (proteostasis), and elevates stress resistance of worms, flies, and mammals. How these functions are orchestrated across the organism is only partially understood. Here, we report that in the nematode Caenorhabditis elegans, the IIS positively regulates the expression of caveolin-1 (cav-1), a gene which is primarily expressed in neurons of the adult worm and underlies the formation of caveolae, a subtype of lipid microdomains that serve as platforms for signaling complexes. Accordingly, IIS reduction lowers cav-1 expression and lessens the quantity of neuronal caveolae. Reduced cav-1 expression extends lifespan and mitigates toxic protein aggregation by modulating the expression of aging-regulating and signaling-promoting genes. Our findings define caveolae as aging-governing signaling centers and underscore the potential for cav-1 as a novel therapeutic target for the promotion of healthy aging.
Collapse
Affiliation(s)
- Noa Roitenberg
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel - Canada, The Hebrew University School of Medicine, Jerusalem, Israel
| | - Michal Bejerano-Sagie
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel - Canada, The Hebrew University School of Medicine, Jerusalem, Israel
| | - Hana Boocholez
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel - Canada, The Hebrew University School of Medicine, Jerusalem, Israel
| | - Lorna Moll
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel - Canada, The Hebrew University School of Medicine, Jerusalem, Israel
| | - Filipa Carvalhal Marques
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel - Canada, The Hebrew University School of Medicine, Jerusalem, Israel
| | - Ludmila Golodetzki
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel - Canada, The Hebrew University School of Medicine, Jerusalem, Israel
| | - Yuval Nevo
- Computation Center, Hebrew University-Hadassah Medical School, Jerusalem, Israel
| | - Tayir Elami
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel - Canada, The Hebrew University School of Medicine, Jerusalem, Israel
| | - Ehud Cohen
- Department of Biochemistry and Molecular Biology, The Institute for Medical Research Israel - Canada, The Hebrew University School of Medicine, Jerusalem, Israel
| |
Collapse
|
9
|
Scholze MJ, Barbieux KS, De Simone A, Boumasmoud M, Süess CCN, Wang R, Gönczy P. PI(4,5)P 2 forms dynamic cortical structures and directs actin distribution as well as polarity in Caenorhabditis elegans embryos. Development 2018; 145:dev.164988. [PMID: 29724757 DOI: 10.1242/dev.164988] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2018] [Accepted: 04/19/2018] [Indexed: 01/25/2023]
Abstract
Asymmetric division is crucial for embryonic development and stem cell lineages. In the one-cell Caenorhabditis elegans embryo, a contractile cortical actomyosin network contributes to asymmetric division by segregating partitioning-defective (PAR) proteins to discrete cortical domains. In the current study, we found that the plasma membrane lipid phosphatidylinositol 4,5-bisphosphate (PIP2) localizes to polarized dynamic structures in C. elegans zygotes, distributing in a PAR-dependent manner along the anterior-posterior (A-P) embryonic axis. PIP2 cortical structures overlap with F-actin, and coincide with the actin regulators RHO-1 and CDC-42, as well as ECT-2. Particle image velocimetry analysis revealed that PIP2 and F-actin cortical movements are coupled, with PIP2 structures moving slightly ahead of F-actin. Importantly, we established that PIP2 cortical structure formation and movement is actin dependent. Moreover, we found that decreasing or increasing the level of PIP2 resulted in severe F-actin disorganization, revealing interdependence between these components. Furthermore, we determined that PIP2 and F-actin regulate the sizing of PAR cortical domains, including during the maintenance phase of polarization. Overall, our work establishes that a lipid membrane component, PIP2, modulates actin organization and cell polarity in C. elegans embryos.
Collapse
Affiliation(s)
- Melina J Scholze
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| | - Kévin S Barbieux
- Geodetic Engineering Laboratory (TOPO), Swiss Federal Institute of Technology (EPFL), Environmental Engineering Institute (IIE), CH-1015 Lausanne, Switzerland
| | - Alessandro De Simone
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| | - Mathilde Boumasmoud
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| | - Camille C N Süess
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| | - Ruijia Wang
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| | - Pierre Gönczy
- Swiss Institute for Experimental Cancer Research (ISREC), School of Life Sciences, Swiss Federal Institute of Technology (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
10
|
Jung W, Sierecki E, Bastiani M, O'Carroll A, Alexandrov K, Rae J, Johnston W, Hunter DJB, Ferguson C, Gambin Y, Ariotti N, Parton RG. Cell-free formation and interactome analysis of caveolae. J Cell Biol 2018; 217:2141-2165. [PMID: 29716956 PMCID: PMC5987714 DOI: 10.1083/jcb.201707004] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 01/24/2018] [Accepted: 03/29/2018] [Indexed: 02/07/2023] Open
Abstract
Caveolae are linked to signaling protein regulation through interactions with caveolins. We describe a cell-free system for the biogenesis of caveolae and show phosphorylated-caveolins preferentially bind signaling proteins. Our validation in vivo shows that phosphorylated CAV1 recruits TRAF2 to the endosome to form a signaling platform. Caveolae have been linked to the regulation of signaling pathways in eukaryotic cells through direct interactions with caveolins. Here, we describe a cell-free system based on Leishmania tarentolae (Lt) extracts for the biogenesis of caveolae and show its use for single-molecule interaction studies. Insertion of expressed caveolin-1 (CAV1) into Lt membranes was analogous to that of caveolin in native membranes. Electron tomography showed that caveolins generate domains of precise size and curvature. Cell-free caveolae were used in quantitative assays to test the interaction of membrane-inserted caveolin with signaling proteins and to determine the stoichiometry of interactions. Binding of membrane-inserted CAV1 to several proposed binding partners, including endothelial nitric-oxide synthase, was negligible, but a small number of proteins, including TRAF2, interacted with CAV1 in a phosphorylation-(CAV1Y14)–stimulated manner. In cells subjected to oxidative stress, phosphorylated CAV1 recruited TRAF2 to the early endosome forming a novel signaling platform. These findings lead to a novel model for cellular stress signaling by CAV1.
Collapse
Affiliation(s)
- WooRam Jung
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Emma Sierecki
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Michele Bastiani
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Ailis O'Carroll
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Kirill Alexandrov
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - James Rae
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Wayne Johnston
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Dominic J B Hunter
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Charles Ferguson
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Yann Gambin
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Nicholas Ariotti
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia
| | - Robert G Parton
- The University of Queensland, The Institute for Molecular Bioscience, Brisbane, Queensland, Australia .,The University of Queensland, The Centre for Microscopy and Microanalysis, Brisbane, Queensland, Australia
| |
Collapse
|
11
|
|
12
|
Mote RD, Mahajan G, Padmanabhan A, Ambati R, Subramanyam D. Dual repression of endocytic players by ESCC microRNAs and the Polycomb complex regulates mouse embryonic stem cell pluripotency. Sci Rep 2017; 7:17572. [PMID: 29242593 PMCID: PMC5730570 DOI: 10.1038/s41598-017-17828-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 11/30/2017] [Indexed: 01/08/2023] Open
Abstract
Cell fate determination in the early mammalian embryo is regulated by multiple mechanisms. Recently, genes involved in vesicular trafficking have been shown to play an important role in cell fate choice, although the regulation of their expression remains poorly understood. Here we demonstrate for the first time that multiple endocytosis associated genes (EAGs) are repressed through a novel, dual mechanism in mouse embryonic stem cells (mESCs). This involves the action of the Polycomb Repressive Complex, PRC2, as well as post-transcriptional regulation by the ESC-specific cell cycle-regulating (ESCC) family of microRNAs. This repression is relieved upon differentiation. Forced expression of EAGs in mESCs results in a decrease in pluripotency, highlighting the importance of dual repression in cell fate regulation. We propose that endocytosis is critical for cell fate choice, and dual repression may function to tightly regulate levels of endocytic genes.
Collapse
Affiliation(s)
- Ridim Dadasaheb Mote
- National Centre for Cell Science, SP Pune University, Ganeshkhind, Pune, 411007, India
| | - Gaurang Mahajan
- National Centre for Cell Science, SP Pune University, Ganeshkhind, Pune, 411007, India
| | - Anup Padmanabhan
- Mechanobiology Institute, National University of Singapore, 5A Engineering Drive 1, Singapore, 117411, Singapore
| | - Ramaraju Ambati
- National Centre for Cell Science, SP Pune University, Ganeshkhind, Pune, 411007, India
| | - Deepa Subramanyam
- National Centre for Cell Science, SP Pune University, Ganeshkhind, Pune, 411007, India.
| |
Collapse
|
13
|
Li X, Yao F, zhang W, Cheng C, Chu B, Liu Y, Mei Y, Wu Y, Zou X, Hou L. Identification, expression pattern, cellular location and potential role of the caveolin-1 gene from Artemia sinica. Gene 2014; 540:161-70. [DOI: 10.1016/j.gene.2014.02.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 02/17/2014] [Accepted: 02/25/2014] [Indexed: 01/15/2023]
|
14
|
Paik YK, Jeong SK, Lee EY, Jeong PY, Shim YH. C. elegans: an invaluable model organism for the proteomics studies of the cholesterol-mediated signaling pathway. Expert Rev Proteomics 2014; 3:439-53. [PMID: 16901202 DOI: 10.1586/14789450.3.4.439] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
With the availability of its complete genome sequence and unique biological features relevant to human disease, Caenorhabditis elegans has become an invaluable model organism for the studies of proteomics, leading to the elucidation of nematode gene function. A journey from the genome to proteome of C. elegans may begin with preparation of expressed proteins, which enables a large-scale analysis of all possible proteins expressed under specific physiological conditions. Although various techniques have been used for proteomic analysis of C. elegans, systematic high-throughput analysis is still to come in order to accommodate studies of post-translational modification and quantitative analysis. Given that no integrated C. elegans protein expression database is available, it is about time that a global C. elegans proteome project is launched through which datasets of transcriptomes, protein-protein interaction and functional annotation can be integrated. As an initial target of a pilot project of the C. elegans proteome project, the cholesterol-mediated signaling pathway will be an excellent example since, like in other organisms, it is one of the key controlling pathways in cell growth and development in C. elegans. As this field tends to broaden to functional proteomics, there is a high demand to develop the versatile proteome informatics tools that can mange many different data in an integrative manner.
Collapse
Affiliation(s)
- Young-Ki Paik
- Yonsei University, Department of Biochemistry, 134 Shinchon-dong, Sudamoon-Ku, Seoul, 120-749, Korea.
| | | | | | | | | |
Collapse
|
15
|
NSBP-1 mediates the effects of cholesterol on insulin/IGF-1 signaling in Caenorhabditis elegans. Cell Mol Life Sci 2012; 70:1623-36. [PMID: 23255046 DOI: 10.1007/s00018-012-1221-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2012] [Revised: 10/30/2012] [Accepted: 11/22/2012] [Indexed: 01/12/2023]
Abstract
Nematode sterol-binding protein 1 (NSBP-1) is a homolog of nucleosome assembly protein 1 in mammals that is expressed widely in Caenorhabditis elegans. NSBP-1 mutants are biologically lethal, demonstrating the significance of the gene in growth and development. We investigated how cholesterol influences the insulin signaling pathway through this novel sterol-binding protein in C. elegans. Here we report that NSBP-1 influences many biological processes mediated by insulin signaling, such as longevity, dauer formation, fat storage, and resistance to oxidative stress. We found that NSBP-1 is phosphorylated by AKT-1 downstream of insulin signaling. In the absence of insulin signaling, NSBP-1 is translocated to the nucleus and binds to DAF-16, a FOXO transcription factor, in a cholesterol-dependent manner. Moreover, NSBP-1 and DAF-16 regulate a common set of genes that can directly modulate fat storage, longevity, and resistance to stress. Together, our results present a new steroid-binding molecule that can connect sterol signaling to insulin signaling through direct interaction with FOXO.
Collapse
|
16
|
LUPIÁÑEZ DARÍOG, REAL FRANCISCAM, DADHICH RAJESHK, CARMONA FRANCISCOD, BURGOS MIGUEL, BARRIONUEVO FRANCISCOJ, JIMÉNEZ RAFAEL. Pattern and Density of Vascularization in Mammalian Testes, Ovaries, and Ovotestes. JOURNAL OF EXPERIMENTAL ZOOLOGY PART B-MOLECULAR AND DEVELOPMENTAL EVOLUTION 2012; 318:170-81. [DOI: 10.1002/jez.b.22000] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
17
|
Rauthan M, Pilon M. The mevalonate pathway in C. elegans. Lipids Health Dis 2011; 10:243. [PMID: 22204706 PMCID: PMC3274489 DOI: 10.1186/1476-511x-10-243] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Accepted: 12/28/2011] [Indexed: 11/10/2022] Open
Abstract
The mevalonate pathway in human is responsible for the synthesis of cholesterol and other important biomolecules such as coenzyme Q, dolichols and isoprenoids. These molecules are required in the cell for functions ranging from signaling to membrane integrity, protein prenylation and glycosylation, and energy homeostasis. The pathway consists of a main trunk followed by sub-branches that synthesize the different biomolecules. The majority of our knowledge about the mevalonate pathway is currently focused on the cholesterol synthesis branch, which is the target of the cholesterol-lowering statins; less is known about the function and regulation of the non-cholesterol-related branches. To study them, we need a biological system where it is possible to specifically modulate these metabolic branches individually or in groups. The nematode Caenorhabditis elegans (C. elegans) is a promising model to study these non-cholesterol branches since its mevalonate pathway seems very well conserved with that in human except that it has no cholesterol synthesis branch. The simple genetic makeup and tractability of C. elegans makes it relatively easy to identify and manipulate key genetic components of the mevalonate pathway, and to evaluate the consequences of tampering with their activity. This general experimental approach should lead to new insights into the physiological roles of the non-cholesterol part of the mevalonate pathway. This review will focus on the current knowledge related to the mevalonate pathway in C. elegans and its possible applications as a model organism to study the non-cholesterol functions of this pathway.
Collapse
Affiliation(s)
- Manish Rauthan
- Department of Cell and Molecular Biology, University of Gothenburg, S-405 30 Gothenburg, Sweden
| | | |
Collapse
|
18
|
Mo S, Yang S, Cui Z. New glimpses of caveolin-1 functions in embryonic development and human diseases. FRONTIERS IN BIOLOGY 2011; 6:367. [PMID: 32215005 PMCID: PMC7089126 DOI: 10.1007/s11515-011-1132-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/14/2010] [Accepted: 12/30/2010] [Indexed: 11/17/2022]
Abstract
Caveolin-1 (Cav-1) isoforms, including Cav-1α and Cav-1β, were identified as integral membrane proteins and the major components of caveolae. Cav-1 proteins are highly conserved during evolution from {itCaenorhabditis elegans} to human and are capable of interacting with many signaling molecules through their caveolin scaffolding domains to regulate the activities of multiple signaling pathways. Thus, Cav-1 plays crucial roles in the regulation of cellular proliferation, differentiation and apoptosis in a cell-specific and contextual manner. In addition, Cav-1 is essential for embryonic development of vertebrates owing to its regulation of BMP, Wnt, TGF-β and other key signaling molecules. Moreover, Cav-1 is mainly expressed in terminally differentiated cells and its abnormal expression is often associated with human diseases, such as tumor progression, cardiovascular diseases, fibrosis, lung regeneration, and diseases related to virus. In this review, we will further discuss the potential of Cav-1 as a target for disease therapy and multiple drug resistance.
Collapse
Affiliation(s)
- Saijun Mo
- Department of Basic Oncology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001 China
| | - Shengli Yang
- Department of Basic Oncology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, 450001 China
| | - Zongbin Cui
- Key Laboratory of Biodiversity and Conservation of Aquatic Organism, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072 China
| |
Collapse
|
19
|
Buschiazzo J, Alonso TS, Biscoglio M, Antollini SS, Bonini IC. Nongenomic steroid- and ceramide-induced maturation in amphibian oocytes involves functional caveolae-like microdomains associated with a cytoskeletal environment. Biol Reprod 2011; 85:808-22. [PMID: 21653896 DOI: 10.1095/biolreprod.110.090365] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Stimulation of full-grown amphibian oocytes with progesterone initiates a nontranscriptional signaling pathway that converges in the activation of Cdc2/cyclin B and reentry into meiosis. We observed that cholesterol depletion mediated by methyl-beta-cyclodextrin (MbetaCD) inhibited meiotic maturation, suggesting involvement of membrane rafts. In the present study, we further characterized caveolae-like membranes from Rhinella arenarum oocytes biochemically and functionally. The identification by mass spectrometry of a nonmuscle myosin heavy-chain associated with caveolar membranes showed evidence of direct involvement of the underlying cytoskeletal environment in the structure of oocyte rafts. Biophysical analysis using the fluorescent probe Laurdan revealed that MbetaCD-mediated cholesterol depletion affected membrane lipid order. In line with this finding, cholesterol removal also affected the localization of the raft marker lipid GM1. Results demonstrated that ceramide is an effective inducer of maturation that alters the distribution of the raft markers caveolin-1, SRC, and GM1, while progesterone seems not to affect membrane microdomain integrity. Cholesterol depletion had a greater effect on ceramide-induced maturation, thus suggesting that ceramide is an inducer more vulnerable to changes in the plasma membrane. MbetaCD treatment delayed tyrosine phosphorylation and MAPK activation in progesterone-induced maturation. Functional studies regarding tyrosine phosphorylation raise the possibility that the hormone receptor is located in the nonraft membrane in the absence of ligand and that it translocates to the caveola when it binds to progesterone. The presence of raft markers and the finding of signaling molecules from MAPK cascade functionally associated to oocyte light membranes suggest that this caveolae-rich fraction efficiently recreates, in part, maturation signaling.
Collapse
Affiliation(s)
- Jorgelina Buschiazzo
- Instituto de Investigaciones Bioquímicas de Bahía Blanca, Universidad Nacional del Sur-Consejo Nacional de Investigaciones Científicas y Técnicas, Bahía Blanca, Argentina
| | | | | | | | | |
Collapse
|
20
|
Branicky R, Desjardins D, Liu JL, Hekimi S. Lipid transport and signaling in Caenorhabditis elegans. Dev Dyn 2010; 239:1365-77. [PMID: 20151418 DOI: 10.1002/dvdy.22234] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The strengths of the Caenorhabditis elegans model have been recently applied to the study of the pathways of lipid storage, transport, and signaling. As the lipid storage field has recently been reviewed, in this minireview we (1) discuss some recent studies revealing important physiological roles for lipases in mobilizing lipid reserves, (2) describe various pathways of lipid transport, with a particular focus on the roles of lipoproteins, (3) debate the utility of using C. elegans as a model for human dyslipidemias that impinge on atherosclerosis, and (4) describe several systems where lipids affect signaling, highlighting the particular properties of lipids as information-carrying molecules. We conclude that the study of lipid biology in C. elegans exemplifies the advantages afforded by a whole-animal model system where interactions between tissues and organs, and functions such as nutrient absorption, distribution, and storage, as well as reproduction can all be studied simultaneously.
Collapse
Affiliation(s)
- Robyn Branicky
- Department of Biology, McGill University, Montreal, Quebec, Canada
| | | | | | | |
Collapse
|
21
|
Parker S, Baylis HA. Overexpression of caveolins in Caenorhabditis elegans induces changes in egg-laying and fecundity. Commun Integr Biol 2010; 2:382-4. [PMID: 19907693 DOI: 10.4161/cib.2.5.8715] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2009] [Accepted: 04/07/2009] [Indexed: 02/06/2023] Open
Abstract
Caveolae are small plasma membrane-associated invaginations that are enriched in proteins of the caveolin family in addition to, sphingolipids, glycosphingolipids and cholesterol. Caveolae have been implicated in several endocytic and trafficking mechanisms. Mutations in caveolins have been shown to cause disease and caveolae offer one site for pathogen entry. The Caenorhabditis elegans genome encodes two caveolins (cav-1 and cav-2); we have shown that these two proteins have distinct expression patterns. CAV-1 is found in the majority of cells in embryos and in the body-wall muscles, neurons and germ line of adult worms. CAV-2 is expressed in the intestine and is required for apical lipid trafficking. In the course of our studies, we generated several constructs to overexpress caveolins in C. elegans. Here we show that overexpression of cav-1 protects against the decrease in brood size associated with the effects of heat shock and the presence of extrachromosomal arrays in heat-shocked animals. Furthermore, we show that overexpression of cav-2 in the nervous system increases the rate of egg-laying and total number of eggs laid.
Collapse
Affiliation(s)
- Scott Parker
- Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK.
| | | |
Collapse
|
22
|
Hadwiger G, Dour S, Arur S, Fox P, Nonet ML. A monoclonal antibody toolkit for C. elegans. PLoS One 2010; 5:e10161. [PMID: 20405020 PMCID: PMC2854156 DOI: 10.1371/journal.pone.0010161] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2010] [Accepted: 03/22/2010] [Indexed: 01/12/2023] Open
Abstract
Background Antibodies are critical tools in many avenues of biological research. Though antibodies can be produced in the research laboratory setting, most research labs working with vertebrates avail themselves of the wide array of commercially available reagents. By contrast, few such reagents are available for work with model organisms. Methodology/Principal Findings We report the production of monoclonal antibodies directed against a wide range of proteins that label specific subcellular and cellular components, and macromolecular complexes. Antibodies were made to synaptobrevin (SNB-1), a component of synaptic vesicles; to Rim (UNC-10), a protein localized to synaptic active zones; to transforming acidic coiled-coil protein (TAC-1), a component of centrosomes; to CENP-C (HCP-4), which in worms labels the entire length of their holocentric chromosomes; to ORC2 (ORC-2), a subunit of the DNA origin replication complex; to the nucleolar phosphoprotein NOPP140 (DAO-5); to the nuclear envelope protein lamin (LMN-1); to EHD1 (RME-1) a marker for recycling endosomes; to caveolin (CAV-1), a marker for caveolae; to the cytochrome P450 (CYP-33E1), a resident of the endoplasmic reticulum; to β-1,3-glucuronyltransferase (SQV-8) that labels the Golgi; to a chaperonin (HSP-60) targeted to mitochondria; to LAMP (LMP-1), a resident protein of lysosomes; to the alpha subunit of the 20S subcomplex (PAS-7) of the 26S proteasome; to dynamin (DYN-1) and to the α-subunit of the adaptor complex 2 (APA-2) as markers for sites of clathrin-mediated endocytosis; to the MAGUK, protein disks large (DLG-1) and cadherin (HMR-1), both of which label adherens junctions; to a cytoskeletal linker of the ezrin-radixin-moesin family (ERM-1), which localized to apical membranes; to an ERBIN family protein (LET-413) which localizes to the basolateral membrane of epithelial cells and to an adhesion molecule (SAX-7) which localizes to the plasma membrane at cell-cell contacts. In addition to working in whole mount immunocytochemistry, most of these antibodies work on western blots and thus should be of use for biochemical fractionation studies. Conclusions/Significance We have produced a set of monoclonal antibodies to subcellular components of the nematode C. elegans for the research community. These reagents are being made available through the Developmental Studies Hybridoma Bank (DSHB).
Collapse
Affiliation(s)
- Gayla Hadwiger
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Scott Dour
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Swathi Arur
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Paul Fox
- Department of Genetics, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Michael L. Nonet
- Department of Anatomy and Neurobiology, Washington University School of Medicine, St. Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
23
|
Hansen CG, Nichols BJ. Exploring the caves: cavins, caveolins and caveolae. Trends Cell Biol 2010; 20:177-86. [PMID: 20153650 DOI: 10.1016/j.tcb.2010.01.005] [Citation(s) in RCA: 222] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2009] [Revised: 01/07/2010] [Accepted: 01/11/2010] [Indexed: 01/29/2023]
Abstract
Caveolae are ampullate (flask-shaped) invaginations that are abundant in the plasma membrane of many mammalian cell types. Although caveolae are implicated in a wide range of processes including endothelial transcytosis, lipid homeostasis and cellular signalling, a detailed molecular picture of many aspects of their function has been elusive. Until recently, the only extensively characterised protein components of caveolae were the caveolins. Recently, data from several laboratories have demonstrated that a family of four related proteins, termed cavins 1-4, plays key roles in caveolar biogenesis and function. Salient properties of the cavin family include their propensity to form complexes with each other and their different but overlapping tissue distribution. This review summarises recent data on the cavins, and sets them in the context of open questions on the construction and function of caveolae. The discovery of cavins implies that caveolae might have unexpectedly diverse structural properties, in accord with the wide range of functions attributed to these 'little caves'.
Collapse
|
24
|
The absence of caveolin-1 increases proliferation and anchorage- independent growth by a Rac-dependent, Erk-independent mechanism. Mol Cell Biol 2009; 29:5046-59. [PMID: 19620284 DOI: 10.1128/mcb.00315-09] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Anchorage-independent growth (AIG) of cancer cells requires escape from integrin-mediated signals. A protein frequently downregulated in cancer, caveolin-1 (Cav1), mediates integrin control of several growth-regulatory pathways. We report that loss of Cav1 results in faster exit from quiescence and progress through the cell cycle, proliferation without anchorage to substrate, and absence of cyclin D1 downregulation upon serum deprivation or detachment. Surprisingly, this proliferative advantage is independent of Erk-mitogen-activated protein kinase signaling; instead, cyclin expression and cell cycle progression in the absence of Cav1 are driven by increased membrane order and Rac targeting. AIG was induced in Cav1-expressing cells by forced membrane targeting of Rac1 or by inhibiting Cav1-mediated internalization of plasma membrane ordered domains at which Rac1 accumulates. Restoring Rho activity, which is downregulated after loss of Cav1, antagonizes Rac1 and prevents cyclin D1 accumulation after serum starvation or loss of adhesion. Anchorage independence and increased proliferation in Cav1-deficient tumoral and null cells are thus due to an increased fraction of active Rac1 at membrane ordered domains. These results provide insight into the mechanisms regulating growth of cancer cells, which frequently lose Cav1 function.
Collapse
|
25
|
Rodríguez H, Silva I, Jiménez L, Sánchez C, Espinoza-Navarro O, Boarelli P, Fornés M. Presencia cualitativa y distribución de caveolina 1 (cav-1) en la celularidad y estadios del ciclo de la espermatogénesis. Rev Int Androl 2009. [DOI: 10.1016/s1698-031x(09)71613-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
26
|
Mao H, Diehl AM, Li YX. Sonic hedgehog ligand partners with caveolin-1 for intracellular transport. J Transl Med 2009; 89:290-300. [PMID: 19139721 PMCID: PMC2647995 DOI: 10.1038/labinvest.2008.163] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Prenatal alcohol exposure is the most common environmental factor leading to congenital birth defects in the United States. Although significant progress has been made in this field, the detailed molecular pathology of fetal alcohol syndrome (FAS) remains to be determined. Previously, we have shown that alcohol exposure perturbs hedgehog signal transduction in zebrafish embryos by inhibiting the post-translational cholesterol modification of Sonic hedgehog (Shh), leading to decreased levels of mature Shh ligand that is associated with the plasma membrane, and causing transient loss of Hh signaling, resulting in permanent FAS-related morphological abnormalities. In the present study, we further elucidate the mechanisms that regulate the intracellular transportation and secretion of Shh using the hepatic stellate cell line HSC8B. We have found that Shh is associated with caveolin-1 in the Golgi apparatus to form protein complexes and that these complexes are packaged as large punctuate structures (transport vesicles) that are transported to the plasma membrane in lipid raft microdomains. Alcohol exposure does not significantly interrupt translation of shh mRNA in endoplasmic reticulum (ER) or the trafficking of Shh from the ER to the Golgi apparatus. However, alcohol does prevent the entry of Shh into transport vesicles from the Golgi to the plasma membrane and specifically decreases the amount of caveolin-1/Shh complex found in lipid rafts, causing cytoplasmic accumulation of Shh and leading to a deficiency of Shh ligand secretion into the extracellular matrix.
Collapse
Affiliation(s)
- Hua Mao
- Division of Gastroenterology and Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Anna Mae Diehl
- Division of Gastroenterology and Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA
| | - Yin-Xiong Li
- Division of Gastroenterology and Department of Medicine, Duke University Medical Center, Durham, North Carolina 27710, USA.,Department of Pediatrics, Duke University Medical Center, Durham, North Carolina 27710, USA
| |
Collapse
|
27
|
Parker S, Walker DS, Ly S, Baylis HA. Caveolin-2 is required for apical lipid trafficking and suppresses basolateral recycling defects in the intestine of Caenorhabditis elegans. Mol Biol Cell 2009; 20:1763-71. [PMID: 19158391 PMCID: PMC2655242 DOI: 10.1091/mbc.e08-08-0837] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 12/08/2008] [Accepted: 01/13/2009] [Indexed: 12/30/2022] Open
Abstract
Caveolins are plasma membrane-associated proteins that colocalize with, and stabilize caveolae. Their functions remain unclear although they are known to be involved in specific events in cell signaling and endocytosis. Caenorhabditis elegans encodes two caveolin genes, cav-1 and cav-2. We show that cav-2 is expressed in the intestine where it is localized to the apical membrane and in intracellular bodies. Using the styryl dye FM4-64 and BODIPY-labeled lactosylceramide, we show that the intestinal cells of cav-2 animals are defective in the apical uptake of lipid markers. These results suggest parallels with the function of caveolins in lipid homeostasis in mammals. We also show that CAV-2 depletion suppresses the abnormal accumulation of vacuoles that result from defective basolateral recycling in rme-1 and rab-10 mutants. Analysis of fluorescent markers of basolateral endocytosis and recycling suggest that endocytosis is normal in cav-2 mutants and thus, that the suppression of basolateral recycling defects in cav-2 mutants is due to changes in intracellular trafficking pathways. Finally, cav-2 mutants also have abnormal trafficking of yolk proteins. Taken together, these data indicate that caveolin-2 is an integral component of the trafficking network in the intestinal cells of C. elegans.
Collapse
Affiliation(s)
- Scott Parker
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, United Kingdom
| | - Denise S. Walker
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, United Kingdom
| | - Sung Ly
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, United Kingdom
| | - Howard A. Baylis
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, United Kingdom
| |
Collapse
|
28
|
Sato M, Grant BD, Harada A, Sato K. Rab11 is required for synchronous secretion of chondroitin proteoglycans after fertilization in Caenorhabditis elegans. J Cell Sci 2008; 121:3177-86. [PMID: 18765566 DOI: 10.1242/jcs.034678] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
We previously identified a novel type of caveolin-enriched secretory vesicle in Caenorhabditis elegans oocytes. These vesicles undergo synchronous fusion with the plasma membrane immediately after fertilization, suggesting that they could be cortical granules that have been described in diverse animal species. Here, we report that these vesicles are indeed cortical granules, delivering essential chondroitin proteoglycans and mucin-like glycoproteins to the early embryonic extracellular matrices (ECMs). Furthermore, we have found that the small GTPase RAB-11 and the target-SNARE SYN-4 are required for cortical granule excoytosis after fertilization. In oocytes, SYN-4 localizes mainly to the plasma membrane, whereas GFP::RAB-11 accumulates transiently on the cortical granules during ovulation, immediately prior to fertilization. Importantly, cytokinesis defects in early embryos are commonly observed after depletion of either rab-11 or syn-4, producing a phenotype very similar to that observed after blockade of chondroitin synthesis. Taken together, our results indicate that at least part of the essential role for RAB-11 and SYN-4 in early embryogenesis is in the targeting of cortical granules to the plasma membrane during the precisely regulated secretion of ECM components.
Collapse
Affiliation(s)
- Miyuki Sato
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, Japan
| | | | | | | |
Collapse
|
29
|
Abstract
N-Glycan branching in the medial-Golgi generates ligands for lattice-forming lectins (e.g., galectins) that regulate surface levels of glycoproteins including epidermal growth factor (EGF) and transforming growth factor-beta (TGF-beta) receptors. Moreover, functional classes of glycoproteins differ in N-glycan multiplicities (number of N-glycans/peptide), a genetically encoded feature of glycoproteins that interacts with metabolic flux (UDP-GlcNAc) and N-glycan branching to differentially regulate surface levels. Oncogenesis increases beta1,6-N-acetylglucosaminyltransferase V (encoded by Mgat5) expression, and its high-affinity galectin ligands promote surface retention of growth receptors with a reduced dependence on UDP-GlcNAc. Mgat5(-/-) tumor cells are less metastatic in vivo and less responsive to cytokines in vitro, but undergo secondary changes that support tumor cell proliferation. These include loss of Caveolin-1, a negative regulator of EGF signaling, and increased reactive oxygen species, an inhibitor of phosphotyrosine phosphatases. These studies suggest a systems approach to cancer treatment where the surface distribution of receptors is targeted through metabolism and N-glycan branching to induce growth arrest.
Collapse
Affiliation(s)
- Ken S Lau
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | | |
Collapse
|
30
|
Vinci G, Xia X, Veitia RA. Preservation of genes involved in sterol metabolism in cholesterol auxotrophs: facts and hypotheses. PLoS One 2008; 3:e2883. [PMID: 18682733 PMCID: PMC2478713 DOI: 10.1371/journal.pone.0002883] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2008] [Accepted: 07/11/2008] [Indexed: 12/02/2022] Open
Abstract
Background It is known that primary sequences of enzymes involved in sterol biosynthesis are well conserved in organisms that produce sterols de novo. However, we provide evidence for a preservation of the corresponding genes in two animals unable to synthesize cholesterol (auxotrophs): Drosophila melanogaster and Caenorhabditis elegans. Principal Findings We have been able to detect bona fide orthologs of several ERG genes in both organisms using a series of complementary approaches. We have detected strong sequence divergence between the orthologs of the nematode and of the fruitfly; they are also very divergent with respect to the orthologs in organisms able to synthesize sterols de novo (prototrophs). Interestingly, the orthologs in both the nematode and the fruitfly are still under selective pressure. It is possible that these genes, which are not involved in cholesterol synthesis anymore, have been recruited to perform different new functions. We propose a more parsimonious way to explain their accelerated evolution and subsequent stabilization. The products of ERG genes in prototrophs might be involved in several biological roles, in addition to sterol synthesis. In the case of the nematode and the fruitfly, the relevant genes would have lost their ancestral function in cholesterogenesis but would have retained the other function(s), which keep them under pressure. Conclusions By exploiting microarray data we have noticed a strong expressional correlation between the orthologs of ERG24 and ERG25 in D. melanogaster and genes encoding factors involved in intracellular protein trafficking and folding and with Start1 involved in ecdysteroid synthesis. These potential functional connections are worth being explored not only in Drosophila, but also in Caenorhabditis as well as in sterol prototrophs.
Collapse
Affiliation(s)
- Giovanna Vinci
- Institut Cochin, Département de Génétique et Développement. Inserm, U567, CNRS, UMR 8104, Université Paris 5, Faculté de Médecine Paris Descartes, UM 3, Paris, France
| | - Xuhua Xia
- CAREG and Biology Department, University of Ottawa, Ottawa, Ontario, Canada
| | - Reiner A. Veitia
- Institut Cochin, Département de Génétique et Développement. Inserm, U567, CNRS, UMR 8104, Université Paris 5, Faculté de Médecine Paris Descartes, UM 3, Paris, France
- Université Denis Diderot/Paris VII, Paris, France
- * E-mail:
| |
Collapse
|
31
|
|
32
|
Goetz JG, Lajoie P, Wiseman SM, Nabi IR. Caveolin-1 in tumor progression: the good, the bad and the ugly. Cancer Metastasis Rev 2008; 27:715-35. [DOI: 10.1007/s10555-008-9160-9] [Citation(s) in RCA: 229] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
33
|
Kirkham M, Nixon SJ, Howes MT, Abi-Rached L, Wakeham DE, Hanzal-Bayer M, Ferguson C, Hill MM, Fernandez-Rojo M, Brown DA, Hancock JF, Brodsky FM, Parton RG. Evolutionary analysis and molecular dissection of caveola biogenesis. J Cell Sci 2008; 121:2075-86. [PMID: 18505796 DOI: 10.1242/jcs.024588] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Caveolae are an abundant feature of mammalian cells. Integral membrane proteins called caveolins drive the formation of caveolae but the precise mechanisms underlying caveola formation, and the origin of caveolae and caveolins during evolution, are unknown. Systematic evolutionary analysis shows conservation of genes encoding caveolins in metazoans. We provide evidence for extensive and ancient, local and genomic gene duplication, and classify distinct caveolin gene families. Vertebrate caveolin-1 and caveolin-3 isoforms, as well as an invertebrate (Apis mellifera, honeybee) caveolin, all form morphologically identical caveolae in caveolin-1-null mouse cells, demonstrating that caveola formation is a conserved feature of evolutionarily distant caveolins. However, coexpression of flotillin-1 and flotillin-2 did not cause caveola biogenesis in this system. In contrast to the other tested caveolins, C. elegans caveolin is efficiently transported to the plasma membrane but does not generate caveolae, providing evidence of diversity of function in the caveolin gene family. Using C. elegans caveolin as a template to generate hybrid caveolin constructs we now define domains of caveolin required for caveolae biogenesis. These studies lead to a model for caveola formation and novel insights into the evolution of caveolin function.
Collapse
Affiliation(s)
- Matthew Kirkham
- Institute for Molecular Bioscience, University of Queensland, Queensland, Brisbane, Australia
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Parker S, Peterkin HS, Baylis HA. Muscular dystrophy associated mutations in caveolin-1 induce neurotransmission and locomotion defects in Caenorhabditis elegans. INVERTEBRATE NEUROSCIENCE 2007; 7:157-64. [PMID: 17629760 DOI: 10.1007/s10158-007-0051-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2007] [Accepted: 06/11/2007] [Indexed: 12/13/2022]
Abstract
Mutations in human caveolin-3 are known to underlie a range of myopathies. The cav-1 gene of Caenorhabditis elegans is a homologue of human caveolin-3 and is expressed in both neurons and body wall muscles. Within the body wall muscle CAV-1 localises adjacent to neurons, most likely at the neuromuscular junction (NMJ). Using fluorescently tagged CAV-1 and pre- and post-synaptic markers we demonstrate that CAV-1 co-localises with UNC-63, a post-synaptic marker, but not with several pre-synaptic markers. To establish a model for human muscular dystrophies caused by dominant-negative mutations in caveolin-3 we created transgenic animals carrying versions of cav-1 with homologous mutations. These animals had increased sensitivity to levamisole, suggesting a role for cav-1 at the NMJ. Animals carrying a deletion in cav-1 show a similar sensitivity. Sensitivity to levamisole and locomotion were also perturbed in animals carrying a dominant-negative cav-1 and a mutation in dynamin, which is a protein known to interact with caveolins. Thus, indicating an interaction between CAV-1 and dynamin at the NMJ and/or in neurons.
Collapse
Affiliation(s)
- Scott Parker
- Department of Zoology, University of Cambridge, Downing Street, Cambridge, CB2 3EJ, UK
| | | | | |
Collapse
|
35
|
Nixon SJ, Carter A, Wegner J, Ferguson C, Floetenmeyer M, Riches J, Key B, Westerfield M, Parton RG. Caveolin-1 is required for lateral line neuromast and notochord development. J Cell Sci 2007; 120:2151-61. [PMID: 17550965 DOI: 10.1242/jcs.003830] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Caveolae have been linked to diverse cellular functions and to many disease states. In this study we have used zebrafish to examine the role of caveolin-1 and caveolae during early embryonic development. During development, expression is apparent in a number of tissues including Kupffer's vesicle, tailbud, intersomite boundaries, heart, branchial arches, pronephric ducts and periderm. Particularly strong expression is observed in the sensory organs of the lateral line, the neuromasts and in the notochord where it overlaps with expression of caveolin-3. Morpholino-mediated downregulation of Cav1alpha caused a dramatic inhibition of neuromast formation. Detailed ultrastructural analysis, including electron tomography of the notochord, revealed that the central regions of the notochord has the highest density of caveolae of any embryonic tissue comparable to the highest density observed in any vertebrate tissue. In addition, Cav1alpha downregulation caused disruption of the notochord, an effect that was enhanced further by Cav3 knockdown. These results indicate an essential role for caveolin and caveolae in this vital structural and signalling component of the embryo.
Collapse
Affiliation(s)
- Susan J Nixon
- Institute for Molecular Bioscience, University of Queensland, Brisbane 4072, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Sato K, Sato M, Audhya A, Oegema K, Schweinsberg P, Grant BD. Dynamic regulation of caveolin-1 trafficking in the germ line and embryo of Caenorhabditis elegans. Mol Biol Cell 2006; 17:3085-94. [PMID: 16672374 PMCID: PMC1483042 DOI: 10.1091/mbc.e06-03-0211] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
Caveolin is the major protein component required for the formation of caveolae on the plasma membrane. Here we show that trafficking of Caenorhabditis elegans caveolin-1 (CAV-1) is dynamically regulated during development of the germ line and embryo. In oocytes a CAV-1-green fluorescent protein (GFP) fusion protein is found on the plasma membrane and in large vesicles (CAV-1 bodies). After ovulation and fertilization the CAV-1 bodies fuse with the plasma membrane in a manner reminiscent of cortical granule exocytosis as described in other species. Fusion of CAV-1 bodies with the plasma membrane appears to be regulated by the advancing cell cycle, and not fertilization per se, because fusion can proceed in spe-9 fertilization mutants but is blocked by RNA interference-mediated knockdown of an anaphase-promoting complex component (EMB-27). After exocytosis, most CAV-1-GFP is rapidly endocytosed and degraded within one cell cycle. CAV-1 bodies in oocytes appear to be produced by the Golgi apparatus in an ARF-1-dependent, clathrin-independent, mechanism. Conversely endocytosis and degradation of CAV-1-GFP in embryos requires clathrin, dynamin, and RAB-5. Our results demonstrate that the distribution of CAV-1 is highly dynamic during development and provides new insights into the sorting mechanisms that regulate CAV-1 localization.
Collapse
Affiliation(s)
- Ken Sato
- *Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma 371-8512, Japan; and
| | - Miyuki Sato
- *Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma 371-8512, Japan; and
| | - Anjon Audhya
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Karen Oegema
- Ludwig Institute for Cancer Research, Department of Cellular and Molecular Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Peter Schweinsberg
- *Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| | - Barth D. Grant
- *Department of Molecular Biology and Biochemistry, Rutgers University, Piscataway, NJ 08854
| |
Collapse
|
37
|
Matallanas D, Sanz-Moreno V, Arozarena I, Calvo F, Agudo-Ibáñez L, Santos E, Berciano MT, Crespo P. Distinct utilization of effectors and biological outcomes resulting from site-specific Ras activation: Ras functions in lipid rafts and Golgi complex are dispensable for proliferation and transformation. Mol Cell Biol 2006; 26:100-16. [PMID: 16354683 PMCID: PMC1317613 DOI: 10.1128/mcb.26.1.100-116.2006] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Ras proteins are distributed in different types of plasma membrane microdomains and endomembranes. However, how microlocalization affects the signals generated by Ras and its subsequent biological outputs is largely unknown. We have approached this question by selectively targeting RasV12 to different cellular sublocalizations. We show here that compartmentalization dictates Ras utilization of effectors and the intensity of its signals. Activated Ras can evoke enhanced proliferation and transformation from most of its platforms, with the exception of the Golgi complex. Furthermore, signals that promote survival emanate primarily from the endoplasmic reticulum pool. In addition, we have investigated the need for the different pools of endogenous Ras in the conveyance of upstream mitogenic and transforming signals. Using targeted RasN17 inhibitory mutants and in physiological contexts such as H-Ras/N-Ras double knockout fibroblasts, we demonstrate that Ras functions at lipid rafts and at the Golgi complex are fully dispensable for proliferation and transformation.
Collapse
Affiliation(s)
- David Matallanas
- Instituto de Investigaciones Biomédicas, Consejo Superior de Investigaciones Científicas (CSIC), Departamento de Biología Molecular, Spain
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Sotgia F, Williams TM, Schubert W, Medina F, Minetti C, Pestell RG, Lisanti MP. Caveolin-1 deficiency (-/-) conveys premalignant alterations in mammary epithelia, with abnormal lumen formation, growth factor independence, and cell invasiveness. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:292-309. [PMID: 16400031 PMCID: PMC1592656 DOI: 10.2353/ajpath.2006.050429] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
During breast cancer development, the luminal space of the mammary acinar unit fills with proliferating epithelial cells that exhibit growth factor-independence, cell attachment defects, and a more invasive fibroblastic phenotype. Here, we used primary cultures of mammary epithelial cells derived from genetically engineered mice to identify caveolin-1 (Cav-1) as a critical factor for maintaining the normal architecture of the mammary acinar unit. Isolated cultures of normal mammary epithelial cells retained the capacity to generate mammary acini within extracellular matrix. However, those from Cav-1 (-/-) mice exhibited defects in three-dimensional acinar architecture, including disrupted lumen formation and epidermal growth factor-independent growth due to hyperactivation of the p42/44 mitogen-activated protein kinase cascade. In addition, Cav-1-null mammary epithelial cells deprived of exogenous extracellular matrix underwent a spontaneous epithelial-mesenchymal transition, with reorganization of the actin cytoskeleton, and E-cadherin redistribution. Mechanistically, these phenotypic changes appear to be caused by increases in matrix metalloproteinase-2/9 secretion and transforming growth factor-beta/Smad-2 hyperactivation. Finally, loss of Cav-1 potentiated the ability of growth factors (hepatocyte growth factor and basic fibroblast growth factor) to induce mammary acini branching, indicative of a more invasive fibroblastic phenotype. Thus, a Cav-1 deficiency profoundly affects mammary epithelia by modulating the activation state of important signaling cascades. Primary cultures of Cav-1-deficient mammary epithelia will provide a valuable new model to study the spatial/temporal progression of mammary cell transformation.
Collapse
Affiliation(s)
- Federica Sotgia
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Ave., Bronx, NY 10461, USA
| | | | | | | | | | | | | |
Collapse
|
39
|
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
|
40
|
Le Lay S, Kurzchalia TV. Getting rid of caveolins: phenotypes of caveolin-deficient animals. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1746:322-33. [PMID: 16019085 DOI: 10.1016/j.bbamcr.2005.06.001] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2005] [Revised: 06/03/2005] [Accepted: 06/06/2005] [Indexed: 10/25/2022]
Abstract
The elucidation of the role of caveolae has been the topic of many investigations which were greatly enhanced after the discovery of caveolin, the protein marker of these flask-shaped plasma membrane invaginations. The generation of mice deficient in the various caveolin genes (cav-1, cav-2 and cav-3) has provided physiological models to unravel the role of caveolins or caveolae at the whole organism level. Remarkably, despite the essential role of caveolins in caveolae biogenesis, all knockout mice are viable and fertile. However, lack of caveolae or caveolins leads to a wide range of phenotypes including muscle, pulmonary or lipid disorders, suggesting their implication in many cellular processes. The aim of this review is to give a broad overview of the phenotypes described for the caveolin-deficient mice and to link them to the numerous functions so far assigned to caveolins/caveolae.
Collapse
Affiliation(s)
- Soazig Le Lay
- MPI of Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | | |
Collapse
|
41
|
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
|
42
|
Li J, Brown G, Ailion M, Lee S, Thomas JH. NCR-1 and NCR-2, theC. eleganshomologs of the human Niemann-Pick type C1 disease protein, function upstream of DAF-9 in the dauer formation pathways. Development 2004; 131:5741-52. [PMID: 15509773 DOI: 10.1242/dev.01408] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Mutations in the human NPC1 gene cause most cases of Niemann-Pick type C(NP-C) disease, a fatal autosomal recessive neurodegenerative disorder. NPC1 is implicated in intracellular trafficking of cholesterol and glycolipids, but its exact function remains unclear. The C. elegans genome contains two homologs of NPC1, ncr-1 and ncr-2, and an ncr-2;ncr-1 double deletion mutant forms dauer larvae constitutively (Daf-c). We have analyzed the phenotypes of ncr single and double mutants in detail, and determined the ncr gene expression patterns. We find that the ncr genes function in a hormonal branch of the dauer formation pathway upstream of daf-9 and daf-12, which encode a cytochrome P450 enzyme and a nuclear hormone receptor, respectively. ncr-1 is expressed broadly in tissues with high levels of cholesterol, whereas expression of ncr-2 is restricted to a few cells. Both Ncr genes are expressed in the XXX cells, which are implicated in regulating dauer formation via the daf-9 pathway. Only the ncr-1 mutant is hypersensitive to cholesterol deprivation and to progesterone, an inhibitor of intracellular cholesterol trafficking. Our results support the hypothesis that ncr-1 and ncr-2 are involved in intracellular cholesterol processing in C. elegans, and that a sterol-signaling defect is responsible for the Daf-c phenotype of the ncr-2; ncr-1 mutant.
Collapse
Affiliation(s)
- Jie Li
- Department of Genome Sciences, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | |
Collapse
|
43
|
Miller MA, Cutter AD, Yamamoto I, Ward S, Greenstein D. Clustered organization of reproductive genes in the C. elegans genome. Curr Biol 2004; 14:1284-90. [PMID: 15268860 DOI: 10.1016/j.cub.2004.07.025] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2004] [Revised: 05/30/2004] [Accepted: 06/02/2004] [Indexed: 10/26/2022]
Abstract
Defining the forces that sculpt genome organization is fundamental for understanding the origin, persistence, and diversification of species. The genomic sequences of the nematodes Caenorhabditis elegans and Caenorhabditis briggsae provide an excellent opportunity to explore the dynamics of chromosome evolution. Extensive chromosomal rearrangement has accompanied divergence from their common ancestor, an event occurring roughly 100 million years ago (Mya); yet, morphologically, these species are nearly indistinguishable and both reproduce primarily by self-fertilization. Here, we show that genes expressed during spermatogenesis (sperm genes) are nonrandomly distributed across the C. elegans genome into three large clusters located on two autosomes. In addition to sperm genes, these chromosomal regions are enriched for genes involved in the hermaphrodite sperm/oocyte switch and in the reception of sperm signals that control fertilization. Most loci are present in single copy, suggesting that cluster formation is largely due to gene aggregation and not to tandem duplication. Comparative mapping indicates that the C. briggsae genome differs dramatically from the C. elegans genome in clustering. Because clustered genes have a direct role in reproduction and thus fitness, their aggregated pattern might have been shaped by natural selection, perhaps as hermaphroditism evolved.
Collapse
Affiliation(s)
- Michael A Miller
- Department of Cell Biology, University of Alabama at Birmingham, 35294, USA.
| | | | | | | | | |
Collapse
|
44
|
Gonzalez E, Nagiel A, Lin AJ, Golan DE, Michel T. Small Interfering RNA-mediated Down-regulation of Caveolin-1 Differentially Modulates Signaling Pathways in Endothelial Cells. J Biol Chem 2004; 279:40659-69. [PMID: 15292187 DOI: 10.1074/jbc.m407051200] [Citation(s) in RCA: 117] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Caveolin-1 is a scaffolding/regulatory protein that interacts with diverse signaling molecules in endothelial cells. To explore the role of this protein in receptor-modulated signaling pathways, we transfected bovine aortic endothelial cells (BAEC) with small interfering RNA (siRNA) duplexes to down-regulate caveolin-1 expression. Transfection of BAEC with duplex siRNA targeted against caveolin-1 mRNA selectively "knocked-down" the expression of caveolin-1 by approximately 90%, as demonstrated by immunoblot analyses of BAEC lysates. We used discontinuous sucrose gradients to purify caveolin-containing lipid rafts from siRNA-treated endothelial cells. Despite the near-total down-regulation of caveolin-1 expression, the lipid raft targeting of diverse signaling proteins (including the endothelial isoform of nitric-oxide synthase, Src-family tyrosine kinases, Galphaq and the insulin receptor) was unchanged. We explored the consequences of caveolin-1 knockdown on kinase pathways modulated by the agonists sphingosine-1 phosphate (S1P) and vascular endothelial growth factor (VEGF). siRNA-mediated caveolin-1 knockdown enhanced basal as well as S1P- and VEGF-induced phosphorylation of the protein kinase Akt and did not modify the basal or agonist-induced phosphorylation of extracellular signal-regulated kinases 1/2. Caveolin-1 knock-down also significantly enhanced the basal and agonist-induced activity of the small GTPase Rac. We used siRNA to down-regulate Rac expression in BAEC, and we observed that Rac knockdown significantly reduced basal, S1P-, and VEGF-induced Akt phosphorylation, suggesting a role for Rac activation in the caveolin siRNA-mediated increase in Akt phosphorylation. By using siRNA to knockdown caveolin-1 and Rac expression in cultured endothelial cells, we have found that caveolin-1 does not seem to be required for the targeting of signaling molecules to caveolae/lipid rafts and that caveolin-1 differentially modulates specific kinase pathways in endothelial cells.
Collapse
Affiliation(s)
- Eva Gonzalez
- Cardiovascular, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | | | | | |
Collapse
|
45
|
Lucius H, Friedrichson T, Kurzchalia TV, Lewin GR. Identification of caveolae-like structures on the surface of intact cells using scanning force microscopy. J Membr Biol 2004; 194:97-108. [PMID: 14502434 DOI: 10.1007/s00232-003-2029-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2002] [Indexed: 10/27/2022]
Abstract
Caveolae are small, functionally important membrane invaginations found on the surface of many different cell types. Using electron microscopy, caveolae can be unequivocally identified in cell membranes by virtue of their size and the presence of caveolin/VIP22 proteins in the caveolar coat. In this study we have applied for the first time scanning force microscopy (SFM), to visualize caveolae on the surface of living and fixed cells. By scanning the membranes of Chinese hamster ovary cells (CHO), using the tapping mode of the SFM in fluid, we could visualize small membrane pits on the cell membranes of living and fixed cells. Two populations of pits with mean diameters of around 100 nm and 200 nm were present. In addition, the location of many pits visualized with the SFM was coincident with membrane spots fluorescently labeled with a green fluorescent protein-caveolin-1 fusion protein. Scanning force microscopy on cells treated with methyl-beta-cyclodextrin, an agent that sequesters cholesterol and disrupts caveolae, abolished pits with a measured diameter of 100 nm but left pits of around 200 nm diameter intact. Thus, the smallest membrane pits measured with the SFM in CHO cells were indeed very likely to be identical to caveolae. These experiments show for the first time that SFM can be used to visualize caveolae in intact cells.
Collapse
Affiliation(s)
- H Lucius
- Department of Neuroscience, Max-Delbrück Institute for Molecular Medicine, D-13122 Berlin, Germany
| | | | | | | |
Collapse
|
46
|
Abstract
Caveolins are markers of caveolae, invaginations in the plasma membrane, and there are three members of the family in vertebrates. Caveolins participate in many important cellular processes, including vesicular transport, cholesterol homeostasis, signal transduction, and tumor suppression. The caveolin gene family has three members in vertebrates: caveolin-1, caveolin-2, and caveolin-3. So far, most caveolin-related research has been conducted in mammals, but the proteins have also been found in other animals, including Xenopus laevis, Fugu rubripes, and Caenorhabditis elegans. Caveolins can serve as protein markers of caveolae ('little caves'), invaginations in the plasma membrane 50-100 nanometers in diameter. Caveolins are found predominantly at the plasma membrane but also in the Golgi, the endoplasmic reticulum, in vesicles, and at cytosolic locations. They are expressed ubiquitously in mammals, but their expression levels vary considerably between tissues. The highest levels of caveolin-1 (also called caveolin, Cav-1 and VIP2I) are found in terminally-differentiated cell types, such as adipocytes, endothelia, smooth muscle cells, and type I pneumocytes. Caveolin-2 (Cav-2) is colocalized and coexpressed with Cav-1 and requires Cav-1 for proper membrane targeting; the Cav-2 gene also maps to the same chromosomal region as Cav-1 (7q31.1 in humans). Caveolin-3 (Cav-3) has greater protein-sequence similarity to Cav-1 than to Cav-2, but it is expressed mainly in muscle cells, including smooth, skeletal, and cardiac myocytes. Caveolins participate in many important cellular processes, including vesicular transport, cholesterol homeostasis, signal transduction, and tumor suppression.
Collapse
Affiliation(s)
- Terence M Williams
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
- The Albert Einstein Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | - Michael P Lisanti
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
- The Albert Einstein Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| |
Collapse
|
47
|
Abstract
The cholesterol-depleting drug methyl-beta-cyclodextrin (Me-beta-CD) was tested for its effects on amphibian oocyte maturation, cholesterol depletion, and low-density membrane recovery. Progesterone-induced oocyte maturation was accelerated by pretreatment of cells with 5-50 mM Me-beta-CD in a dose-dependent manner. Treatment of oocytes with 50 mM Me-beta-CD alone was sufficient to induce germinal vesicle breakdown, stimulate formation of meiotic spindles, and stimulate phosphorylation of mitogen-activated protein kinase over time courses longer than those observed after progesterone treatment. After short-term (30 min) labeling of oocytes with [(3)H]cholesterol, 30-90 min of treatment with 5-50 mM Me-beta-CD removed 50%-70% of cell- associated label, and cholesterol depletion was not observed with alpha-cyclodextrin. After long-term (20-23 h) labeling of oocytes with [(3)H]cholesterol, Me-beta-CD treatment resulted in dose- dependent cholesterol depletion in the 5-50 mM range, and 50 mM Me-beta-CD removed approximately 50% of cell-associated label after 9 h. Treatment of oocytes with 5-50 mM Me-beta-CD also decreased recovery of low-density membrane by detergent-free sucrose gradient centrifugation. These results implicate cholesterol and low-density membrane domains in the signaling mechanisms leading to germinal vesicle breakdown in amphibian oocytes.
Collapse
Affiliation(s)
- Susan E Sadler
- Department of Biological Sciences, University of Denver, Denver, Colorado 80208, USA.
| | | |
Collapse
|
48
|
Zurzolo C, van Meer G, Mayor S. The order of rafts. Conference on microdomains, lipid rafts and caveolae. EMBO Rep 2003; 4:1117-21. [PMID: 14634694 PMCID: PMC1326424 DOI: 10.1038/sj.embor.7400032] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2003] [Accepted: 10/24/2003] [Indexed: 01/08/2023] Open
Affiliation(s)
- Chiara Zurzolo
- Dipartimento di Biologia e
Patologia Cellulare e Molecolare, Università Federico II,
Via Pansini 5-80131, Napoli, Italy
- Pasteur Institute, 25 rue du Dr
Roux 75724, Paris, France
- Tel: +39 081 545 3033; Fax: +39 081 770 1016;
| | - Gerrit van Meer
- Department of Membrane Enzymology, Institute of
Biomembranes, Utrecht University, Padualaan 8,
3584 CH Utrecht, the
Netherlands
| | - Satyajit Mayor
- National Centre for Biological Sciences, Tata
Institute of Fundamental Research, UAS-GKVK Campus, Bangalore
560 065, Karnataka, India
- Tel: +91 80 363 6421/29; Fax; +91 80 363 6662/75;
| |
Collapse
|
49
|
Abstract
Cholesterol is a structural component of animal membranes that influences fluidity, permeability and formation of lipid microdomains. It is also a precursor to signalling molecules, including mammalian steroid hormones and insect ecdysones. The nematode Caenorhabditis elegans requires too little cholesterol for it to have a major role in membrane structure. Instead, its most probable signalling functions are to control molting and induce a specialized non-feeding larval stage, although no cholesterol-derived signalling molecule has yet been identified for these or any other functions.
Collapse
Affiliation(s)
- Teymuras V Kurzchalia
- Max Planck Institute for Molecular Cell Biology and Genetics, Dresden, D-01307, Germany.
| | | |
Collapse
|
50
|
Capozza F, Williams TM, Schubert W, McClain S, Bouzahzah B, Sotgia F, Lisanti MP. Absence of caveolin-1 sensitizes mouse skin to carcinogen-induced epidermal hyperplasia and tumor formation. THE AMERICAN JOURNAL OF PATHOLOGY 2003; 162:2029-39. [PMID: 12759258 PMCID: PMC1868132 DOI: 10.1016/s0002-9440(10)64335-0] [Citation(s) in RCA: 122] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Caveolin-1 is the principal protein component of caveolae membrane domains, which are located at the cell surface in most cell types. Evidence has accumulated suggesting that caveolin-1 may function as a suppressor of cell transformation in cultured cells. The human CAV-1 gene is located at a putative tumor suppressor locus (7q31.1/D7S522) and a known fragile site (FRA7G) that is deleted in a variety of epithelial-derived tumors. Mechanistically, caveolin-1 is known to function as a negative regulator of the Ras-p42/44 MAP kinase cascade and as a transcriptional repressor of cyclin D1, possibly explaining its transformation suppressor activity in cultured cells. However, it remains unknown whether caveolin-1 functions as a tumor suppressor gene in vivo. Here, we examine the tumor suppressor function of caveolin-1 using Cav-1 (-/-) null mice as a model system. Cav-1 null mice and their wild-type counterparts were subjected to carcinogen-induced skin tumorigenesis, using 7,12-dimethylbenzanthracene (DMBA). Mice were monitored weekly for the development of tumors. We demonstrate that Cav-1 null mice are dramatically more susceptible to carcinogen-induced tumorigenesis, as they develop skin tumors at an increased rate. After 16 weeks of DMBA-treatment, Cav-1 null mice showed a 10-fold increase in tumor incidence, a 15-fold increase in tumor number per mouse (multiplicity), and a 35-fold increase in tumor area per mouse, as compared with wild-type littermate mice. Moreover, before the development of tumors, DMBA-treatment induced severe epidermal hyperplasia in Cav-1 null mice. Both the basal cell layer and the suprabasal cell layers were expanded in treated Cav-1 null mice, as evidenced by immunostaining with cell-type specific differentiation markers (keratin-10 and keratin-14). In addition, cyclin D1 and phospho-ERK1/2 levels were up-regulated during epidermal hyperplasia, suggesting a possible mechanism for the increased susceptibility of Cav-1 null mice to tumorigenesis. However, the skin of untreated Cav-1 null mice appeared normal, without any evidence of epidermal hyperplasia, despite the fact that Cav-1 null keratinocytes failed to express caveolin-1 and showed a complete ablation of caveolae formation. Thus, Cav-1 null mice require an appropriate oncogenic stimulus, such as DMBA treatment, to reveal their increased susceptibility toward epidermal hyperplasia and skin tumor formation. Our results provide the first genetic evidence that caveolin-1 indeed functions as a tumor suppressor gene in vivo.
Collapse
Affiliation(s)
- Franco Capozza
- Department of Molecular Pharmacology and The Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | | | | | | | | | | | | |
Collapse
|