Chapter Nine - Recent Progress in the Topology, Structure, and Oligomerization of Caveolin: A Building Block of Caveolae

https://doi.org/10.1016/bs.ctm.2015.03.007Get rights and content

Abstract

Caveolae are cholesterol-rich plasma membrane invaginations that are found in a plethora of cell types. They play many roles including signal transduction, endocytosis, and mechanoprotection. The most critical protein in caveolae is the integral membrane protein, caveolin, which has been shown to be necessary for caveolae formation, and governs the major functions attributed to caveolae. Caveolin is postulated to act as a scaffold in the high molecular weight striated coat that surrounds the caveolar bulb, stabilizing it. Caveolin interacts, both directly and indirectly, with a large number of signaling molecules, and presides over the endocytosis of molecular cargo by caveolae. However, many of the key biophysical aspects of the caveolin protein, its structure, topology, and oligomeric behavior, are just beginning to come to light. Herein is an up-to-date summary and critique of the progress that has been made in understanding caveolin on a molecular and atomic level.

Introduction

Caveolae are 50–100 nm invaginations in the plasma membrane that are found in many cell types such as adipocytes, endothelial cells, fibroblasts, and smooth muscle cells (Mobley and Eisenberg, 1975, Palade, 1953, Thorn et al., 2003, Wheaton et al., 2001). Using electron microscopy, caveolae were first observed in the 1950s as flask-shaped domains within the heart and also the gall bladder (Palade, 1953, Yamada, 1955). Caveolae are proposed to have three major functions in the cell: mechanoprotection; recruiting, concentrating, and effecting signaling molecules; and endocytosis (Chang et al., 1994, Lisanti et al., 1994, Liu et al., 1997, Parton et al., 1994). Caveolae act as membrane reservoirs that allow cells to sustain mechanical stresses (Sinha et al., 2011). Facilitated by their highly curved nature, caveolae can bud off from the membrane in an endocytic manner and form vesicle-like structures that can carry cargo from the cell membrane to other parts of the cell (Kiss, 2012).

At the center of all of these functions is the integral membrane protein caveolin. Initially, there were two key proteins that were associated with caveolae, VIP-21 and caveolin. Caveolin was first isolated as a marker protein for caveolae, while VIP-21 was identified as part of the trans-golgi network (Kurzchalia et al., 1992, Rothberg et al., 1992). Shortly afterward, VIP-21 was sequenced and shown to be the same protein as caveolin (Glenney, 1992). This led to the consensus of calling the protein VIP-21/caveolin and now the protein is referred to simply as caveolin.

Caveolin interacts and effects, both directly and indirectly, a large number of signaling molecules including Src-like kinases, Ha-Ras, endothelial nitric oxide synthase (eNOS), and G-proteins (Carman et al., 1999, Couet et al., 1997, Ju et al., 1997, Lisanti et al., 1994, Okamoto et al., 1998, Roy et al., 1999, Sargiacomo et al., 1993). One well-studied interaction is between caveolin (specifically caveolin-1) and eNOS (García-Cardeña et al., 1997, Ghosh et al., 1998, Ju et al., 1997). The disruption of the binding between caveolin-1 and eNOS has negative effects on cellular function by causing a buildup of nitric oxide within the cell, as caveolin acts as a negative regulator of eNOS (Feron, Saldana, Michel, & Michel, 1998). Initially, many of the caveolin-signaling protein interactions were proposed to occur directly through a consensus sequence dubbed the caveolin-binding motif (CBM). However, recent structural data and bioinformatic analyses suggest that this motif (CBM) is not indicative of direct caveolin-partner interactions (Byrne et al., 2012, Collins et al., 2012). Although these studies call into question the direct interaction of caveolin with signaling molecules through a well-defined consensus motif, they do not preclude a specific nonconsensus interface for caveolin-partner interactions. Clearly, more work will be needed to establish which caveolin-partner interactions are direct versus indirect.

Caveolin-1 has been shown to play an integral role in regulating and mediating the endocytosis of caveolae. For example, the overexpression of caveolin-1 appears to inhibit the endocytosis of caveolae (Le, Guay, Altschuler, & Nabi, 2002). Caveolin-1 has also been implicated in the endocytosis of virus particles through caveolae (Pelkmans, Kartenbeck, & Helenius, 2001). Clearly, caveolin is involved in how molecules are transported from the plasma membrane to other parts of the cell through non-clathrin-mediated pathways.

Caveolae allow for a cell to experience abrupt mechanical stress without being deformed or lysed (Sinha et al., 2011). For example, when a cell is subjected to mechanical force, caveolae rapidly dissipate to allow the cell to accommodate the additional force. When the force is removed, caveolae are reformed.

Caveolin is an approximately 22 kDa protein that can be described by four domains: the N-terminal domain, the scaffolding domain, the intramembrane domain (IMD), and the C-terminal domain (Figure 1). Caveolin has three isoforms, caveolin-1, -2, and -3 (Figure 2). Caveolin-1 is the most ubiquitous, being found in most terminally differentiated cell types, and the protein has been shown to be necessary for caveolae formation (Drab et al., 2001, Park et al., 2002, Razani et al., 2001, Razani et al., 2002). Caveolin-1 also has two specific isoforms referred to as caveolin-1α and caveolin-1β. The caveolin-1β isoform can be thought of as a truncated construct of the α-isoform lacking the first 31 amino acid residues (Scherer et al., 1995). Caveolin-2 is coexpressed with caveolin-1 and the two are thought to form heterooligomeric complexes that are part of the striated coat (Scherer et al., 1996). However, caveolin-2 appears to lack the ability to form caveolae in the absence of caveolin-1 (Mora et al., 1999, Parolini et al., 1999). Caveolin-3 is expressed mostly in muscle tissue and also has the ability to form caveolae (Song et al., 1996, Tang et al., 1996, Way and Parton, 1995). This is supported by the high degree of sequence identity between caveolin-1 and -3. In contrast, the sequence of caveolin-2 deviates from that of the other isoforms, particularly in its scaffolding domain (Figure 2). In addition, the lengths of the N-terminal domains vary significantly between the isoforms, with caveolin-1 having the longest and caveolin-3 having the shortest (Figure 2).

Caveolin-1 has been shown to be palmitoylated at all three of its cysteine residues located in or near the C-terminal domain (C133, C143, and C156). The removal of this posttranslational modification by mutation of the cysteines to serines has been shown to have no effect on the trafficking of caveolin-1 to the plasma membrane (Dietzen, Hastings, & Lublin, 1995). Caveolin-2 has five cysteines and it has recently been shown that three of its cysteines are palmitoylated (Kwon, Lee, Jeong, Jang, & Pak, 2015). Caveolin-3 contains nine cysteines and has been shown to be heavily palmitoylated, but the exact sites have not been experimentally verified (Galbiati, Volonte, Minetti, Chu, & Lisanti, 1999). However, because of the sequence similarity to caveolin-1, it has been assumed that caveolin-3 is palmitoylated at the analogous three sites (Figure 2) (Dietzen et al., 1995, Galbiati et al., 1999).

The composition of caveolae is distinct compared to the bulk membrane because it is enriched in cholesterol and sphingomyelin (Ortegren et al., 2004). This has led to the postulation that caveolae have significantly less fluidity than the bulk membrane, a composition not unlike that of lipid rafts. The relationship between cholesterol and caveolin appears to be very strong and has been implicated in the proper folding and behavior of caveolin (Murata et al., 1995). In particular, the scaffolding domain of caveolin contains a CRAC (cholesterol recognition amino acid consensus) motif that is purported to directly bind cholesterol (Epand, Sayer, & Epand, 2005).

Mutations in the caveolin proteins as well as cellular expression levels have been linked to many diseases such as cancer, diabetes, cardiovascular disease, artherosclerosis, pulmonary fibrosis, and many muscular degenerative diseases (Capozza et al., 2005, Cohen et al., 2003, Fernández-Hernando et al., 2010, Komers et al., 2006, Sotgia et al., 2005, Weiss et al., 2008, Williams et al., 2004, Xia et al., 2010). The literature on the links between caveolin and disease is extensive and is beyond the scope and purpose of this review; therefore, only a few examples will be highlighted (For extensive reviews of caveolin and disease, see Engelman et al., 1998, Razani and Lisanti, 2001; Williams & Lisanti, 2004, and others). The knockout of caveolin-1 in mice can cause an increase in tumor proliferation indicating that caveolin-1 is a tumor suppressor (Sloan et al., 2004, Williams et al., 2003). However, in prostate cancer it is found that caveolin-1 is upregulated and that it assists in the metastasis of the cancer (Williams et al., 2005). This dual role of caveolin-1 in cancer highlights that normal levels of caveolin-1 expressed within specific cell types is critical for maintaining proper cell function. Other studies have also shown that the overexpression of caveolin-1 can mirror the phenotype of a putative disease mutant (P132L) implicated in breast cancer by causing a disruption of caveolin migration from the Golgi to the plasma membrane (Bonuccelli et al., 2009, Hanson et al., 2013). However the true role and extent that this mutation appears in affected patients is unclear (Koike et al., 2010, Lacroix-Triki et al., 2010). Caveolin-3 has been linked to many muscular diseases such as heart disease and muscular dystrophy (Horikawa et al., 2008, Weiss et al., 2008). P104 in caveolin-3 which is analogous to P132 in caveolin-1 is also implicated in increased disease states and misregulation of the protein (Ohsawa et al., 2004, Stoppani et al., 2011).

Despite caveolin's preeminent role in cell homeostasis, many of the biophysical details surrounding caveolin function remain elusive. Much like other proteins whose improper function has been linked to disease states, a complete understanding of the determinants of cellular disruption will only come to fruition once a clear picture of the caveolin protein is obtained in molecular and atomic detail. This review will strive to report the progress that has been made in the field of caveolin structure, topology, and oligomerization and to highlight the work that still needs to be done.

Section snippets

Caveolins have an unusual topology

Caveolin-1 has been shown to be an integral membrane protein using carbonate extraction, a method which distinguishes between peripherally bound membrane proteins and integral membrane proteins (Dupree et al., 1993, Schlegel et al., 1999). Primary sequence analysis of caveolin-1 indicates that the protein has an unusually long 33-amino acid hydrophobic segment (residues 102–134, IMD, Figure 1). The length of the caveolin hydrophobic segment is much longer than the typical 15–20 amino acid

Caveolin Structure

The structure of caveolin clearly has implications for how it governs its many proposed functions. Within this section the progress of understanding the structure of caveolin-1, -2, and -3 will be discussed.

Caveolin-1 homo-oligomerization

The first hint that caveolin may oligomerize arose from the observation that caveolin-1 (VIP21) was part of a complex derived from trans-Golgi network vesicles isolated from MDCK (Madin–Darby canine kidney) cells (Kurzchalia et al., 1992). These complexes were isolated based on their insolubility in the mild detergents, Triton X-114 and CHAPS (3-[(3-Cholamidopropyl)dimethylammonio]-1-propanesulfonate). This result was followed shortly afterward with a study that analyzed these complexes

Conclusions

Caveolae are a critical component of the eukaryotic plasma membrane. In the time since their discovery, the number of the functions and events postulated to be caveolae localized is both diverse and immense. The central player involved in the formation and function of these microdomains is the integral protein caveolin, whose importance is belied by the fact that mutation and misregulation of the protein results in disease states. Much is known about the involvement of this protein in disease

References (107)

  • S. Aoki et al.

    Caveolin-1 hydrophobic segment peptides insertion into membrane mimetic systems: role of proline residue

    Biochimica et Biophysica Acta

    (2012)
  • S. Aoki et al.

    The role of proline in the membrane re-entrant helix of caveolin-1

    The Journal of Biological Chemistry

    (2010)
  • M. Bastiani et al.

    MURC/Cavin-4 and cavin family members form tissue-specific caveolar complexes

    The Journal of Cell Biology

    (2009)
  • G. Bonuccelli et al.

    Caveolin-1 (P132L), a common breast cancer mutation, confers mammary cell invasiveness and defines a novel stem cell/metastasis-associated gene signature

    The American Journal of Pathology

    (2009)
  • D.P. Byrne et al.

    Evaluating caveolin interactions: do proteins interact with the caveolin scaffolding domain through a widespread aromatic residue-rich motif?

    PloS One

    (2012)
  • F. Capozza et al.

    Caveolin-3 knockout mice show increased adiposity and whole body insulin resistance, with ligand-induced insulin receptor instability in skeletal muscle

    American Journal of Physiology–Cell Physiology

    (2005)
  • C.V. Carman et al.

    Regulation of G protein-coupled receptor kinases by caveolin

    Journal of Biological Chemistry

    (1999)
  • W.J. Chang et al.

    Purification and characterization of smooth muscle cell caveolae

    Journal of Cell Biology

    (1994)
  • A.W. Cohen et al.

    Caveolin-1 null mice develop cardiac hypertrophy with hyperactivation of p42/44 MAP kinase in cardiac fibroblasts

    American Journal of Physiology–Cell Physiology

    (2003)
  • B.M. Collins et al.

    Structure-based reassessment of the caveolin signaling model: do caveolae regulate signaling through caveolin-protein interactions?

    Developmental Cell

    (2012)
  • J. Couet et al.

    Interaction of a receptor tyrosine kinase, EGF-R, with caveolins – caveolin binding negatively regulates tyrosine and serine/threonine kinase activities

    The Journal of Biological Chemistry

    (1997)
  • K. Das et al.

    The membrane-spanning domains of caveolins-1 and -2 mediate the formation of caveolin hetero-oligomers. Implications for the assembly of caveolae membranes in vivo

    The Journal of Biological Chemistry

    (1999)
  • D.J. Dietzen et al.

    Caveolin is palmitoylated on multiple cysteine residues. Palmitoylation is not necessary for localization of caveolin to caveolae

    The Journal of Biological Chemistry

    (1995)
  • M. Drab et al.

    Loss of caveolae, vascular dysfunction, and pulmonary defects in caveolin-1 gene-disrupted mice

    Science

    (2001)
  • P. Dupree et al.

    Caveolae and sorting in the trans-golgi network of epithelial cells

    The EMBO Journal

    (1993)
  • J.A. Engelman et al.

    Molecular genetics of the caveolin gene family: Implications for human cancers, diabetes, Alzheimer disease, and muscular dystrophy

    American Journal of Human Genetics

    (1998)
  • R.M. Epand et al.

    Caveolin scaffolding region and cholesterol-rich domains in membranes

    Journal of Molecular Biology

    (2005)
  • I. Fernandez et al.

    Mechanism of caveolin filament assembly

    Proceedings of the National Academy of Sciences of the United States of America

    (2002)
  • C. Fernández-Hernando et al.

    Endothelial-specific overexpression of caveolin-1 accelerates atherosclerosis in apolipoprotein E-deficient mice

    The American Journal of Pathology

    (2010)
  • O. Feron et al.

    The endothelial nitric-oxide synthase-caveolin regulatory cycle

    The Journal of Biological Chemistry

    (1998)
  • F. Galbiati et al.

    Phenotypic behavior of caveolin-3 mutations that cause autosomal dominant limb girdle muscular dystrophy (LGMD-1C). Retention of LGMD-1C caveolin-3 mutants within the golgi complex

    Journal of Biological Chemistry

    (1999)
  • Y. Gambin et al.

    Single-molecule analysis reveals self assembly and nanoscale segregation of two distinct cavin subcomplexes on caveolae

    Elife

    (2013)
  • G. García-Cardeña et al.

    Dissecting the interaction between nitric oxide synthase (NOS) and caveolin: functional significance of the NOS caveolin binding domain in vivo

    The Journal of Biological Chemistry

    (1997)
  • S. Ghosh et al.

    Interaction between caveolin-1 and the reductase domain of endothelial nitric-oxide synthase. Consequences for catalysis

    The Journal of Biological Chemistry

    (1998)
  • J.R. Glenney

    The sequence of human caveolin reveals identity with VIP21, a component of transport vesicles

    FEBS Letters

    (1992)
  • Han, B., Tiwari, A., & Kenworthy, A. K. Tagging strategies strongly impact the fate of overexpressed caveolin-1....
  • C.G. Hansen et al.

    SDPR induces membrane curvature and functions in the formation of caveolae

    Nature Cell Biology

    (2009)
  • C.G. Hansen et al.

    Deletion of cavin genes reveals tissue-specific mechanisms for morphogenesis of endothelial caveolae

    Nature Communications

    (2013)
  • C.A. Hanson et al.

    Overexpression of caveolin-1 is sufficient to phenocopy the behavior of a disease-associated mutant

    Traffic

    (2013)
  • A. Hayer et al.

    Biogenesis of caveolae: stepwise assembly of large caveolin and cavin complexes

    Traffic

    (2010)
  • M. Hill et al.

    PTRF-cavin, a conserved cytoplasmic protein required for caveola formation and function

    Cell

    (2008)
  • C.L. Hoop et al.

    Structural characterization of the caveolin scaffolding domain in association with cholesterol-rich membranes

    Biochemistry

    (2012)
  • Y.T. Horikawa et al.

    Caveolin-3 expression and caveolae are required for isoflurane-induced cardiac protection from hypoxia and ischemia/reperfusion injury

    Journal of Molecular and Cell Cardiology

    (2008)
  • H. Ju et al.

    Direct interaction of endothelial nitric-oxide synthase and caveolin-1 inhibits synthase activity

    The Journal of Biological Chemistry

    (1997)
  • J. Kim et al.

    Modest effects of lipid modifications on the structure of caveolin-3

    Biochemistry

    (2014)
  • A. Kiss

    Caveolae and the regulation of endocytosis

    Advances in Experimental Medicine and Biology

    (2012)
  • S. Koike et al.

    Absence of the caveolin-1 P132L mutation in cancers of the breast and other organs

    The Journal of Molecular Diagnostics

    (2010)
  • R. Komers et al.

    Altered endothelial nitric oxide synthase targeting and conformation and caveolin-1 expression in the diabetic kidney

    Diabetes

    (2006)
  • O. Kovtun et al.

    Structural insights into the organization of the cavin membrane coat complex

    Developmental Cell

    (2014)
  • T.V. Kurzchalia et al.

    VIP21, a 21-kD membrane protein is an integral component of trans-golgi-network-derived transport vesicles

    The Journal of Cell Biology

    (1992)
  • H. Kwon et al.

    Fatty acylated caveolin-2 is a substrate of insulin receptor tyrosine kinase for insulin receptor substrate-1-directed signaling activation

    Biochimica et Biophysica Acta

    (2015)
  • M. Lacroix-Triki et al.

    Caveolin-1 P132L mutation in human cancers: 1 CAVeat to be voiced

    The Journal of Molecular Diagnostics

    (2010)
  • P.U. Le et al.

    Caveolin-1 is a negative regulator of caveolae-mediated endocytosis to the endoplasmic reticulum

    The Journal of Biological Chemistry

    (2002)
  • C. Le Lan et al.

    Structural and dynamic properties of juxta-membrane segments of caveolin-1 and caveolin-2 at the membrane interface

    European Biophysics Journal

    (2010)
  • C. Le Lan et al.

    Role of the membrane interface on the conformation of the caveolin scaffolding domain: a CD and NMR study

    FEBS Letters

    (2006)
  • J. Lee et al.

    The transmembrane domain of caveolin-1 exibits a helix-break-helix structure

    Biochimica et Biophysica Acta

    (2012)
  • H. Lee et al.

    Caveolin-1 mutations (P132L and null) and the pathogenesis of breast cancer – caveolin-1 (P132L) behaves in a dominant-negative manner and caveolin-1 (−/−) null mice show mammary epithelial cell hyperplasia

    American Journal of Pathology

    (2002)
  • M.P. Lisanti et al.

    Characterization of caveolin-rich membrane domains isolated from an endothelial-rich source: implications for human-disease

    The Journal of Cell Biology

    (1994)
  • L. Liu et al.

    Deletion of Cavin/PTRF causes global loss of caveolae, dyslipidemia, and glucose intolerance

    Cell Metabolism

    (2008)
  • J. Liu et al.

    Organized endothelial cell surface signal transduction in caveolae distinct from glycosylphosphatidylinositol-anchored protein microdomains

    The Journal of Biological Chemistry

    (1997)
  • Cited by (0)

    a

    These two authors contributed equally to this work.

    View full text