Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2009 Aug 13.
Published in final edited form as: Mol Cell Endocrinol. 2008 Apr 22;290(1-2):8–13. doi: 10.1016/j.mce.2008.04.005

Caveolin Proteins and Estrogen Signaling in the Brain

Jessie I Luoma 1, Marissa I Boulware 1, Paul G Mermelstein 1
PMCID: PMC2565274  NIHMSID: NIHMS65990  PMID: 18502030

Abstract

Best described outside the nervous system, caveolins are structural proteins that form caveolae, functional microdomains at the plasma membrane that cluster related signaling molecules. Caveolin associated proteins include G protein coupled receptors and G proteins, receptor tyrosine kinases, as well as protein kinases, ion channels and various other signaling enzymes. Not surprisingly, a wide array of biological disorders are thought to be rooted in caveolin dysfunction. In addition, caveolins also traffic and cluster estrogen receptors to caveolae. Interactions between the estrogen receptors ERα and ERβ with caveolins appear critical in many non-neuronal cell types, e.g. disruption of normal function may underlie many forms of breast cancer. Recent findings suggest caveolins may also play an essential role in membrane estrogen receptor function in the nervous system. Not only are they expressed in neurons and glia, but different caveolin isoforms also appear necessary to generate distinct functional signaling complexes. With membrane estrogen receptors responsible for the efficient activation of a multitude of intracellular signaling pathways, which in turn influence a wide variety of nervous system functions, caveolin proteins are poised to act as the central coordinators of these processes.

Keywords: estradiol, estrogen receptor, mGluR, lordosis, membrane receptor, rapid actions, caveolae, flotillin

Introduction

The role of estradiol on brain function and its consequent influence upon behavior has been studied for over 60 years. Once thought to be the sole mechanism of estrogen action, estradiol binding to the intracellular estrogen receptors ERα and ERβ acts to affect gene expression and protein synthesis [1,2]. This classical mechanism of estrogen action, i.e stimulation of steroid regulated transcription factors, plays a crucial role in brain functions involving sexual development, sexual maturation, and the expression of sexual receptivity [38]. However, in addition to its actions on intracellular estrogen receptors, estradiol can also affect a variety of cellular processes through stimulation of surface membrane receptors. Not only have these rapid acting effects of estrogens been shown to play a role in sex behavior, but also in brain and spinal cord regions involved with, but not limited to, learning and memory, motor function, nociception and drug addiction [915]. The majority of these reported membrane-initiated actions of estradiol in the nervous system appear dependent on a subpopulation of ERα and/or ERβ that are localized to the membrane surface [1618], but see [1921]. To this end, several biological questions remain unanswered: (1) how are intracellular estrogen receptors trafficked to the membrane, and (2) once in the membrane, how are estrogen receptors targeted to the appropriate signaling complexes for the precise activation of specific intracellular signaling cascades? This review will focus on the putative role caveolin proteins may play in mediating these two processes.

Caveolins: Important for the Trafficking and Clustering of Membrane-Associated Signaling Proteins

Caveolin proteins are the fundamental components of caveolae, which form distinct structural and functional microdomains in many cell types [22,23]. Caveolae when associated with the plasma membrane exhibit invaginations described as omega- or cave-like structures that cluster functionally related membrane-associated proteins [24]. Caveolae were first recognized in epithelial cells by electron micrograph techniques over 50 years ago [25]. There are three known caveolin proteins, caveolin 1 (CAV1, with splice variants α and β), caveolin 2 (CAV2), and caveolin 3 (CAV3) [2628]. CAV1 and CAV2 have overlapping expression patterns in a variety of cell types including, neurons and glia [29,30], endothelial [31], and epithelial cells [32]. Disruption of CAV2 expression does not affect caveolae formation in vivo [33], inasmuch it is hypothesized that CAV2 only forms caveolae as hetero-oligomers with CAV1, and not in isolation [34]. In comparison, knockout of either CAV1 or CAV3 results in a loss of caveolae formations in the specific cell type for which they are expressed [35,36]. Notably, it was originally believed that expression of CAV3 was restricted to skeletal and smooth muscle cells [26,3739]. We now know this not to be the case, as expression of CAV3 is more widespread, including its presence in nervous tissue [30].

The cavernous structure of caveolae supports a functional domain where various proteins can cluster and associate for efficient activation of discrete signaling pathways. As such, caveolae are often described as signaling regulators that serve to orchestrate the interaction of receptors and signaling molecules, modulating transmembrane signaling in a rapid and specific manner [40,41]. This is thought to occur via direct protein-protein interactions between caveolins and signaling components found at the plasma membrane. In various cell types, caveolin proteins have been shown to be associated with G protein-coupled receptors, G protein subunits, tyrosine kinase receptors, various intracellular kinases, voltage-gated ion channels, ion pumps, and various second messenger molecules [4245].

In addition to its role of clustering related signaling molecules, caveolin proteins also play a role in the trafficking of various receptors to the membrane. Surface receptors in which trafficking to the membrane has been reported to be dependent on caveolin function include, but are not limited to, the D1 dopamine receptor [46], M1 muscarinic receptor [47], angiotensin II type 1 receptor [48], and glucagon-like peptide 1 receptor [49]. Notably, caveolins also play a role in receptor endocytosis [47,5054] providing an additional regulatory mechanism to modulate cell signaling. Caveolin-dependent endocytosis is a mechanism involving internalization of membrane components within caveolae resulting in the diminution of function. Caveolin-dependent sequestration of receptors can be thought of as a means to negatively modulate signaling via the storage of signaling complexes within the cell.

With their importance for the trafficking and clustering of various signaling transduction molecules, it stands to reason caveolins play critical roles in many cellular processes. Indeed, alteration/disruption of caveolin expression has been implicated, breast cancer [5557], vascular abnormalities [5860], pulmonary malfunction [33], and muscle disease [35,37].

Caveolin Proteins and Estrogen Receptors

A functional link between caveolin proteins and membrane estrogen receptors was first reported in non-neuronal cells approximately ten years ago. Initial studies identified ERα-dependent nitric oxide (NO) production required ERα to be associated with caveolae that contained endothelial nitric oxide synthase (eNOS) [61,62]. Concurrent with these studies, CAV1 was shown to potentiate classical ERα-mediated gene expression [63], a process dependent upon the direct interaction between ERα and CAV1 [63]. Thus, the signaling of estradiol via classical and novel actions appears to be intertwined, with activation of each influenced by the other. Furthermore, estradiol appears to directly influence ERα interaction with CAV1 [63,64], and estrogens modulate expression of caveolins [65], providing additional levels of regulation. Of note, ERβ can also associate with caveolae, and they too are functionally coupled to enzymatic signaling machinery via this process. For example, ERβ within eNOS containing caveolae affords estradiol/ERβ regulation of NO production [66].

A necessary step in estrogen receptor localization to caveolae is the palmitoylation of the receptor. Although research has focused primarily on palmitoylation of ERα and its variants [64,6769], the same mechanism for caveolae association has been described for ERβ [64,70]. Specifically, palmitoylation of human ERα at cysteine 447 (mouse 451) is essential for receptor interaction with CAV1 and its subsequent localization to the plasma membrane. In CHO cells, mutation of cysteine 447 to an alanine results in a loss of membrane ERα. In addition, the physical interaction between ERα and CAV1 is abolished, and membrane estrogen effects are eliminated [64,70]. It is through regulation of palmitoylation that estradiol appears to affect the interaction between ERα and CAV1. In particular, stimulation of HELA cells with estradiol reduces ERα binding to CAV1 with a corresponding reduction in membrane ERα [64,70]. It is significant to note that it is the palmitoylation of a single amino acid that regulates the trafficking of ERα, as this residue is well conserved across species. Similarly, a single cysteine residue in ERβ (mouse 418) appears to be the critical amino acid for palmitoylation and trafficking to the membrane. The question to which enzyme(s) is/are responsible for palmitoylation of the estrogen receptors remains unanswered. As 23 separate palmitoyl acyl transferases are known to exist and appear to be ubiquitously expressed across various tissues [71], this determination may not be rapidly forthcoming.

A second amino acid within the ligand-binding domain of ERα is also necessary for interaction of ERα with CAV1. Mutation of mouse serine 522 to an alanine reduces ERα binding to CAV1 by ~60% in CHO cells and reduces membrane-localization of ERα by a similar percentage [72]. The S522A mutation also acts as a dominant-negative in relation to membrane estrogen receptor signaling [73]. Currently, the mechanism by which serine 522 facilitates ERα binding to CAV1 and caveolin-dependent trafficking of the receptor to the membrane remains unknown. Mutation of residues within the palmitoylation motif (ERα: F449A, IL456-7AA; ERβ: Y416A, IL423-4AA), have also been shown to interfere with palmitoylation and membrane localization of mouse ERα and ERβ.

The physiological relevance of caveolin proteins and estrogen receptors is best described in relationship to breast cancer. Both ERα and caveolin proteins have been implicated in breast cancer etiology, as alterations in the expression or function of either is prevalent in many forms of the disease. In addition, proliferation of estrogen receptor-positive breast cancer cells has been shown to be sensitive and facilitated by estrogen [74], which may be a result of elevated levels of estrogen receptors in breast cancer tissue [75] as these proliferative effects of estrogen can be reduced by treatment with anti-estrogens [76]. Of note is the finding that cell cycle progression in breast cancer cells has been shown to be mediated through mechanisms activated by membrane-localized estrogen receptors [77]. Mutations of CAV1 are found in ~35% of ERα-positive human breast cancer samples [78], and CAV1 RNA and protein levels are reduced in many cases of human primary breast carcinomas [79]. Mechanistically, it has been theorized that these alterations in CAV1 function or expression contribute to the increased sensitivity of mammary tissue to estrogen by leading to an increase of ERα expression [78,80]. Moreover, overexpression of CAV1 in mammary tissue protects against the tumorigenic phenotype that mammary tissue lacking CAV1 shows [81]. Collectively, these data indicate a possible role for both proteins in the biology of a subset of breast cancers that may result from a loss of caveolin-mediated modulation of estrogen receptor availability in breast cancer tissue. This provides additional support for a mechanism of membrane estrogen signaling that depends on interactions between caveolins and estrogen receptors.

In terms of caveolin proteins and nervous system function, until a decade ago, it was believed caveolin expression was limited to glial cells [82]. Thus, when first hypothesizing that caveolae-like structures may too be responsible for the localization of membrane estrogen receptors in neurons, a review article by Dominique Toran-Allerand discussed structures comprised of the caveolin-related, flotillin proteins [21]. Indeed, caveolar-like microdomains (CLMs) were termed as the neuronal counterpart to non-neuronal caveolae. More recently, however, various reports have demonstrated the expression off all three caveolin isoforms in neurons [29,30,8386]. Data described below is consistent with reports from non-neuronal tissue and the initial descriptions in Dominique Toran-Allerand’s review that caveolin proteins play an essential role in brain membrane estrogen receptor function.

Our experiments in hippocampal neurons delineated two distinct signaling pathways by which membrane estrogen receptors regulate cell function. The first pathway found estradiol to activate ERα, leading to stimulation of the metabotropic glutamate receptor mGluR1a. This in turn led to activation of Gq, PLC, IP3 and MAPK signaling, and eventually, phosphorylation of the transcription factor CREB. The second pathway was initiated by estradiol activation of either ERα or ERβ. Under these conditions, stimulation of mGluR2/3 led to Gi/o signaling, with a subsequent decrease in L-type calcium channel currents due to inhibition of PKA. As such, L-type calcium channel-dependent CREB phosphorylation was attenuated [87]

In a follow-up report, we found CAV1 and CAV3 to be responsible for the segregation and functional compartmentalization of these two distinct signaling pathways. ERα activation of mGluR1a signaling was dependent on CAV1. Correspondingly, ERα/ERβ activation of mGluR2/3 was dependent on CAV3 [73]. Presumably, CAV1 is responsible for the localization of ERα with mGluR1a and its downstream signaling partners. Conversely, CAV3 traffics and/or clusters either ERα or ERβ with mGluR2/3 and its second messengers (Figure 1). These data are both consistent with and expand upon previous work. Previous studies have not only indicated CAV1 to localize with ERα, but also mGluR1a [88]. Similarly, L-type calcium channels cluster in CAV3-generated caveolae [89,90]. As such, we find several individual signaling molecules linked to caveolin proteins to which they were previously ascribed. In addition, distinct caveolae generated by different caveolin isoforms, responsible for the functional isolation of separate signaling pathways in the same cell illustrates the complexity by which neurons transmit information from the membrane surface.

Figure 1.

Figure 1

Caveolin proteins are responsible for the isolation of functionally distinct signaling pathways in neurons. Schematic of caveolin 1 and caveolin 3 comprised caveolae in hippocampal neurons. Caveolin 1 is required for ERα activation of mGluR1a signaling, leading to MAPK-dependent CREB phosphorylation. In comparison, caveolin 3 mediates ERα or ERβ activation of mGluR2/3 signaling, leading to a decrease in L-type calcium channel-dependent CREB phosphorylation.

The hypothesis that CAV1 and CAV3 play important roles in membrane estrogen receptor signaling expands to paradigms outside the hippocampus. mGluRs appear required for membrane estrogen receptor signaling in several other systems including neurons of the arcuate nucleus [91], striatal neurons [92], dorsal root ganglia [93] and hypothalamic astrocytes [94]. Thus far, it has been determined that caveolin proteins functionally link estrogen receptors with mGluRs in striatal neurons [92]; other regions await verification. Through influencing these additional regions, rapid estrogen receptor signaling has been suggested to modulate multiple cellular processes, such as motor control and drug addiction [12,9597], sexual receptivity [91], the control of the estrous cycle [98], and nociception [14,99]. Furthermore, while we and others hypothesize GPCRs to act as intermediaries between estrogen receptors and G proteins, others have postulated that estrogen receptors directly activate the G proteins [100]. This alternative hypothesis does not negate the possibility that caveolin proteins are essential for membrane estrogen receptor function. In fact, with caveolin proteins clustering specific G proteins, as well as GPCRs, their influence on membrane estrogen receptor signaling within the nervous system may be far reaching. Consistent with this idea, there are many parallels between caveolin and estrogen receptor function in brain. A wealth of information regarding caveolins and their influence on brain function has recently emerged. Topics include, but are not limited to, caveolin regulation of synaptic strength, motor control, and Alzheimer’s disease [101106]. These studies demonstrate that caveolins play critical roles in both neuronal and glial processes. Interestingly, each of these processes has also been demonstrated to be under the regulation of estrogens [9,107110].

It is intriguing to consider that divergent functions in neurons are often localized to discrete functional regions [111,112]. The notion of subcellular compartmentalization of various signaling and effector proteins has been best demonstrated in neurons in regard to transsynaptic signaling. For example, PDZ domain-containing scaffolding proteins have been studied in great detail, elucidating their significance in the localization and clustering of proper neurotransmitter receptors, downstream second messengers, and cytoskeletal proteins [111]. Thus, the separation of Gq-coupled mGluR1a and Gi/o-coupled group II mGluRs, imparted by their localization within distinct caveolae, makes sense in terms of glutamatergic synaptic function. The spatial segregation of various estrogen receptor-dependent signaling pathways is necessitated by the isolation of separate glutamatergic receptors, but can it also be significant in terms of steroid-regulated processes? We now know that estrogens can be synthesized and released in various brain regions [113116]. Based upon these data, it has been suggested that estrogens may play physiological roles similar to neurotransmitters [117]. Thus, whereas ovarian estradiol reaching the hippocampus would most likely activate both mGluR signaling pathways simultaneously, locally synthesized estradiol could potentially activate one pathway over another. Increased temporal and spatial control over estradiol-sensitive signaling by the synthesis and release of the steroid in brain is an exciting possibility that deserves future study.

Conclusions

Caveolae play an important role in the organization of signal transduction by shuttling signaling proteins and molecules to and from the membrane surface. At the membrane surface they may function by partitioning signaling components into particular combinations, increasing specificity without increasing complexity. Additionally, caveolae can rapidly modulate signaling through an endocytotic mechanism by which the caveolae internalize receptors or other signaling components so that they are non-functional. These endocytosed structures can remain near the membrane surface so that they can be made available for signaling by reintroduction to the membrane surface facilitated by the caveolae [46,118,119].

Membrane estrogen receptors can affect a variety of cellular processes both in and outside the nervous system. Caveolin proteins have been demonstrated to play critical roles in many of these systems, whereby they both traffic estrogen receptors as well as cluster the additional signaling machinery at the membrane surface into functional caveolae. While current research in brain has been limited to examination of estradiol regulation of CREB signaling, undoubtedly many other cellular processes dependent on membrane estrogen receptors will too require the proper functioning of caveolins.

Acknowledgements

This work was supported by NIH grant NS41302.

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • 1.McKenna NJ, Lanz RB, O'Malley BW. Nuclear receptor coregulators: cellular and molecular biology. Endocr Rev. 1999;20:321–344. doi: 10.1210/edrv.20.3.0366. [DOI] [PubMed] [Google Scholar]
  • 2.McInerney EM, Weis KE, Sun J, Mosselman S, Katzenellenbogen BS. Transcription activation by the human estrogen receptor subtype beta (ER beta) studied with ER beta and ER alpha receptor chimeras. Endocrinology. 1998;139:4513–4522. doi: 10.1210/endo.139.11.6298. [DOI] [PubMed] [Google Scholar]
  • 3.Odor DL. The temporal relationship of the first maturation division of rat ova to the onset of heat. Am. J. Anat. 1955;97:461–491. doi: 10.1002/aja.1000970306. [DOI] [PubMed] [Google Scholar]
  • 4.Gorski RA, Yanase M. Estrogen facilitation of lordosis behavior in the female rat. Exp Brain Res Suppl. 1981;3:222–237. doi: 10.1007/978-3-642-45525-4_18. [DOI] [PubMed] [Google Scholar]
  • 5.Young WC, Boling JL, Blandau RJ. The vaginal smear picture, sexual receptivity and time of ovulation in the albino rat. Anat. Rec. 1941;80:37–45. [Google Scholar]
  • 6.Levine S, Mullins R., Jr Estrogen Administered Neonatally Affects Adult Sexual Behavior in Male and Female Rats. Science. 1964;144:185–187. doi: 10.1126/science.144.3615.185. [DOI] [PubMed] [Google Scholar]
  • 7.Feder HH, Whalen RE. Feminine Behavior in Neonatally Castrated and Estrogen-Treated Male Rats. Science. 1965;147:306–307. doi: 10.1126/science.147.3655.306. [DOI] [PubMed] [Google Scholar]
  • 8.Booth JE. Sexual behaviour of neonatally castrated rats injected during infancy with oestrogen and dihydrotestosterone. J Endocrinol. 1977;72:135–141. doi: 10.1677/joe.0.0720135. [DOI] [PubMed] [Google Scholar]
  • 9.McEwen BS, Alves SE. Estrogen actions in the central nervous system. Endocr Rev. 1999;20:279–307. doi: 10.1210/edrv.20.3.0365. [DOI] [PubMed] [Google Scholar]
  • 10.McEwen B. Estrogen actions throughout the brain. Recent Prog Horm Res. 2002;57:357–384. doi: 10.1210/rp.57.1.357. [DOI] [PubMed] [Google Scholar]
  • 11.Becker D, Creutzfeldt OD, Schwibbe M, Wuttke W. Changes in physiological, EEG and psychological parameters in women during the spontaneous menstrual cycle and following oral contraceptives. Psychoneuroendocrinology. 1982;7:75–90. doi: 10.1016/0306-4530(82)90057-9. [DOI] [PubMed] [Google Scholar]
  • 12.Hampson E, Kimura D. Reciprocal effects of hormonal fluctuations on human motor and perceptual-spatial skills. Behav Neurosci. 1988;102:456–459. doi: 10.1037//0735-7044.102.3.456. [DOI] [PubMed] [Google Scholar]
  • 13.Hampson E. Estrogen-related variations in human spatial and articulatory-motor skills. Psychoneuroendocrinology. 1990;15:97–111. doi: 10.1016/0306-4530(90)90018-5. [DOI] [PubMed] [Google Scholar]
  • 14.Riley JL, 3rd, Robinson ME, Wise EA, Price DD. A meta-analytic review of pain perception across the menstrual cycle. Pain. 1999;81:225–235. doi: 10.1016/S0304-3959(98)00258-9. [DOI] [PubMed] [Google Scholar]
  • 15.Brinton RD. Cellular and molecular mechanisms of estrogen regulation of memory function and neuroprotection against Alzheimer's disease: recent insights and remaining challenges. Learn Mem. 2001;8:121–133. doi: 10.1101/lm.39601. [DOI] [PubMed] [Google Scholar]
  • 16.McCarthy MM. Out with the new, in with the old: classical estrogen receptors mediate novel estradiol actions in brain. Endocrinology. 2004;145:3053–3054. doi: 10.1210/en.2004-0336. [DOI] [PubMed] [Google Scholar]
  • 17.Vasudevan N, Pfaff DW. Non-genomic actions of estrogens and their interaction with genomic actions in the brain. Front Neuroendocrinol. 2007 doi: 10.1016/j.yfrne.2007.08.003. [DOI] [PubMed] [Google Scholar]
  • 18.Boulware MI, Mermelstein PG. The influence of estradiol on nervous system function. Drug News Perspect. 2005;18:631–637. doi: 10.1358/dnp.2005.18.10.959578. [DOI] [PubMed] [Google Scholar]
  • 19.Prossnitz ER, Arterburn JB, Sklar LA. GPR30: A G protein-coupled receptor for estrogen. Mol Cell Endocrinol. 2007:265–266. 138–142. doi: 10.1016/j.mce.2006.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Ronnekleiv OK, Malyala A, Kelly MJ. Membrane-initiated signaling of estrogen in the brain. Semin Reprod Med. 2007;25:165–177. doi: 10.1055/s-2007-973429. [DOI] [PubMed] [Google Scholar]
  • 21.Toran-Allerand CD. Minireview: A plethora of estrogen receptors in the brain: where will it end? Endocrinology. 2004;145:1069–1074. doi: 10.1210/en.2003-1462. [DOI] [PubMed] [Google Scholar]
  • 22.Glenney JR, Jr, Soppet D. Sequence and expression of caveolin, a protein component of caveolae plasma membrane domains phosphorylated on tyrosine in Rous sarcoma virus-transformed fibroblasts. Proc Natl Acad Sci U S A. 1992;89:10517–10521. doi: 10.1073/pnas.89.21.10517. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Rothberg KG, Heuser JE, Donzell WC, Ying YS, Glenney JR, Anderson RG. Caveolin, a protein component of caveolae membrane coats. Cell. 1992;68:673–682. doi: 10.1016/0092-8674(92)90143-z. [DOI] [PubMed] [Google Scholar]
  • 24.Parton RG, Hanzal-Bayer M, Hancock JF. Biogenesis of caveolae: a structural model for caveolin-induced domain formation. J Cell Sci. 2006;119:787–796. doi: 10.1242/jcs.02853. [DOI] [PubMed] [Google Scholar]
  • 25.Yamada E. The fine structure of the gall bladder epithelium of the mouse. J Biophys Biochem Cytol. 1955;1:445–458. doi: 10.1083/jcb.1.5.445. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Tang Z, Scherer PE, Okamoto T, Song K, Chu C, Kohtz DS, Nishimoto I, Lodish HF, Lisanti MP. Molecular cloning of caveolin-3, a novel member of the caveolin gene family expressed predominantly in muscle. J Biol Chem. 1996;271:2255–2261. doi: 10.1074/jbc.271.4.2255. [DOI] [PubMed] [Google Scholar]
  • 27.Scherer PE, Okamoto T, Chun M, Nishimoto I, Lodish HF, Lisanti MP. Identification, sequence, and expression of caveolin-2 defines a caveolin gene family. Proc Natl Acad Sci U S A. 1996;93:131–135. doi: 10.1073/pnas.93.1.131. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Glenney JR., Jr The sequence of human caveolin reveals identity with VIP21, a component of transport vesicles. FEBS Lett. 1992;314:45–48. doi: 10.1016/0014-5793(92)81458-x. [DOI] [PubMed] [Google Scholar]
  • 29.Galbiati F, Volonte D, Gil O, Zanazzi G, Salzer JL, Sargiacomo M, Scherer PE, Engelman JA, Schlegel A, Parenti M, Okamoto T, Lisanti MP. Expression of caveolin-1 and -2 in differentiating PC12 cells and dorsal root ganglion neurons: caveolin-2 is up-regulated in response to cell injury. Proc Natl Acad Sci U S A. 1998;95:10257–10262. doi: 10.1073/pnas.95.17.10257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ikezu T, Ueda H, Trapp BD, Nishiyama K, Sha JF, Volonte D, Galbiati F, Byrd AL, Bassell G, Serizawa H, Lane WS, Lisanti MP, Okamoto T. Affinity-purification and characterization of caveolins from the brain: differential expression of caveolin-1, -2, and -3 in brain endothelial and astroglial cell types. Brain Res. 1998;804:177–192. doi: 10.1016/s0006-8993(98)00498-3. [DOI] [PubMed] [Google Scholar]
  • 31.Lisanti MP, Scherer PE, Vidugiriene J, Tang Z, Hermanowski-Vosatka A, Tu YH, Cook RF, Sargiacomo M. Characterization of caveolin-rich membrane domains isolated from an endothelial-rich source: implications for human disease. J Cell Biol. 1994;126:111–126. doi: 10.1083/jcb.126.1.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Vogel U, Sandvig K, van Deurs B. Expression of caveolin-1 and polarized formation of invaginated caveolae in Caco-2 and MDCK II cells. J Cell Sci. 1998;111(Pt 6):825–832. doi: 10.1242/jcs.111.6.825. [DOI] [PubMed] [Google Scholar]
  • 33.Razani B, Wang XB, Engelman JA, Battista M, Lagaud G, Zhang XL, Kneitz B, Hou H, Jr, Christ GJ, Edelmann W, Lisanti MP. Caveolin-2-deficient mice show evidence of severe pulmonary dysfunction without disruption of caveolae. Mol Cell Biol. 2002;22:2329–2344. doi: 10.1128/MCB.22.7.2329-2344.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Scherer PE, Lewis RY, Volonte D, Engelman JA, Galbiati F, Couet J, Kohtz DS, van Donselaar E, Peters P, Lisanti MP. Cell-type and tissue-specific expression of caveolin-2. Caveolins 1 and 2 co-localize and form a stable hetero-oligomeric complex in vivo. J Biol Chem. 1997;272:29337–29346. doi: 10.1074/jbc.272.46.29337. [DOI] [PubMed] [Google Scholar]
  • 35.Galbiati F, Engelman JA, Volonte D, Zhang XL, Minetti C, Li M, Hou H, Jr, Kneitz B, Edelmann W, Lisanti MP. Caveolin-3 null mice show a loss of caveolae, changes in the microdomain distribution of the dystrophin-glycoprotein complex, and t-tubule abnormalities. J Biol Chem. 2001;276:21425–21433. doi: 10.1074/jbc.M100828200. [DOI] [PubMed] [Google Scholar]
  • 36.Razani B, Combs TP, Wang XB, Frank PG, Park DS, Russell RG, Li M, Tang B, Jelicks LA, Scherer PE, Lisanti MP. Caveolin-1-deficient mice are lean, resistant to diet-induced obesity, and show hypertriglyceridemia with adipocyte abnormalities. J Biol Chem. 2002;277:8635–8647. doi: 10.1074/jbc.M110970200. [DOI] [PubMed] [Google Scholar]
  • 37.Song KS, Scherer PE, Tang Z, Okamoto T, Li S, Chafel M, Chu C, Kohtz DS, Lisanti MP. Expression of caveolin-3 in skeletal, cardiac, and smooth muscle cells. Caveolin-3 is a component of the sarcolemma and co-fractionates with dystrophin and dystrophin-associated glycoproteins. J Biol Chem. 1996;271:15160–15165. doi: 10.1074/jbc.271.25.15160. [DOI] [PubMed] [Google Scholar]
  • 38.Way M, Parton RG. M-caveolin, a muscle-specific caveolin-related protein. FEBS Lett. 1995;376:108–112. doi: 10.1016/0014-5793(95)01256-7. [DOI] [PubMed] [Google Scholar]
  • 39.Chang WJ, Ying YS, Rothberg KG, Hooper NM, Turner AJ, Gambliel HA, De Gunzburg J, Mumby SM, Gilman AG, Anderson RG. Purification and characterization of smooth muscle cell caveolae. J Cell Biol. 1994;126:127–138. doi: 10.1083/jcb.126.1.127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Lisanti MP, Scherer PE, Tang Z, Sargiacomo M. Caveolae, caveolin and caveolin-rich membrane domains: a signalling hypothesis. Trends Cell Biol. 1994;4:231–235. doi: 10.1016/0962-8924(94)90114-7. [DOI] [PubMed] [Google Scholar]
  • 41.Simons K, Toomre D. Lipid rafts and signal transduction. Nat Rev Mol Cell Biol. 2000;1:31–39. doi: 10.1038/35036052. [DOI] [PubMed] [Google Scholar]
  • 42.Anderson RG. Plasmalemmal caveolae and GPI-anchored membrane proteins. Curr Opin Cell Biol. 1993;5:647–652. doi: 10.1016/0955-0674(93)90135-d. [DOI] [PubMed] [Google Scholar]
  • 43.Stefanova I, Horejsi V, Ansotegui IJ, Knapp W, Stockinger H. GPI-anchored cell-surface molecules complexed to protein tyrosine kinases. Science. 1991;254:1016–1019. doi: 10.1126/science.1719635. [DOI] [PubMed] [Google Scholar]
  • 44.Anderson RG. The caveolae membrane system. Annu Rev Biochem. 1998;67:199–225. doi: 10.1146/annurev.biochem.67.1.199. [DOI] [PubMed] [Google Scholar]
  • 45.Patel HH, Murray F, Insel PA. Caveolae as organizers of pharmacologically relevant signal transduction molecules. Annu Rev Pharmacol Toxicol. 2008;48:359–391. doi: 10.1146/annurev.pharmtox.48.121506.124841. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Kong MM, Hasbi A, Mattocks M, Fan T, O'Dowd BF, George SR. Regulation of D1 dopamine receptor trafficking and signaling by caveolin-1. Mol Pharmacol. 2007;72:1157–1170. doi: 10.1124/mol.107.034769. [DOI] [PubMed] [Google Scholar]
  • 47.Shmuel M, Nodel-Berner E, Hyman T, Rouvinski A, Altschuler Y. Caveolin 2 regulates endocytosis and trafficking of the M1 muscarinic receptor in MDCK epithelial cells. Mol Biol Cell. 2007;18:1570–1585. doi: 10.1091/mbc.E06-07-0618. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Wyse BD, Prior IA, Qian H, Morrow IC, Nixon S, Muncke C, Kurzchalia TV, Thomas WG, Parton RG, Hancock JF. Caveolin interacts with the angiotensin II type 1 receptor during exocytic transport but not at the plasma membrane. J Biol Chem. 2003;278:23738–23746. doi: 10.1074/jbc.M212892200. [DOI] [PubMed] [Google Scholar]
  • 49.Syme CA, Zhang L, Bisello A. Caveolin-1 regulates cellular trafficking and function of the glucagon-like Peptide 1 receptor. Mol Endocrinol. 2006;20:3400–3411. doi: 10.1210/me.2006-0178. [DOI] [PubMed] [Google Scholar]
  • 50.Lajoie P, Nabi IR. Regulation of raft-dependent endocytosis. J Cell Mol Med. 2007;11:644–653. doi: 10.1111/j.1582-4934.2007.00083.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Pelkmans L, Helenius A. Endocytosis via caveolae. Traffic. 2002;3:311–320. doi: 10.1034/j.1600-0854.2002.30501.x. [DOI] [PubMed] [Google Scholar]
  • 52.Hommelgaard AM, Roepstorff K, Vilhardt F, Torgersen ML, Sandvig K, Van Deurs B. Caveolae: stable membrane domains with a potential for internalization. Traffic. 2005;6:720–724. doi: 10.1111/j.1600-0854.2005.00314.x. [DOI] [PubMed] [Google Scholar]
  • 53.Becher A, McIlhinney RA. Consequences of lipid raft association on G-protein-coupled receptor function. Biochem Soc Symp. 2005:151–164. doi: 10.1042/bss0720151. [DOI] [PubMed] [Google Scholar]
  • 54.de Weerd WF, Leeb-Lundberg LM. Bradykinin sequesters B2 bradykinin receptors and the receptor-coupled Galpha subunits Galphaq and Galphai in caveolae in DDT1 MF-2 smooth muscle cells. J Biol Chem. 1997;272:17858–17866. doi: 10.1074/jbc.272.28.17858. [DOI] [PubMed] [Google Scholar]
  • 55.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. Am J Pathol. 2006;168:292–309. doi: 10.2353/ajpath.2006.050429. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Williams TM, Medina F, Badano I, Hazan RB, Hutchinson J, Muller WJ, Chopra NG, Scherer PE, Pestell RG, Lisanti MP. Caveolin-1 gene disruption promotes mammary tumorigenesis and dramatically enhances lung metastasis in vivo. Role of Cav-1 in cell invasiveness and matrix metalloproteinase (MMP-2/9) secretion. J Biol Chem. 2004;279:51630–51646. doi: 10.1074/jbc.M409214200. [DOI] [PubMed] [Google Scholar]
  • 57.Chen ST, Lin SY, Yeh KT, Kuo SJ, Chan WL, Chu YP, Chang JG. Mutational, epigenetic and expressional analyses of caveolin-1 gene in breast cancers. Int J Mol Med. 2004;14:577–582. [PubMed] [Google Scholar]
  • 58.Li XA, Everson WV, Smart EJ. Caveolae, lipid rafts, and vascular disease. Trends Cardiovasc Med. 2005;15:92–96. doi: 10.1016/j.tcm.2005.04.001. [DOI] [PubMed] [Google Scholar]
  • 59.Drab M, Verkade P, Elger M, Kasper M, Lohn M, Lauterbach B, Menne J, Lindschau C, Mende F, Luft FC, Schedl A, Haller H, Kurzchalia TV. Loss of caveolae, vascular dysfunction, and pulmonary defects in caveolin-1 gene-disrupted mice. Science. 2001;293:2449–2452. doi: 10.1126/science.1062688. [DOI] [PubMed] [Google Scholar]
  • 60.Razani B, Engelman JA, Wang XB, Schubert W, Zhang XL, Marks CB, Macaluso F, Russell RG, Li M, Pestell RG, Di Vizio D, Hou H, Jr, Kneitz B, Lagaud G, Christ GJ, Edelmann W, Lisanti MP. Caveolin-1 null mice are viable but show evidence of hyperproliferative and vascular abnormalities. J Biol Chem. 2001;276:38121–38138. doi: 10.1074/jbc.M105408200. [DOI] [PubMed] [Google Scholar]
  • 61.Kim HP, Lee JY, Jeong JK, Bae SW, Lee HK, Jo I. Nongenomic stimulation of nitric oxide release by estrogen is mediated by estrogen receptor alpha localized in caveolae. Biochem Biophys Res Commun. 1999;263:257–262. doi: 10.1006/bbrc.1999.1348. [DOI] [PubMed] [Google Scholar]
  • 62.Chambliss KL, Yuhanna IS, Mineo C, Liu P, German Z, Sherman TS, Mendelsohn ME, Anderson RG, Shaul PW. Estrogen receptor alpha and endothelial nitric oxide synthase are organized into a functional signaling module in caveolae. Circ Res. 2000;87:E44–E52. doi: 10.1161/01.res.87.11.e44. [DOI] [PubMed] [Google Scholar]
  • 63.Schlegel A, Wang C, Katzenellenbogen BS, Pestell RG, Lisanti MP. Caveolin-1 potentiates estrogen receptor alpha (ERalpha) signaling. caveolin-1 drives ligand-independent nuclear translocation and activation of ERalpha. J Biol Chem. 1999;274:33551–33556. doi: 10.1074/jbc.274.47.33551. [DOI] [PubMed] [Google Scholar]
  • 64.Acconcia F, Ascenzi P, Bocedi A, Spisni E, Tomasi V, Trentalance A, Visca P, Marino M. Palmitoylation-dependent estrogen receptor alpha membrane localization: regulation by 17beta-estradiol. Mol Biol Cell. 2005;16:231–237. doi: 10.1091/mbc.E04-07-0547. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zschocke J, Manthey D, Bayatti N, van der Burg B, Goodenough S, Behl C. Estrogen receptor alpha-mediated silencing of caveolin gene expression in neuronal cells. J Biol Chem. 2002;277:38772–38780. doi: 10.1074/jbc.M205664200. [DOI] [PubMed] [Google Scholar]
  • 66.Chambliss KL, Shaul PW. Estrogen modulation of endothelial nitric oxide synthase. Endocr Rev. 2002;23:665–686. doi: 10.1210/er.2001-0045. [DOI] [PubMed] [Google Scholar]
  • 67.Li L, Haynes MP, Bender JR. Plasma membrane localization and function of the estrogen receptor alpha variant (ER46) in human endothelial cells. Proc Natl Acad Sci USA. 2003;100:4807–4812. doi: 10.1073/pnas.0831079100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Acconcia F, Ascenzi P, Fabozzi G, Visca P, Marino M. S-palmitoylation modulates human estrogen receptor-alpha functions. Biochem Biophys Res Commun. 2004;316:878–883. doi: 10.1016/j.bbrc.2004.02.129. [DOI] [PubMed] [Google Scholar]
  • 69.Pedram A, Razandi M, Aitkenhead M, Hughes CC, Levin ER. Integration of the non-genomic and genomic actions of estrogen. Membrane-initiated signaling by steroid to transcription and cell biology. J Biol Chem. 2002;277:50768–50775. doi: 10.1074/jbc.M210106200. [DOI] [PubMed] [Google Scholar]
  • 70.Pedram A, Razandi M, Sainson RC, Kim JK, Hughes CC, Levin ER. A conserved mechanism for steroid receptor translocation to the plasma membrane. J Biol Chem. 2007;282:22278–22288. doi: 10.1074/jbc.M611877200. [DOI] [PubMed] [Google Scholar]
  • 71.Mitchell DA, Vasudevan A, Linder ME, Deschenes RJ. Protein palmitoylation by a family of DHHC protein S-acyltransferases. J Lipid Res. 47:1118–1127. doi: 10.1194/jlr.R600007-JLR200. [DOI] [PubMed] [Google Scholar]
  • 72.Razandi M, Alton G, Pedram A, Ghonshani S, Webb P, Levin ER. Identification of a structural determinant necessary for the localization and function of estrogen receptor alpha at the plasma membrane. Mol Cell Biol. 2003;23:1633–1646. doi: 10.1128/MCB.23.5.1633-1646.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Boulware MI, Kordasiewicz H, Mermelstein PG. Caveolin proteins are essential for distinct effects of membrane estrogen receptors in neurons. J Neurosci. 2007;27:9941–9950. doi: 10.1523/JNEUROSCI.1647-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Anderson E, Clarke RB. Steroid receptors and cell cycle in normal mammary epithelium. J Mammary Gland Biol Neoplasia. 2004;9:3–13. doi: 10.1023/B:JOMG.0000023584.01750.16. [DOI] [PubMed] [Google Scholar]
  • 75.Shoker BS, Jarvis C, Clarke RB, Anderson E, Hewlett J, Davies MP, Sibson DR, Sloane JP. Estrogen receptor-positive proliferating cells in the normal and precancerous breast. Am J Pathol. 1999;155:1811–1815. doi: 10.1016/S0002-9440(10)65498-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Song RX, McPherson RA, Adam L, Bao Y, Shupnik M, Kumar R, Santen RJ. Linkage of rapid estrogen action to MAPK activation by ERalpha-Shc association and Shc pathway activation. Mol Endocrinol. 2002;16:116–127. doi: 10.1210/mend.16.1.0748. [DOI] [PubMed] [Google Scholar]
  • 77.Razandi M, Pedram A, Rosen EM, Levin ER. BRCA1 inhibits membrane estrogen and growth factor receptor signaling to cell proliferation in breast cancer. Mol Cell Biol. 2004;24:5900–5913. doi: 10.1128/MCB.24.13.5900-5913.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Li T, Sotgia F, Vuolo MA, Li M, Yang WC, Pestell RG, Sparano JA, Lisanti MP. Caveolin-1 mutations in human breast cancer: functional association with estrogen receptor alpha-positive status. Am J Pathol. 2006;168:1998–2013. doi: 10.2353/ajpath.2006.051089. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Bouras T, Lisanti MP, Pestell RG. Caveolin-1 in breast cancer. Cancer Biol Ther. 2004;3:931–941. doi: 10.4161/cbt.3.10.1147. [DOI] [PubMed] [Google Scholar]
  • 80.Adams BD, Furneaux H, White BA. The micro-ribonucleic acid (miRNA) miR-206 targets the human estrogen receptor-alpha (ERalpha) and represses ERalpha messenger RNA and protein expression in breast cancer cell lines. Mol Endocrinol. 2007;21:1132–1147. doi: 10.1210/me.2007-0022. [DOI] [PubMed] [Google Scholar]
  • 81.Williams TM, Sotgia F, Lee H, Hassan G, Di Vizio D, Bonuccelli G, Capozza F, Mercier I, Rui H, Pestell RG, Lisanti MP. Stromal and epithelial caveolin-1 both confer a protective effect against mammary hyperplasia and tumorigenesis: Caveolin-1 antagonizes cyclin D1 function in mammary epithelial cells. Am J Pathol. 2006;169:1784–1801. doi: 10.2353/ajpath.2006.060590. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Cameron PL, Ruffin JW, Bollag R, Rasmussen H, Cameron RS. Identification of caveolin and caveolin-related proteins in the brain. J Neurosci. 1997;17:9520–9535. doi: 10.1523/JNEUROSCI.17-24-09520.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Cabello OA, Schilling WP. Vectorial Ca2+ flux from the extracellular space to the endoplasmic reticulum via a restricted cytoplasmic compartment regulates inositol 1,4,5-trisphosphate-stimulated Ca2+ release from internal stores in vascular endothelial cells. Biochem J. 1993;295:357–366. doi: 10.1042/bj2950357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Mikol DD, Hong HL, Cheng HL, Feldman EL. Caveolin-1 expression in Schwann cells. Glia. 1999;27:39–52. [PubMed] [Google Scholar]
  • 85.Gaudreault SB, Blain JF, Gratton JP, Poirier J. A role for caveolin-1 in post-injury reactive neuronal plasticity. J Neurochem. 2005;92:831–839. doi: 10.1111/j.1471-4159.2004.02917.x. [DOI] [PubMed] [Google Scholar]
  • 86.Masserini M, Palestini P, Pitto M. Glycolipid-enriched caveolae and caveolae-like domains in the nervous system. J Neurochem. 1999;73:1–11. doi: 10.1046/j.1471-4159.1999.0730001.x. [DOI] [PubMed] [Google Scholar]
  • 87.Boulware MI, Weick JP, Becklund BR, Kuo SP, Groth RD, Mermelstein Estradiol activates group I and II metabotropic glutamate receptor signaling, leading to opposing influences on cAMP response element-binding protein. J Neurosci. 2005;25:5066–5078. doi: 10.1523/JNEUROSCI.1427-05.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Burgueno J, Canela EI, Mallol J, Lluis C, Franco R, Ciruela F. Mutual regulation between metabotropic glutamate type 1alpha receptor and caveolin proteins: from traffick to constitutive activity. Exp Cell Res. 2004;300:23–34. doi: 10.1016/j.yexcr.2004.06.013. [DOI] [PubMed] [Google Scholar]
  • 89.Balijepalli RC, Foell JD, Hall DD, Hell JW, Kamp TJ. Localization of cardiac L-type Ca(2+) channels to a caveolar macromolecular signaling complex is required for beta(2)-adrenergic regulation. Proc Natl Acad Sci U S A. 2006;103:7500–7505. doi: 10.1073/pnas.0503465103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Couchoux H, Allard B, Legrand C, Jacquemond V, Berthier C. Loss of caveolin-3 induced by the dystrophy-associated P104L mutation impairs L-type calcium channel function in mouse skeletal muscle cells. J Physiol. 2007;580:745–754. doi: 10.1113/jphysiol.2006.124198. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Dewing P, Boulware MI, Sinchak K, Christensen A, Mermelstein PG, Micevych P. Membrane estrogen receptor-alpha interactions with metabotropic glutamate receptor 1a modulate female sexual receptivity in rats. J Neurosci. 2007;27:9294–9300. doi: 10.1523/JNEUROSCI.0592-07.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Grove-Strawser D, Mermelstein PG. Estrogen receptors activate different mGluRs across distinct brain regions. Abstract 195.13/XX10; Society for Neuroscience Annual Meeting 2007; San Diego, CA. 2007. [Google Scholar]
  • 93.Chaban VV, Li J, McDonald J, Rapkin A, Micevych P. Estradiol attenuates ATP-induced increase of [Ca2+]i through group II metabotropic glutamate receptors in rat DRG neurons. Abstract 519.7/WW16; Society for Neuroscience Annual Meeting 2007; SanDiego, CA. 2007. [Google Scholar]
  • 94.Micevych P, Sinchak K. Mini-reviews: Synthesis and function of hypothalamic neuroprogesterone in reproduction. Endocrinology. 2008 doi: 10.1210/en.2008-0011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Becker JB, Snyder PJ, Miller MM, Westgate SA, Jenuwine MJ. The influence of estrous cycle and intrastriatal estradiol on sensorimotor performance in the female rat. Pharmacol Biochem Behav. 1987;27:53–59. doi: 10.1016/0091-3057(87)90476-x. [DOI] [PubMed] [Google Scholar]
  • 96.Xiao L, Jackson LR, Becker JB. The effect of estradiol in the striatum is blocked by ICI 182,780 but not tamoxifen: pharmacological and behavioral evidence. Neuroendocrinology. 2003;77:239–245. doi: 10.1159/000070279. [DOI] [PubMed] [Google Scholar]
  • 97.Gajjar TM, Anderson LI, Dluzen DE. Acute effects of estrogen upon methamphetamine induced neurotoxicity of the nigrostriatal dopaminergic system. J Neural Transm. 2003;110:1215–1224. doi: 10.1007/s00702-003-0045-3. [DOI] [PubMed] [Google Scholar]
  • 98.Micevych PE, Chaban V, Ogi J, Dewing P, Lu JK, Sinchak K. Estradiol stimulates progesterone synthesis in hypothalamic astrocyte cultures. Endocrinology. 2007;148:782–789. doi: 10.1210/en.2006-0774. [DOI] [PubMed] [Google Scholar]
  • 99.Stoffel EC, Ulibarri CM, Craft RM. Gonadal steroid hormone modulation of nociception, morphine antinociception and reproductive indices in male and female rats. Pain. 2003;103:285–302. doi: 10.1016/s0304-3959(02)00457-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Vasudevan N, Pfaff DW. Membrane-initiated actions of estrogens in neuroendocrinology: emerging principles. Endocr Rev. 2007;28:1–19. doi: 10.1210/er.2005-0021. [DOI] [PubMed] [Google Scholar]
  • 101.Trushina E, Du Charme J, Parisi J, McMurray CT. Neurological abnormalities in caveolin-1 knock out mice. Behav Brain Res. 2006;172:24–32. doi: 10.1016/j.bbr.2006.04.024. [DOI] [PubMed] [Google Scholar]
  • 102.Ikezu T, Trapp BD, Song KS, Schlegel A, Lisanti MP, Okamoto T. Caveolae, plasma membrane microdomains for alpha-secretase-mediated processing of the amyloid precursor protein. J Biol Chem. 1998;273:10485–10495. doi: 10.1074/jbc.273.17.10485. [DOI] [PubMed] [Google Scholar]
  • 103.Nishiyama K, Trapp BD, Ikezu T, Ransohoff RM, Tomita T, Iwatsubo T, Kanazawa I, Hsiao KK, Lisanti MP, Okamoto T. Caveolin-3 upregulation activates beta-secretase-mediated cleavage of the amyloid precursor protein in Alzheimer's disease. J Neurosci. 1999;19:6538–6548. doi: 10.1523/JNEUROSCI.19-15-06538.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104.Braun JE, Madison DV. A novel SNAP25-caveolin complex correlates with the onset of persistent synaptic potentiation. J Neurosci. 2000;20:5997–6006. doi: 10.1523/JNEUROSCI.20-16-05997.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105.Arvanitis DN, Wang H, Bagshaw RD, Callahan JW, Boggs JM. Membrane-associated estrogen receptor and caveolin-1 are present in central nervous system myelin and oligodendrocyte plasma membranes. J Neurosci Res. 2004;75:603–613. doi: 10.1002/jnr.20017. [DOI] [PubMed] [Google Scholar]
  • 106.Gaudreault SB, Dea D, Poirier J. Increased caveolin-1 expression in Alzheimer's disease brain. Neurobiol Aging. 2004;25:753–759. doi: 10.1016/j.neurobiolaging.2003.07.004. [DOI] [PubMed] [Google Scholar]
  • 107.Paganini-Hill A, Henderson VW. Estrogen deficiency and risk of Alzheimer's disease in women. Am J Epidemiol. 1994;140:256–261. doi: 10.1093/oxfordjournals.aje.a117244. [DOI] [PubMed] [Google Scholar]
  • 108.Van der Horst VG, Holstege G. Sensory and motor components of reproductive behavior: pathways and plasticity. Behav Brain Res. 1998;92:157–167. doi: 10.1016/s0166-4328(97)00188-5. [DOI] [PubMed] [Google Scholar]
  • 109.Cordoba Montoya DA, Carrer HF. Estrogen facilitates induction of long term potentiation in the hippocampus of awake rats. Brain Res. 1997;778:430–438. doi: 10.1016/s0006-8993(97)01206-7. [DOI] [PubMed] [Google Scholar]
  • 110.Yankova M, Hart SA, Woolley CS. Estrogen increases synaptic connectivity between single presynaptic inputs and multiple postsynaptic CA1 pyramidal cells: a serial electron-microscopic study. Proc Natl Acad Sci U S A. 2001;98:3525–3530. doi: 10.1073/pnas.051624598. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Hata Y, Nakanishi H, Takai Y. Synaptic PDZ domain-containing proteins. Neurosci Res. 1998;32:1–7. doi: 10.1016/s0168-0102(98)00069-8. [DOI] [PubMed] [Google Scholar]
  • 112.Augustine GJ, Santamaria F, Tanaka K. Local calcium signaling in neurons. Neuron. 2003;40:331–346. doi: 10.1016/s0896-6273(03)00639-1. [DOI] [PubMed] [Google Scholar]
  • 113.Wehrenberg U, Prange-Kiel J, Rune GM. Steroidogenic factor-1 expression in marmoset and rat hippocampus: co-localization with StAR and aromatase. J Neurochem. 2001;76:1879–1886. doi: 10.1046/j.1471-4159.2001.00207.x. [DOI] [PubMed] [Google Scholar]
  • 114.Rune GM, Frotscher M. Neurosteroid synthesis in the hippocampus: role in synaptic plasticity. Neuroscience. 2005;136:833–842. doi: 10.1016/j.neuroscience.2005.03.056. [DOI] [PubMed] [Google Scholar]
  • 115.Zhou L, Lehan N, Wehrenberg U, Disteldorf E, von Lossow R, Mares U, Jarry H, Rune GM. Neuroprotection by estradiol: a role of aromatase against spine synapse loss after blockade of GABA(A) receptors. Exp Neurol. 2007;203:72–81. doi: 10.1016/j.expneurol.2006.07.020. [DOI] [PubMed] [Google Scholar]
  • 116.Mukai H, Tsurugizawa T, Ogiue-Ikeda M, Murakami G, Hojo Y, Ishii H, Kimoto T, Kawato S. Local neurosteroid production in the hippocampus: influence on synaptic plasticity of memory. Neuroendocrinology. 2006;84:255–263. doi: 10.1159/000097747. [DOI] [PubMed] [Google Scholar]
  • 117.Balthazart J, Ball GF. Is brain estradiol a hormone or a neurotransmitter? Trends Neurosci. 2006;29:241–249. doi: 10.1016/j.tins.2006.03.004. [DOI] [PubMed] [Google Scholar]
  • 118.Sigismund S, Woelk T, Puri C, Maspero E, Tacchetti C, Transidico P, Di Fiore PP, Polo S. Clathrin-independent endocytosis of ubiquitinated cargos. Proc Natl Acad Sci U S A. 2005;102:2760–2765. doi: 10.1073/pnas.0409817102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Di Guglielmo GM, Le Roy C, Goodfellow AF, Wrana JL. Distinct endocytic pathways regulate TGF-beta receptor signalling and turnover. Nat Cell Biol. 2003;5:410–421. doi: 10.1038/ncb975. [DOI] [PubMed] [Google Scholar]

RESOURCES