Open access

Caveolae-Dependent Endocytosis in Viral Infection

Written By

Norica Branza-Nichita, Alina Macovei and Catalin Lazar

Submitted: 12 April 2012 Published: 06 July 2012

DOI: 10.5772/48538

From the Edited Volume

Molecular Regulation of Endocytosis

Edited by Brian Ceresa

Chapter metrics overview

2,762 Chapter Downloads

View Full Metrics

1. Introduction

1.1. Caveolae structure and morphogenesis

Morphologically described as small “caves” in the plasma membrane, caveolae are highly specialized membrane domains with important roles in cell endocytosis, lipid metabolism, and signaling. Since their discovery sixty years ago [1,2], the functional relevance of caveolae has challenged many scientists, raising numerous debates and controversies. The electron microscopy images of caveolae show a rather cell-type-dependent appearance. In endothelial cells, caveolae opening is more constricted [3], while in epithelial cells they appear open to the extracellular medium and smaller in size [4]. In muscle cells, multiple caveolae units cluster together forming T-like tubules invaginating from sarcolemma [5]. Regardless their shape, caveolae appear as immobile structures, in tight connection with the cortical actin cytoskeleton underlying the plasma membrane. Video microscopy and fluorescence recovery after photobleaching analysis have shown that caveolae detach from the membrane only upon ligand binding and specific signaling [6].

The discovery of caveolins (Cav), the structural proteins enveloping caveolae in a spike-like coat [7] marked a significant breakthrough in understanding the nature and importance of these organelles.

Three members of the Cav family have been described in mammalian cells, to date: Cav-1, -2, -3, which share a significant homology and are conserved throughout evolution [8]. Cav-1 and -2 are relatively ubiquitous, with highest distribution in fibroblasts, adipocytes, endothelial cells, and pneumocytes, being co-expressed in most cells types [9]. Cav-3 is expressed independently of Cav-1 and -2 and is limited to skeletal muscle fibers and cardiac myocytes [10-12]. Over-expression of Cav-1 in caveolae-deficient cells is necessary and apparently sufficient to drive caveolae biogenesis [13]. Moreover, Cav-1 expression is required for the membrane localization and stability of Cav-2. Although unable to drive caveolae biogenesis on its own, Cav-2 may however influence it, at least in several polarized, epithelial cells [14,15]. The capacity to modulate caveolae assembly, shape and size has been shown to depend on Cav-2 phosphorylation status [16]. Similar to Cav-1, Cav-3 protein is sufficient to drive formation of caveolae in muscle cells [17].

Most of the molecular data available on caveolins refer to Cav-1; therefore the following discussion will focus on this protein, as a representative of the caveolin family, which is shown schematically in Fig.1.

Cav-1 is an integral membrane protein of 21kDa with an unusual topology. Both the N- and C-termini are cytoplasmically oriented and connected by a central hydrophobic domain, comprising approximately residues 102-134, inserted into, but not spanning the membrane bilayer, in a hairpin (or U bent, or horseshoe) configuration [18,19]. Interestingly, a peptide corresponding to the last 20 residues of the hydrophilic N-domain, (amino acids 82–101) enriched in aromatic residues, can also bind to membranes independently [20,21]. This so-called caveolin scaffolding domain (CSD) is a highly conserved region responsible for many functions associated with Cav-1.

In silico analysis of the conformation of this hydrophobic domain, showed that mutation of a single residue, Pro (110), changes the stable conformation to a straight hydrophobic helix that would span the membrane. Expression of the Cav-1 P110A mutant in HEK 293 cells followed by confocal immunofluorescence microscopy further confirmed the in silico data and the estimated topology [22].

Figure 1.

Schematic presentation of the structural similarities of the caveolin family of proteins.

Very recently, circular dichroism and NMR spectroscopy analysis have shown that the transmembrane domain of Cav-1 is primarily α-helical (57-65%). In addition, the helix–break–helix structure was suggested to be critical for the formation of the intra-membrane horseshoe conformation predicted for the protein. Interestingly, mutations of Ile (109) and Pro (110) to Ala dramatically altered the helix-break-helix structure. Moreover, it was shown that substitution of Pro (110) with any other residue results in disruption of the helix-break-helix structure, confirming the importance of the residue in the stability of the hydrophobic domain [23].

An important structural feature of Cav-1 is represented by its arrangement in high molecular mass oligomers of about 350 kDa, composed of 14-16 individual molecules [24,25]. Oligomerization is initiated in the endoplasmic reticulum (ER), where the Cav-1 monomer is co-translationally inserted into the membrane [19] and is rapidly assembled to form SDS-resistant, 8S complexes considered the building units of caveolae structure [24,25]. Intriguingly, blue native gel analysis evidenced only a few intermediate sized oligomers, suggesting that Cav-1 oligomerization is a highly cooperative process [26]. Oligomerization of the full-length protein requires the presence of the CSD and of the C-terminal domain [27] and appears to be stabilized by the palmitoylation of cysteine residues located at positions 133, 143 and 156 [18,28]. At this stage of their assembly, the complexes appear highly mobile in the ER membrane and rapidly concentrate at the ER exit sites, a process favored by the existence of a di-acidic export sequence located at the N-terminal domain. In the absence of this signal sequence, Cav-1 accumulates in lipid droplets [29,30]. This is an important observation, suggesting that ER exit and lipid droplets localization of caveolin complexes are competing processes, highlighting the role of the di-acidic motif in caveolin trafficking.

Interestingly, co-expression of Cav-1 and -2 results in assembly of mixt 8S complexes, where the Cav-1 to Cav-2 ratio may vary from 2:1 to 4:1 [31]; however, expression of Cav-2 alone does not result in oligomer formation.

The process continues in the trans-Golgi where the oligomers are exported in a COPII-dependent manner and self-associate into a large network of caveolin. However, formation of the 8S complex is not a prerequisite for Golgi transport, as expression of Cav- 2 alone, as well as that of an oligomerization-incompetent variant of Cav-1 does not result in their retention within the ER [32,33].

The Golgi oligomerization step is sensitive to BFA, clearly indicating that formation of large oligomer complexes is dependent on caveolins trafficking to this compartment, probably requiring a specific lipid composition of the membrane. The assembly process continues in a cholesterol-dependent manner, resulting in formation of 70S stable complexes, also evidenced by using a panel of anti-Cav-1 conformational antibodies. These complexes were assumed to correspond to the intact protein scaffold of the caveolae structure [34]. It is interesting that the caveolin assemblies colocalized with medial, rather than trans-Golgi markers, suggesting that, unlike other cargos transported through the secretory pathway, caveolar carrier vesicles are formed in the medial cisternae, being further exported to the plasma membrane in a dynamin-2 independent manner, similar to other raft-associated proteins [34]. This assembly process is schematically shown in Fig.2.

Figure 2.

Caveolin oligomerization and assembly

It can be concluded from these observations that the tight regulation of Cav-1 trafficking along the secretory compartments (and the multiple check points) is totally justified by the complexity of the assembly process.

In addition to oligomerization, caveola formation involves association of the complexes with cholesterol-rich lipid-raft domains at plasma membrane. It is estimated that Cav-1 binds to 1–2 cholesterol molecules through the conserved basic and hydrophobic residues of the scaffolding domain [35]; thus, the relative amount of cholesterol concentrated in isolated caveolae can be as high as 20.000 molecules [36]. The relationship between cholesterol and caveolins is very complex. Treatment of cells with cholesterol binding or depleting agents results in caveolae with altered morphology and disrupted protein coat [7]. Moreover, cholesterol regulates Cav-1 expression at both, transcriptional and translational levels, through binding to either two steroid regulatory elements in the Cav-1 promoter, or the protein itself, thus modulating the level of Cav-1 mRNA or the protein stability [37,38].

Caveolae are enriched in glycosphingolipids (like GM1 and GM3) and sphingomyelin, the total lipid density being significantly higher than within the rest of the plasma membrane [36]. This is an important observation implying that certain lipids are recruited in caveolae, possibly to ensure their invagination-competent composition.

Recently, a crucial role in the last steps of caveolae biogenesis has been attributed to PTRF (Polymerase I and transcript release factor), originally described as an RNA Pol I transcription factor (also called Cav-P60 or cavin-1) [39,40]. Interestingly, cavin-1 is able to associate with plasma membrane caveolae but not with caveolins with other intracellular distribution (such as Golgi caveolins) [34]. Cavin-1 is recruited by Cav-1 to plasma membrane caveolar domains, where the two proteins are found to be in close proximity and an approximate ration of 1:1 [39]; however, whether or not they directly interact with each other is still a matter of debate. It was clearly demonstrated that cavins bind to phosphatidylserine and are phosphorylated at multiple sites, suggesting they may act as regulatory proteins of caveolae functions [41].

Based on sequence homology with cavin-1, three other proteins named cavin-2 to 4, sharing similar molecular organization, have been identified as part of the cavin family [42-44]. While cavin-1 expression is strictly associated to that of Cav-1 [39] contributing to the stability of the caveolae unit like a scaffolding protein, cavin-2 promotes recruitment of cavin-1 in caveolae and appears to have a role in the membrane-curvature [45].

The role of cavin-3 and -4 in caveolae biogenesis is less well understood. Cavin-3 was shown to regulate caveolae budding and Cav-1 trafficking, suggesting a function in coupling caveolae to the intracellular transport network [44]. Cavin-4 is co-expressed with Cav-3 in cardiac and muscle tissues where their function appears to be tightly correlated [42].

All members of the cavin family interact in a multimeric complex of about 60-80 cavins, in a Cav-1 independent manner. These complexes were detected both in the cytosol and plasma membrane fractions, suggesting they are the result of a succession of events, starting with cavin association into the cytosol and ending with the recruitment of the multimeric complexes to caveolae, during the final step of their biogenesis [42].

In contrast to caveolins, cavins are peripheral membrane proteins, and bind molecular components of the caveolar domain facing the cytosol. Given the high affinity of cavins for phosphatidylserine and the rapid dissociation from caveolae in the presence of nonionic detergents, it was suggested that binding to the lipid membrane, rather than to the protein scaffold, was highly probable.

The identification of cavins in caveolae opened new perspectives in understanding the complexity of caveolar structure. Although our knowledge on caveolae architecture and molecular composition has improved since their discovery, the main structural pillars defined at the time have not dramatically changed. Thus, today, caveolae are referred to as invaginations of the plasma membrane lipid bilayer, enriched in cholesterol and sphingolipids, embedding an integral membrane scaffold formed by caveolin oligomers assembled in a stable network, peripherally covered by a protein layer of cavin complexes. Once formed, this structure appears to remain stable also during endocytosis [46].

Advertisement

2. Caveolae signaling

The protein composition of caveolae has been addressed in a more systematic manner within the last years, using proteomic approaches [47-49]. A variety of signal transduction proteins were found to localize in caveolae, in tight connection with either the CSD or the lipid domains.

According to the caveolin signaling hypothesis, the role of caveolae is to trigger specific signal transduction by concentrating downstream effectors close to plasma membrane receptors, through direct interaction with the CSD [50,51]. Palmitoylation appears to play an important function in this process by facilitating the caveolar localization of proteins [52]. Of the signaling molecules identified, several have been more thoroughly investigated:

  1. G proteins were abundant in caveolae; their binding to caveolin has a role in maintaining the Gα subunits in an inactive GDP-bound state [53]. Small GTP-binding proteins of the Ras superfamily also localized in caveolae, a process enhanced by palmitoylation of the C-terminal hypervariable region [54,55]. Binding of the H-Ras to the CSD results in inactivation of the protein, which is relevant in certain human cancers were H-Ras -caveolin interaction is prevented and the protein is maintained in an active state [56].

  2. Src family kinases, such as c-Src, Fyn, Lyn [57] are nonreceptor tyrosine kinases, also enriched in caveolae. Their localization depends on the N-terminal myristoylation and subsequent interaction with the caveolins. Interestingly, Cav-1 palmitoylation is equally important for caveolae/c-Src interaction [52], which results in c-Src and Fyn inactivation [58]. In turn, the tyrosine phosphorylation of Cav-1 and -2 facilitates the recruitment of matrix metalloproteinases [58,59], and promotes caveolins localization to focal adhesions [60,61].

  3. Several steroid hormone receptors were localized in caveolae, a process depending on both, palmitoylation and association with Cav-1 [62] and facilitating their activation [63].

  4. Endothelial nitric oxide synthase (eNOS) is one of the most extensively studied Cav-1 interacting protein [64]. eNOS binds to the CSD of both Cav-1 and -3, which inhibit its enzymatic activity [65,66]. This observation lead to a novel concept of eNOS regulation, whereby, the interaction with caveolins is necessary to keep the enzyme inactive under basal conditions, while its concentration in caveolae will allow a quick response upon stimulation [67].

  5. Many ion channels and pumps are targeted to caveolae and interact with caveolins, such as the calcium signaling molecules calmodulin, Ca2+-ATPase, L-type Ca2+ channels [68,69]. Transient receptor potential (TRP) channels, and large-conductance Ca2+-activated K+ channels also localize in cholesterol-rich membrane areas, suggesting an important role of these domains in Ca2+ homeostasis [70,71]. Other transporters, like the Na/K-ATPase, involved in maintaining the Na+ membrane gradient, are also found in caveolae, owing this localization to the existence of two caveolin-binding motifs in their amino acid sequence [72].

  6. Protein kinases of different families were found in caveolae, due to their direct interaction with the Cav-1 CSD. For PKA, this interaction results in inhibition of the enzymatic activity [73], with consequences on regulation of other proteins, such as ATP-dependent K+ channels or eNOS in muscle and endothelial cells, respectively [74,75]. Different isoforms of the PKC family of enzymes are also caveolae resident and appear to participate in regulation of caveolar proteins [76]. Caveolae interaction with PKC is more complex, leading to either activation (via ceramide interaction) [77], or inactivation, following endosomal delivery [78]. Caveolae also recruit the phosphatidylinositol-3-kinase (PI3K) through direct binding to Cav-1 [79] and the protein kinase B (PKB). Integration of this signaling pathway by caveolae plays a significant role in managing the cellular physiological stress, and regulating cell survival and death [80].

  7. Phosphodiesterases (PDEs), involved in cyclic nucleotides (cAMP and cGMP) hydrolysis, have a preference for lipid rafts association (Abrahamsen H, 2004). For some isoformes, such as PDE3B, a direct interaction with Cav-1, with a stabilizing effect on PDE3B, has been clearly confirmed by co-immunoprecipitation [81]. Other PDEs (PDE5, PDE4A4) appear to be recruited by caveoale through indirect mechanisms, possibly involving adapter proteins; [82]. Nevertheless, this association influences the establishment of cAMP/cGMP gradients and the downstream events [83].

Advertisement

3. Caveolae internalization and trafficking

Electron microscopy data show that caveolae are tightly connected to submembranous actin filaments [84,85], suggesting a function of the cytoskeleton in caveolae-mediated endocytosis. However, the exact role of the actin cytoskeleton is not clearly defined, as its disruption inhibits uptake of the caveolae ligand, alkaline phosphatase, on one hand [86] and promotes clustering of caveolae and internalization of Cav-1-labeled vesicles, on the other hand [6,87]. These observations fit into a model whereby actin would play a dual role in caveolae internalization: one is to keep the organization of caveolae and maintain their immobility at the plasma membrane, and the other to promote vesicle budding and release from the membrane.

Caveolae endocytosis relies heavily on dynamin, a multi-domain GTPase [88,89], which was shown to interact directly with Cav-1 [90]. Ligand binding initially disrupts the local actin cytoskeleton and promotes dynamin II recruitment to the site of internalization [91,92]. Dynamin oligomerization and the GTP-dependent conformational changes result in a structural collar around the neck of caveolae, directly mediating formation of free transport vesicles, following vesicular fission from the plasma membrane. It was shown that the protein regulates the actin tail formation [93,94], possibly through binding to cortactin [95-97] or intersectin, which promotes actin polymerization [98].

Another player in this complex molecular game was recently suggested, following the initial observation that Cav-1 binds to actin cross-linking proteins, filamin A and B, both in vitro and in vivo [99]. The main intracellular function of these proteins is to organize the actin cytoskeleton. The Cav-1 filamin A interaction was further confirmed in different cell types [100,101] and it was implicated in activation of the actin-folding and chaperone protein T-complex protein-1, [100] and inhibition of calpain-mediated cleavage of filamin A [102]. Thus, by providing the missing link between Cav-1 and the actin cytoskeleton, filamin is an important regulator of caveolae-mediated endocytosis and trafficking [103].

It was proposed that following budding, caveolae can fuse with either preformed vesicles, called “caveosome” at the time of their discovery, or early endosome, the latter process being dependent on Rab5 expression [46].

The caveosome was initially described as an immobile structure which did not co-localize with fluid phase markers or ligands of the clathrin-dependent pathway [104]. Moreover, the compartment was characterized by neutral pH and was unable to accumulate a lysosomal dye (lysotracker), reinforcing the notion of an independent organelle, clearly distinct from other endocytic compartments, which delivers its cargo to other cellular locations, such as the ER [104,105]. However, in a recent investigation of Cav-1 trafficking using pH-sensitive fluorophores, the existence of a neutral pH compartment was seriously doubted [106]. Rather, it was suggested that the caveosomes would correspond to modified late endosomes, where Cav-1 accumulates when over-expressed, undergoing ubiquitination and being further targeted to degradation [106]. Conversely, under physiological conditions, caveolae would bud from the plasma membrane transporting their viral cargo to early endosomes and eventually to the ER, in a microtubule-dependent manner [107], following a series of maturation events, which will be detailed below.

Clearly, more work is necessary to have the correct picture of the highly atypical caveolar trafficking, which appears to allow access of its ligands to intracellular destinations that are not reachable from other endocytic pathways. Despite the remaining uncertainties, the continuous development of the field has considerable advanced our knowledge of virus infection of host cells using caveolae endocytosis.

The current view of caveolae internalization and trafficking is depicted in Fig. 3.

Figure 3.

Ligand internalization and intracellular trafficking following caveolae-mediated endocytosis

Advertisement

4. Caveolae dependent viral infections

Owing to the vast amount of experimental data characterizing cell infection by the simian virus 40 (SV40), the pathogen has become the “star” of the caveolae-mediated entry pathway, being now extensively used in other studies as a marker of it [104,108]. Other well-characterized viruses using this entry pathway belong to the polyoma virus family, which has gained more interest recently, with the increasing number of human viruses identified. These include the KI polyoma virus, the WU polyoma virus and the Merkel cell polyoma virus [109-111], the latter being associated with an aggressive form of neuroendocrine skin cancer, the Merkel cell carcinoma. These are all non enveloped DNA viruses that replicate in the nucleus.

Viruses that use the same pathway to initiate a productive infection in target cells are listed in Table 1. Amongst them, Echovirus 1 (EV1), Human Hepatitis B virus (HBV) [112], Murine Leukemia Virus (MLV) [113], enteroviruses [114], have been more intensely investigated.

VirusReference
Simian virus 40Stang, E., J. Kartenbeck, and R.G. Parton. 1997. Major histocompatibility complex class I molecules mediate association of SV40 with caveolae. Mol Biol Cell. 8:47-57.
Pelkmans, L., J. Kartenbeck, and A. Helenius. 2001. Caveolar endocytosis of simian virus 40 reveals a new two-step vesicular-transport pathway to the ER. Nat Cell Biol. 3:473-83.
KI polyoma virusAllander, T., K. Andreasson, S. Gupta, A. Bjerkner, G. Bogdanovic, M.A. Persson, T. Dalianis, T. Ramqvist, and B. Andersson. 2007. Identification of a third human polyomavirus. J Virol. 81:4130-6
WU polyoma virusGaynor, A.M., M.D. Nissen, D.M. Whiley, I.M. Mackay, S.B. Lambert, G. Wu, D.C. Brennan, G.A. Storch, T.P. Sloots, and D. Wang. 2007. Identification of a novel polyomavirus from patients with acute respiratory tract infections. PLoS Pathog. 3:e64
Merkel cell polyoma virusFeng, H., M. Shuda, Y. Chang, and P.S. Moore. 2008. Clonal integration of a polyomavirus in human Merkel cell carcinoma. Science. 319:1096-100
Echovirus 1Marjomaki, V., V. Pietiainen, H. Matilainen, P. Upla, J. Ivaska, L. Nissinen, H. Reunanen, P. Huttunen, T. Hyypia, and J. Heino. 2002. Internalization of echovirus 1 in caveolae. J Virol. 76:1856-65.
Stuart, A.D., H.E. Eustace, T.A. McKee, and T.D. Brown. 2002. A novel cell entry pathway for a DAF-using human enterovirus is dependent on lipid rafts. J Virol. 76:9307-22
Human Hepatitis B virusMacovei, A., C. Radulescu, C. Lazar, S. Petrescu, D. Durantel, R.A. Dwek, N. Zitzmann, and N.B. Nichita. 2010. Hepatitis B virus requires intact caveolin-1 function for productive infection in HepaRG cells. J Virol. 84:243-53.
Murine Leukemia VirusBeer, C., D.S. Andersen, A. Rojek, and L. Pedersen. 2005. Caveola-dependent endocytic entry of amphotropic murine leukemia virus. J Virol. 79:10776-87
Tiger Frog VirusGuo, C.J., D. Liu, Y.Y. Wu, X.B. Yang, L.S. Yang, S. Mi, Y.X. Huang, Y.W. Luo, K.T. Jia, Z.Y. Liu, W.J. Chen, S.P. Weng, X.Q. Yu, and J.G. He. 2011. Entry of tiger frog virus (an Iridovirus) into HepG2 cells via a pH-dependent, atypical, caveola-mediated endocytosis pathway. J Virol. 85:6416-26
Infectious spleen and kidney necrosis virusGuo, C.J., Y.Y. Wu, L.S. Yang, X.B. Yang, J. He, S. Mi, K.T. Jia, S.P. Weng, X.Q. Yu, and J.G. He. 2011. Infectious spleen and kidney necrosis virus (a fish iridovirus) enters Mandarin fish fry cells via caveola-dependent endocytosis. J Virol. 86:2621-31.
Influenza virusesNunes-Correia, I., A. Eulalio, S. Nir, and M.C. Pedroso de Lima. 2004. Caveolae as an additional route for influenza virus endocytosis in MDCK cells. Cell Mol Biol Lett. 9:47-60.
Coronavirus 229ENomura, R., A. Kiyota, E. Suzaki, K. Kataoka, Y. Ohe, K. Miyamoto, T. Senda, and T. Fujimoto. 2004. Human coronavirus 229E binds to CD13 in rafts and enters the cell through caveolae. J Virol. 78:8701-8
Papillomavirus 31Bousarghin, L., A. Touze, P.Y. Sizaret, and P. Coursaget. 2003. Human papillomavirus types 16, 31, and 58 use different endocytosis pathways to enter cells. J Virol. 77:3846-50
Respiratory syncytial virusBrown G, Jeffree CE, McDonald T, Rixon HW, Aitken JD, et al. Analysis of the interaction between respiratory syncytial virus and lipid-rafts in Hep2 cells during infection. Virology 2004;327(2) 175-185.
Werling D, Hope JC, Chaplin P, Collins RA, Taylor G, et al. Involvement of caveolae in the uptake of respiratory syncytial virus antigen by dendritic cells. J Leukoc Biol 1999;66(1) 50-58.
Newcastle disease virusCantin, C., J. Holguera, L. Ferreira, E. Villar, and I. Munoz-Barroso. 2007. Newcastle disease virus may enter cells by caveolae-mediated endocytosis. J Gen Virol. 88:559-69
Ebola virusEmpig, C.J., and M.A. Goldsmith. 2002. Association of the caveola vesicular system with cellular entry by filoviruses. J Virol. 76:5266-70.
BK polyoma virusEash, S., W. Querbes, and W.J. Atwood. 2004. Infection of vero cells by BK virus is dependent on caveolae. J Virol. 78:11583-90.
Marburg virusEmpig, C.J., and M.A. Goldsmith. 2002. Association of the caveola vesicular system with cellular entry by filoviruses. J Virol. 76:5266-70.

Table 1.

Viruses that use caveolae-mediated endocytosis

After binding to the host cell, the virus particles are able to cluster the receptor molecules such as certain integrins (a2h1 in the case of EV1) [118] or glycosphingolipids (GM1 or GD1a in the case of SV40 or polyoma viruses), within the lipid rafts [119]. Accumulation of viral particles in caveolae induces a cascade of tyrosine phosphorylation reactions followed by rearrangements of the cortical actin cytoskeleton, as described above [91].

Several models addressing virus capturing into the plasma membrane invaginations have been proposed, the most recent relying on the observation that Cav-1 polymer remains intact once formed in the Golgi complex, during transport to the plasma membrane. According to this model, caveolae result from the fusion of a pre-existing caveolar vesicle with the plasma membrane [46]. As a consequence of virus binding to an increasing number of sphingolipids and/or integrins, the affinity of the pathogen for caveolar domains increases, facilitating entrapment in these areas. Another possibility is that virus particles bind and release the plasma membrane gangliosides in a transient manner, thus screening the whole cell surface. When reaching a caveolar region where multiple gangliosides interactions can occur simultaneously, binding becomes permanent and the virus particles are sequestered [104].

The intracellular trafficking of caveolae cargos has been recently re-evaluated using SV40 as a model and a series of complementary, state-of-the art techniques, including live-cells and electron microscopy, video recordings, pharmacological inhibitors and inhibition of expression of trafficking regulators [120]. It was shown that productive SV40 infection depends on the virus transport through a series of classical endocytic vesicles. Initially, the virus is found in Rab5-, EEA1-positive early endosomes and subsequently becomes associated with Rab7-positive domains, during endosome maturation. As this process proceeds, SV40 co-localizes with LAMP1-, Rab9-, and Rab7-positive late endosomes resembling multivesicular bodies and possibly endolysosomes. Endosome maturation also involves acidification of the compartments, as a consequence of vacuolar ATPase (v-ATPase) recruitment and activity. At this stage, acidification is required for SV40 subsequent transport steps and the initiation of productive infection. Interestingly, BK and JC viruses were also shown to enter the endosomes and depend on acidification for infection [121-123]. In the case of the mouse polyoma virus, the recycling, as well as late endosomes have been involved in infection [124,125].

From the late compartments of the endocytic pathway, SV40 appears to be directly transported to the ER, although an indirect ER targeting, via the Golgi complex, has not been completely excluded. Similarly, other polyoma viruses are transported to the ER lumen, before reaching the nucleus [121,125].

However, there are some notable exceptions of virus trafficking diverting from this pathway, despite being internalized through caveolae. Thus, cellular penetration of the EV1, a positive-stranded RNA human pathogen, depends on caveolins, dynamin II, and signaling events but does not require actin filaments or microtubules. The virus uptake was much faster than that of SV40 and was followed by rapid co-localization with Cav-1. Beyond this step, the virus failed to enter the Golgi complex, the ER, or the lysosomes, as none of the markers used to label these organelles co-localized with viral proteins. This observation raised the hypothesis that the virus particles remain sequestered in the Cav-1 positive endocytic vesicles until replication is initiated, further using them for cytoplasmic penetration and uncoating.

The lack of transport to the ER or Golgi was also confirmed by the absence of any inhibition of infection in the presence of nocodazole. Although more experimental data is needed to substantiate it, it is tempting to speculate that the different sorting pathway of the two viruses is related to their replication mechanism; thus, a DNA virus such as SV40 needs the nucleus for replication, which might be more accessible through the ER, whereas for a positive-stranded RNA virus the release of the genome into the cytoplasm is sufficient to initiate replication.

Either way, the precise molecular details characterizing this segment of the trafficking pathway are still to be defined, but understanding the factors involved in these unusual trafficking pathways is crucially important as other, yet uninvestigated viruses, may well use them when accessing the host cell via caveolae.

Importantly, there is accumulating evidence suggesting that several viruses take advantage of cross talk between endocytosis routes. For instance, JCV, bovine papillomavirus type 1 and human papillomavirus type 16 have been shown to access cells using the clathrin-dependent endocytosis, but intriguingly, they require Cav-1-mediated trafficking to initiate productive infection [123,126,127]. The internalized virions were trafficked to early endosomes before being transported to the caveolar pathway. From the Cav-1-positive vesicles, the viral cargo is further moved to the ER in a COPI-mediated, BFA-sensitive manner [127].

The newly described trafficking routes taken by these viruses may have an explanation in their absolute requirement to reach the ER compartment, a target that is not on the clathrin-mediated route.

The intriguing question as to why these viruses travel to the lumen of the ER, instead of using the endosomes for genome release, have received several interesting answers lately. One possibility is that the pathogens take advantage of the ER machinery of folding enzymes and chaperones, for uncoating and membrane penetration, being activated by lumenal thiol oxidoreductases before release into the cytosol/nucleoplasm [128].

Very recently, BiP and the ER-membrane protein BAP31 (both involved in ERAD) were shown to be essential factors for SV40 infection; thus incoming SV40 particles are structurally remodeled leading to exposure of the amino-terminal sequence of the minor viral protein VP2. These hydrophobic sequences anchor the virus to the ER membranes helping the particles release into the cytosol [129].

Advertisement

5. Investigation of the caveolae entry pathway

The molecular details of virus entry have been investigated through a variety of techniques, by perturbing endocytotic internalization with various inhibitors or interfering with the expression or function of key regulator proteins, using siRNA or dominant-negative mutants of the proteins, or by using transgenic animals. Because no single method to assess caveolae is perfect, the use of complementary techniques is crucial for such a task. These can employ cell fractionation, immunoprecipitation, protein and organelle labeling, immunofluorescence microscopy. Since caveolae are best characterized by their microscopic appearance, studies employing alteration of the intracellular level of caveolins, cholesterol, or different molecules enriched, but not exclusively present in caveolae, should ideally follow the impact of such changes to other internalization and trafficking pathways, by electron microscopy

5.1. Pharmacological approaches

A pharmacologic approach involves the treatment of cells with agents that deplete membranes of cholesterol, or inhibit various structural or signaling molecules involved in controlling the pathway. Having the advantage of being readily available and convenient to use, chemical inhibitors have been extensively employed to characterize different endocytic pathway; however, stringent controls must be included and results should be interpreted with care, because of the pleiotropic effects these drugs may have within the treated cell.

The usefulness as well as the pitfalls associated with the use of these agents will be detailed below.

Methyl-β-cyclodextrin (MβCD) – Cyclodextrins are cyclic oligomers of glucose that have the property to bind and extract lipophiles, including cholesterol, from their hydrophobic core [130]. Based on the tight dependency of caveolae stability and function on the amount of cholesterol present in the lipid rafts, the compound has been widely used to define the caveolar-mediated entry of many pathogens [6,117,131-135].

A major issue of MβCD treatment is its cellular toxicity, which was initially associated with longer incubation times (for example, the cell viability decreases form 90% during a 30 minutes incubation, to as low as 64%, if the drug is used for 12 h) [117]. A thorough study performed on multiple cell lines showed, however, that MβCD significantly decreased cellular viability, even after short treatment and at concentrations routinely used to inhibit endocytosis, a phenomenon which was cell line dependent [136]. Moreover, a low level of plasma membrane cholesterol was shown to interfere with other endocytic pathways, such as the clathrin-mediated endocytosis [137,138], or even with cholesterol independent endocytosis [139], demonstrating a rather poor specificity of the drug in inhibiting a distinct pathway.

Statins – are a group of drugs that lower the intracellular cholesterol level by competitively inhibiting the 3 hydroxy 3 methylglutaryl coenzyme A reductase involved in its biosynthesis. As a consequence, efficient depletion of membrane cholesterol and decreased formation of caveolae are observed [140]. Despite showing good toxicity profiles, statins also exert pleiotropic effects through a variety of mechanisms, which appear to be unrelated to their cholesterol-lowering activity. Thus, several immunosuppressive effects have been involved; amongst them well-documented are the prevention of activation of the transcription factor NF-kappaB or up-regulation of the pro-inflammatory cytokine production [141,142].

Filipin - is a macrolide pentene polyene with antibiotic properties relying on sterol binding with high affinity [143]. Filipin III has been employed to block caveolae entry since it complexes with membrane cholesterol, thus interfering with cholesterol-sensitive processes [144]. Treatment with filipin dispersed the receptors found in caveolae and promoted disassembly of these structures [145]. Similar to MβCD, filipin treatment is toxic and at least for certain cell lines, its inhibitory effect on endocytosis was exclusively due to cytotoxicity [146]. However, a narrow window of specific inhibitory function can be identified in most cell lines.

Genistein – inhibits several tyrosine kinases in mammalian cells and thus, caveolae internalization [86,147]. Genistein has been however shown to suppress the entry of several types of viruses that use different endocytic ways to gain access to the replication sites: SV40 [91], adenoviruses [148], human herpesvirus 8 (HHV-8) [149], HBV [112]. However, the general need of tyrosine kinases-mediated signaling of diverse families of viruses, both at early entry steps, or later in infection, makes it difficult to clearly ascertain the endocytic route used, only by using genistein.

U18666A - is an amphiphilic amine that arrests cholesterol transport and suppresses sterol biosynthesis. Treatment of cells with this inhibitor was shown to induce cholesterol accumulation in late endosomes/lysosomes and deplete cholesterol from the Golgi complex [150]. Interestingly, the mobility of Cav-1 significantly increased in the Golgi complex of U18666A-treated cells.

Phorbol 12-myristate 13-acetate (PMA) – is an activator of classical 2,3-diacylglycerol (DAG)-dependent protein kinase isoforms, owing this property to their high affinity for the DAG binding site. Interestingly, PMA treatment results in constitutive phosphorylation of caveolin [151] with significant inhibitory effects on caveola invagination from the intracellular face of the plasma membrane [86,152,153].

PMA has been shown to specifically inhibit the caveolae-mediated entry of Ebola and Marburg viruses, two negative-stranded RNA pathogens, members of the Filoviridae family [154]. PMA has a low citotoxicity even when used for longer incubation times (82% of the cell are still viable following incubation for 24 h, at concentrations required to inhibit caveolae endocytosis [117].

It is important to note that PMA can stimulate endocytosis of other ligands and interferes with the endocytic trafficking by stimulating a factor required for endosome fusion after Rab5 activation [155]. PMA treatment may have opposite effects on internalization of certain ligands (such as FITC-dextran) in polarized cells, increasing for instance, its basolateral, but not apical uptake.

Okadaic acid – is an inhibitor of phosphatases 1 and 2A, which are important in caveolae function [156]. Treatment with okadaic acid has been shown to promote removal of caveolar structures from the cell surface and stimulate endocytosis via these structures [6,86]. Importantly, the drug also inhibits the clathrin-mediated endocytosis [86] making the time of addition to the cells an important experimental factor in drawing the right conclusions on viral entry. Thus, pretreatment of cells with okadaic acid interferes with both, caveolae and clathrin pathways, while addition after virus binding to the target cells enhances infection, as it was shown for MLV [113].

Cytoskeleton inhibitors - the actin cytoskeleton localized near the plasma membrane appear to be a critical regulator of caveolae endocytosis [157].

Depolymerization of microtubules with colchicine or disruption of actin microfilaments with cytochalasin D resulted in a significant reduction of the amount of Cav-3 in plasma membrane fractions isolated from cardiac myocytes. Treatment with either drug also led to the exclusion of Cav-1 and -2 from similar fractions and the decrease of tyrosine-phosphorylated Cav-1 [101].

Other drugs interfering with actin polymerization are latrunculin A (actin monomer-sequestering drug) and jasplakinolide (an actin polymer-stabilizing compound), both were shown to reduced SV40 internalization by more than 60% [91]. Since function of many cellular processes such as trafficking and organelle movement are regulated by microtubules and the actin cytoskeleton, the biochemical assays should be combined with microscopy analysis to clearly define the role of the cytoskeleton in viral infection mediated by caveolae. Also, it is important to keep in mind that viruses induce cytoskeletal reorganization and reconfiguration to initiate, maintain and spread the infection. Therefore, the impact of the cytoskeleton perturbation on the outcome of infection highly depends on the stage of the viral life cycle the drug is acting upon [158].

5.2. Interference with expression and function of caveolae regulating proteins

As threshold levels of Cav-1 regulate caveolae formation [155,159,160], modulating the expression and/or function of this protein is one the most reliable approach to investigate caveolae entry. Cav-1 down-regulation using anti-sense, small interfering (si) or short hairpin (sh) RNA results in a significant decrease in the number of caveolae. For instance, the siRNA-mediated knockdown of Cav-1 expression was sufficient to inhibit albumin uptake in endothelial cells; however, intriguingly enough, the caveolae localization of signaling proteins, including eNOS, Rac, tyrosine kinase Src and insulin receptor was not altered. Using this technique, several viruses were shown to depend on caveolae for productive infection such as the Avian Reovirus [161], BK polyomavirus [133].

Cavin proteins are also important targets to study caveolae-mediated entry, since absence of cavin-1 results in lower expression level of caveolins and eventually, the loss of caveole. Down-regulation of cavin-1, using specific shRNA, increases mobility of caveolin-1, which is released from the cell surface and rapidly internalized and degraded [39]. Interfering with cavin-2 expression is also a valuable tool when assessing the role of caveolae in viral infection, since its down-regulation induces loss of cavin-1 and caveolin expression and therefore, it limits caveolae formation [43]. Similarly, suppression of cavin-3 biosynthesis uncouples caveolae from the intracellular transport machinery [44].

Caveolae budding from the plasma membrane and subsequent internalization strictly depends on dynamin II [89], thus silencing its expression is also often used in combination with caveolin inhibition.

An elegant alternative to silencing the expression of the proteins involved in caveolae architecture and function is over-expression of their mutant counterparts, which compete with the wild-type proteins for the same function. An important advantage of this technique is that, at any time during the experiment, the wild-type protein is still expressed ensuring the functioning of the pathway at a basal level and reducing toxicity. This approach was used to show the dependence of HBV internalization on functional Cav-1 and dynamin II (Fig. 4) [112].

Figure 4.

Hepatitis B Virus (HBV) infection of permissive HepaRG cells stably expressing dynamin II (A and B) and caveolin-1 (C and D) proteins with either wild-type (A and C) or dominant-negative (B and D) functions. HBV infection is evidenced by immunofluorescence microscopy using antibodies against the envelope proteins (in red). The dominant-negative dynamin II contains the K44A mutation which abolishes the GTP-ase activity (B). The dominant negative caveolin-1 contains a deletion of the 1-81 amino acid domain, at the N-terminal end (D). Expression of the wild-type and dominant-negative variants is evidenced through the Green Fluorescent Protein (GFP), which is either co-expressed from bicistronic GFP-caveolin DNA constructs (C and D) or expressed in fusion with dynamin II (A and B).

Cav -1 dominant negative proteins – disturb the formation of Cav-1-positive lipid rafts and cause the redistribution of endogenous caveolin to detergent soluble membrane fractions [162]. These are N-terminally truncated or N-terminally GFP-tagged caveolin constructs, which strongly inhibit SV40 entry [104,163] and were used to characterize the internalization pathway of many other viruses [134,164,165].

Other caveolin constructs containing the point mutations, Y14F and P132L were recently demonstrated to have dominant-negative activity [166]. Expression of the Cav-1 variant containing the P110A mutation was shown to determine a profound inhibition of caveolae endocytosis, cellular lipid accumulation and lipid droplet biogenesis. Moreover, this is a potent mutant to take into account when investigating the caveolae pathway, as it significantly reduces the Cav-1 localization into detergent-resistant domains of the plasma membrane and caveolae formation [22].

An interesting caveolin mutant is cavDGV, a deleted Cav-3 form, which lacks the first 53 residues of the protein, but contains an intact scaffolding domain. The truncated protein acts as a dominant negative inhibitory mutant, causing the intracellular accumulation of free cholesterol in late endosomes, a reduction of surface cholesterol, efflux and synthesis [30].

Dynamin -II dominant negative proteins - as important regulators of clathrin, caveolae and other endocytic pathaways [88,167], dynamin II inhibition is often used in combination with modulation of other, more specific proteins involved in caveolae function (listed above). By far, the most used mutant dynamin is the K44A variant, defective in GTP hydrolysis, which was clearly shown to inhibit release of caveolae from plasma membranes in an in vitro assay [89].

A long term expression of a dominant negative protein may be toxic for the cells, determining changes of morphology. Also it is important to keep in mind that down-regulation of a certain pathway may promote up-regulation of other, compensatory entry mechanisms, if cells express dominant negative proteins for a long time. A solution to overcome these potential problems is the use controlled/inducible expression systems (such as the TetOn/Tet Off switch).

Generation of knockout (KO) mice – is a powerful approach for the study of caveolae in vivo. Caveolin-KO mice (Cav-1, -2, -3 and Cav-1/-3 double KO mice have already been generated and characterized. They displayed different phenotypes, but interestingly, were viable and fertile [168]. While Cav-2 KO mice retain normal expression of caveolae, Cav-1 KO mice are devoid of Cav-2 expression and caveolae in certain cell lines, and develop many cardiac and pulmonary diseases. More work is needed to understand whether or not these pathologies are directly correlated with the loss of expression of caveolins and caveolae and a good approach toward this aim would be to investigate each individual caveolins and the development of the corresponding phenotypes over a longer period of time.

Advertisement

6. Concluding remarks

The new experimental evidence emerged with the advance of the techniques used to investigate ligand internalization and intracellular trafficking, have consolidated the notion that endocytosis through caveolae is a true alternative to the clathrin-mediated pathway. By employing this route for entry into the target cell, viruses could benefit from the enormous advantage of being targeted directly to specific organelles that are essential for their replication; moreover, degradative compartments can be bypassed, which could enhance the efficiency of productive infection. Nevertheless, despite the tremendous development of the field in the last decade, many conceptual and mechanistic aspects are still to be clarified or reevaluated. Certainly, important issues regarding: a) the regulation of the crosstalk between different internalization pathways; b) the similarities between caveolae and other clathrin-independent entry routes; c) the exact mechanism of ligand sorting; d) the properties of the compartment(s) where it occurs; d) the preferential targeting of caveolae ligands to other intracellular compartments than the ER; are already under the scrutiny of many cell biologists and will find an answer in the near future.

Advertisement

Acknowledgement

This work was supported by the Romanian Academy project 3 of the Institute of Biochemistry, the IDEI grant ID-84 and the POSDRU/89/1.5/S/60746 grant.

References

  1. 1. Palade GE. Caveolae-Dependent Endocytosis in Viral Infection. J Histochem Cytochem 1 1953 4 188211 .
  2. 2. YamadaE. Caveolae-Dependent Endocytosis in Viral Infection. J Biophys Biochem Cytol 1 1955 6 551566 .
  3. 3. StanR. V.RobertsW. G.PredescuD.IhidaK.SaucanL.et al. Caveolae-Dependent Endocytosis in Viral Infection Mol Biol Cell 8 1997 4 595605 .
  4. 4. MontesanoR.RothJ.RobertA.OrciL. Caveolae-Dependent Endocytosis in Viral Infection Nature 296 1982 5858 651653 .
  5. 5. PartonR. G.WayM.ZorziN.StangE.Caveolinassociateswith.developingT-tubules.duringmuscle.differentiation J Cell Biol 136 1997 1 137154 .
  6. 6. ThomsenP.RoepstorffK.StahlhutM.van DeursB. Caveolae-Dependent Endocytosis in Viral Infection Mol Biol Cell 13 2002 1 238250 .
  7. 7. Rothberg KG, Heuser JE, Donzell WC, Ying YS, Glenney JR, et al. Caveolin, a protein component of caveolae membrane coats. Cell 68 1992 4 673682 .
  8. 8. TangZ.OkamotoT.BoontrakulpoontaweeP.KatadaT.OtsukaA. J.et al.Identificationsequence.expressionof.aninvertebrate.caveolingene.familyfrom.thenematode.Caenorhabditiselegans. Implications for the molecular evolution of mammalian caveolin genes. J Biol Chem 272 1997 4 24372445 .
  9. 9. SchererP. E.LewisR. Y.VolonteD.EngelmanJ. A.GalbiatiF.et al. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 1997;272 46 2933729346 .
  10. 10. SongK. S.SchererP. E.TangZ.OkamotoT.LiS.et al.Expressionof.caveolininskeletal.cardiacsmoothmuscle.cells Caveolin-3 is a component of the sarcolemma and co-fractionates with dystrophin and dystrophin-associated glycoproteins. J Biol Chem 271 1996 25 1516015165 .
  11. 11. TangZ.SchererP. E.OkamotoT.SongK.ChuC.et al. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 271 1996 4 22552261 .
  12. 12. WayM.PartonR. G.M-caveolina.muscle-specificcaveolin-related.proteinF. E. B. FEBS Lett 1995 108 EOF12 EOF .
  13. 13. LiS.SongK. S.LisantiM. P. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 271 1996 1 568573 .
  14. 14. FujimotoT.KogoH.NomuraR.UneT.Isoformsof.caveolincaveolarstructure. J.CellSci. 2000 Pt 193509 .
  15. 15. LahtinenU.HonshoM.PartonR. G.SimonsK.VerkadeP.Involvementof.caveolinincaveolar.biogenesisin. M. D. C. K.cellsF. E. B. FEBS Lett 2003;538(1-3) 85-88.
  16. 16. SowaG.PypaertM.FultonD.SessaW. C. Caveolae-Dependent Endocytosis in Viral Infection. Proc Natl Acad Sci U S A 2003;100 11 65116516 .
  17. 17. GalbiatiF.RazaniB.LisantiM. P.Caveolaecaveolininmuscular.dystrophy Trends Mol Med 7 2001 10 435441 .
  18. 18. DupreeP.PartonR. G.RaposoG.KurzchaliaT. V.SimonsK. Caveolae-Dependent Endocytosis in Viral Infection Embo J 12 1993 4 15971605 .
  19. 19. MonierS.PartonR. G.VogelF.BehlkeJ.HenskeA.etal. V. I.P21-caveolina.membraneprotein.constituentof.thecaveolar.coatoligomerizes.invivo.invitro. Mol Biol Cell 6 1995 7 911927 .
  20. 20. SchlegelA.SchwabR. B.SchererP. E.LisantiM. P. A.rolefor.thecaveolin.scaffoldingdomain.inmediating.themembrane.attachmentof.caveolin- The caveolin scaffolding domain is both necessary and sufficient for membrane binding in vitro. J Biol Chem 274 1999 32 2266022667 .
  21. 21. ArbuzovaA.WangL.WangJ.Hangyas-MihalyneG.MurrayD.et al. Caveolae-Dependent Endocytosis in Viral Infection Biochemistry 39 2000 33 1033010339 .
  22. 22. AokiS.ThomasA.DecaffmeyerM.BrasseurR.EpandR. M. Caveolae-Dependent Endocytosis in Viral Infection. J Biol Chem 285 2010 43 3337133380 .
  23. 23. LeeJ.GloverK. J. Caveolae-Dependent Endocytosis in Viral Infection. Biochim Biophys Acta 1818 2012 5 11581164 .
  24. 24. FernandezI.YingY.AlbanesiJ.AndersonR. G. Caveolae-Dependent Endocytosis in Viral Infection. Proc Natl Acad Sci U S A 99 2002 17 1119311198 .
  25. 25. SargiacomoM.SchererP. E.TangZ.KublerE.SongK. S.et al. Caveolae-Dependent Endocytosis in Viral Infection Proc Natl Acad Sci U S A 92 1995 20 94079411 .
  26. 26. RenX.OstermeyerA. G.RamcharanL. T.ZengY.LublinD. M.et al. Caveolae-Dependent Endocytosis in Viral Infection. Mol Biol Cell 15 2004 10 45564567 .
  27. 27. SongK. S.TangZ.LiS.LisantiM. P. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 272 1997 7 43984403 .
  28. 28. MonierS.DietzenD. J.HastingsW. R.LublinD. M.KurzchaliaT. V. Caveolae-Dependent Endocytosis in Viral Infection FEBS Lett 1996 143-149.
  29. 29. OstermeyerA. G.PaciJ. M.ZengY.LublinD. M.MunroS.et al. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 152 2001 5 10711078 .
  30. 30. PolA.LuetterforstR.LindsayM.HeinoS.IkonenE.etal. A.caveolindominant.negativemutant.associateswith.lipidbodies.inducesintracellular.cholesterolimbalance. J Cell Biol 152 2001 5 10571070 .
  31. 31. ScheiffeleP.VerkadeP.FraA. M.VirtaH.SimonsK.et al.Caveolin 2 in the exocytic pathway of MDCK cells. J Cell Biol 1998;140 4 795806 .
  32. 32. LeeH.DSParkWang. X. B.SchererP. E.SchwartzP. E.et al. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 2002 34556-34567.
  33. 33. MoraR.BonilhaV. L.MarmorsteinA.SchererP. E.BrownD.et al.Caveolinlocalizesto.thegolgi.complexbut.redistributesto.plasmamembrane.caveolaeraftswhen.co-expressedwith.caveolin- J Biol Chem 274 1999 36 2570825717 .
  34. 34. HayerA.StoeberM.BissigC.HeleniusA.Biogenesisof.caveolaestepwise.assemblyof.largecaveolin.cavincomplexes. Traffic 11 2010 3 361382 .
  35. 35. MurataM.PeranenJ.SchreinerR.WielandF.KurzchaliaT. V.etal. V. I. Caveolae-Dependent Endocytosis in Viral Infection. Proc Natl Acad Sci U S A 92 1995 22 1033910343 .
  36. 36. OrtegrenU.KarlssonM.BlazicN.BlomqvistM.NystromF. H.et al. Caveolae-Dependent Endocytosis in Viral Infection. Eur J Biochem 271 2004 10 20282036 .
  37. 37. FieldingC. J.BistA.FieldingP. E.Caveolinm. R. N. A.levelsare.up-regulatedby.freecholesterol.down-regulatedby.oxysterolsin.fibroblastmonolayers. Proc Natl Acad Sci U S A 94 1997 8 37533758 .
  38. 38. FrankP. G.MarcelY. L.MAConnellyLublin. D. M.FranklinV.et al.Stabilizationof.caveolinbycellular.cholesterolscavengerreceptor.classB.typeI. Biochemistry 41 2002 39 1193111940 .
  39. 39. MMHillBastiani. M.LuetterforstR.KirkhamM.KirkhamA.et-Cavinal. P. T. R. F.conserveda.cytoplasmicprotein.requiredfor.caveolaformation.function Cell 132 2008 1 113124 .
  40. 40. LiuL.BrownD.Mc KeeM.LebrasseurN. K.YangD.et al.Deletionof.CavinP. T. R. F.causesglobal.lossof.caveolaedyslipidemia.glucoseintolerance. Cell Metab 8 2008 4 310317 .
  41. 41. AboulaichN.VainonenJ. P.StralforsP.VenerA. V. Caveolae-Dependent Endocytosis in Viral Infection. Biochem J 2004 2) 237-248.
  42. 42. BastianiM.LiuL.MMHillJedrychowski. M. P.NixonS. J.etal. M. U. R. C.Cavincavinfamily.membersform.tissue-specificcaveolar.complexes Jedrychowski MP, Nixon SJ, et al. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 185 2009 7 12591273 .
  43. 43. HansenC. G.BrightN. A.HowardG.NicholsB. J. S. D. P. R.inducesmembrane.curvaturefunctionsin.theformation.ofcaveolae. Nat Cell Biol 11 2009 7 807814 .
  44. 44. Mc MahonK. A.ZajicekH.LiW. P.MJPeytonMinna.JDetal. S. R. B.C/cavinisa.caveolinadapter.proteinthat.regulatescaveolae.function Embo J 28 2009 8 10011015 .
  45. 45. ZimmerbergJ.MMKozlov Caveolae-Dependent Endocytosis in Viral Infection Nat Rev Mol Cell Biol 7 2006 1 919 .
  46. 46. PelkmansL.BurliT.ZerialM.HeleniusA. Caveolae-Dependent Endocytosis in Viral Infection Cell 118 2004 6 767780 .
  47. 47. BanfiC.BrioschiM.WaitR.BegumS.GianazzaE.et al. Caveolae-Dependent Endocytosis in Viral Infection Proteomics 6 2006 6 19761988 .
  48. 48. Mc MahonK. A.ZhuM.KwonS. W.LiuP.ZhaoY.et al. Caveolae-Dependent Endocytosis in Viral Infection Proteomics 6 2006 1 143152 .
  49. 49. DurrE.YuJ.KrasinskaK. M.CarverL. A.YatesJ. R.et al. Caveolae-Dependent Endocytosis in Viral Infection Nat Biotechnol 22 2004 8 985992 .
  50. 50. Ostrom RS, Insel PA. Caveolae-Dependent Endocytosis in Viral Infection Br J Pharmacol 143 2004 2 235245 .
  51. 51. CohenA. W.RazaniB.SchubertW.WilliamsT. M.WangX. B.et al.Roleof.caveolininthe.modulationof.lipolysislipiddroplet.formation Diabetes 53 2004 5 12611270 .
  52. 52. LeeH.WoodmanS. E.EngelmanJ. A.VolonteD.GalbiatiF.et al.Palmitoylationof.caveolinata.singlesite. .Cys-15controlsits.couplingto.the-Srcc.tyrosinekinase.targetingof.duallyacylated.molecules. G. P.I-linkedtransmembrane.orcytoplasmic.tocaveolae.effectively-Srcuncouples. c.caveolinR-14. T. Y. JA, Volonte D, Galbiati F, et al. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 276 2001 37 3515035158 .
  53. 53. Insel PA, Head BP, Ostrom RS, Patel HH, Swaney JS, et al. Caveolae-Dependent Endocytosis in Viral Infection Ann N Y Acad Sci 1047166 .
  54. 54. RoyS.PlowmanS.RotblatB.PriorI. A.MunckeC.et al. Caveolae-Dependent Endocytosis in Viral Infection. Mol Cell Biol 25 2005 15 67226733 .
  55. 55. PriorI. A.HardingA.YanJ.SluimerJ.PartonR. G.etal. G. T. Caveolae-Dependent Endocytosis in Viral Infection Nat Cell Biol 3 2001 4 368375 .
  56. 56. LiS.OkamotoT.ChunM.SargiacomoM.CasanovaJ. E.et al. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 270 1995 26 1569315701 .
  57. 57. SargiacomoM.SudolM.TangZ.LisantiM. P. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 122 1993 4 789807 .
  58. 58. LiS.CouetJ.LisantiM. P. Src tyrosine kinases, Galpha subunits, and H-Ras share a common membrane-anchored scaffolding protein, caveolin. Caveolin binding negatively regulates the auto-activation of Src tyrosine kinases. J Biol Chem 271 1996 46 2918229190 .
  59. 59. LeeH.VolonteD.GalbiatiF.IyengarP.LublinD. M.et al. Caveolae-Dependent Endocytosis in Viral Infection Mol Endocrinol 2000;14 11 17501775 .
  60. 60. SwaneyJ. S.PatelH. H.YokoyamaU.HeadB. P.RothD. M.et al. Focal adhesions in (myo)fibroblasts scaffold adenylyl cyclase with phosphorylated caveolin. J Biol Chem 281 2006 25 1717317179 .
  61. 61. Del Pozo MA, Schwartz MA. Rac, membrane heterogeneity, caveolin and regulation of growth by integrins. Trends Cell Biol 17 2007 5 246250 .
  62. 62. AcconciaF.BocediA.AscenziP.MarinoM. Does palmitoylation target estrogen receptors to plasma membrane caveolae? IUBMB Life 55 2003 1 3335 .
  63. 63. LiL.RenC. H.TahirS. A.RenC.ThompsonT. C.Caveolinmaintainsactivated.Aktin.prostatecancer.cellsthrough.scaffoldingdomain.bindingsite.interactionswith.inhibitionof.serine/threonineprotein.phosphatasesP. 1 1 and PP2A. Mol Cell Biol 2003;23(24) 9389-9404.
  64. 64. Garcia-CardenaG.FanR.SternD. F.LiuJ.SessaW. C. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 271 1996 44 2723727240 .
  65. 65. VenemaV. J.JuH.ZouR.VenemaR. C. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 272 1997 45 2818728190 .
  66. 66. Garcia-CardenaG.MartasekP.BSMastersSkidd. P. M.CouetJ.et al. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 272 1997 41 2543725440 .
  67. 67. SbaaE.FrerartF.FeronO. Caveolae-Dependent Endocytosis in Viral Infection Trends Cardiovasc Med 15 2005 5 157162 .
  68. 68. IsshikiM.AndersonR. G. Function of caveolae 2 Ca2+ entry and Ca2+-dependent signal transduction. Traffic 2003;4 11 717723 .
  69. 69. BergdahlA.SwardK. Caveolae-Dependent Endocytosis in Viral Infection Can J Physiol Pharmacol 82 2004 5 289299 .
  70. 70. WangX. L.YeD.PetersonT. E.CaoS.ShahV. H.et al. Caveolae targeting and regulation of large conductance Ca(2+)-activated K+ channels in vascular endothelial cells. J Biol Chem 280 2005 12 1165611664 .
  71. 71. Darby PJ, Kwan CY, Daniel EE. Caveolae-Dependent Endocytosis in Viral Infection. Am J Physiol Lung Cell Mol Physiol 2000;279(6) L12261235 .
  72. 72. LiuL.AskariA. Beta-subunit of cardiac Na+-K+-ATPase dictates the concentration of the functional enzyme in caveolae. Am J Physiol Cell Physiol 2006 C569578 .
  73. 73. RazaniB.LisantiM. P.Twodistinct.caveolindomainsmediate.thefunctional.interactionof.caveolinwithprotein.kinaseA. Am J Physiol Cell Physiol 2001 C12411250 .
  74. 74. SampsonL. J.HayabuchiY.StandenN. B.DartC. Caveolae-Dependent Endocytosis in Viral Infection Circ Res 95 2004 10 10121018 .
  75. 75. HeijnenH. F.WaaijenborgS.JDCrapoBowler. R. P.AkkermanJ. W.et al.Colocalizationof. e. N. O. S.thecatalytic.subunitof. P. K. A.inendothelial.celljunctions. a.cluefor.regulatedN. O.production JD, Bowler RP, Akkerman JW, et al. Caveolae-Dependent Endocytosis in Viral Infection J Histochem Cytochem 52 2004 10 12771285 .
  76. 76. Peart JN, Headrick JP. Adenosinergic cardioprotection: multiple receptors, multiple pathways. Pharmacol Ther 114 2007 2 208221 .
  77. 77. FoxT. E.HouckK. L.O’NeillS. M.NagarajanM.StoverT. C.et al. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 282 2007 17 1245012457 .
  78. 78. PrevostelC.AliceV.JoubertD.ParkerP. J.Proteinkinase.C(alphaactively.downregulatesthrough.caveolae-dependenttraffic.toan.endosomalcompartment. Joubert D, Parker PJ. Protein kinase C(alpha) actively downregulates through caveolae-dependent traffic to an endosomal compartment. J Cell Sci 2000 ( Pt 14) 2575-2584.
  79. 79. KrajewskaW. M.MaslowskaI.Caveolinsstructure.functionin.signaltransduction. Cell Mol Biol Lett 9 2004 2 195220 .
  80. 80. SeddingD. G.HermsenJ.SeayU.EickelbergO.KummerW.et al.Caveolinfacilitatesmechanosensitive.proteinkinase. B. .Aktsignaling.invitro.invivo. Circ Res 96 2005 6 635642 .
  81. 81. NilssonR.AhmadF.SwardK.AnderssonU.WestonM.et al. Caveolae-Dependent Endocytosis in Viral Infection. Cell Signal 2006;18 10 17131721 .
  82. 82. MurrayF.InselP. A.YuanJ. X. Role of O(2)-sensitive K(+) and 2 channels in the regulation of the pulmonary circulation: potential role of caveolae and implications for high altitude pulmonary edema. Respir Physiol Neurobiol 2006;151(2-3) 192-208.
  83. 83. Houslay MD, Baillie GS, Maurice DH. cAMP-Specific phosphodiesterase-4 enzymes in the cardiovascular system: a molecular toolbox for generating compartmentalized cAMP signaling. Circ Res 100 2007 7 950966 .
  84. 84. MoroneN.FujiwaraT.MuraseK.KasaiR. S.IkeH.et al. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 174 2006 6 851862 .
  85. 85. RichterT.FloetenmeyerM.FergusonC.GaleaJ.GohJ.et al.High-resolution. D.quantitativeanalysis.ofcaveolar.ultrastructurecaveola-cytoskeletoninteractions. Traffic 9 2008 6 893909 .
  86. 86. PartonR. G.JoggerstB.SimonsK. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 127 1994 5 11991215 .
  87. 87. MundyD. I.MachleidtT.YingY. S.AndersonR. G.BloomG. S. Caveolae-Dependent Endocytosis in Viral Infection J Cell Sci 2002 22) 4327 EOF39 EOF .
  88. 88. Henley JR, Krueger EW, Oswald BJ, McNiven MA. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 141 1998 1 8599 .
  89. 89. OhP.Mc IntoshD. P.SchnitzerJ. E. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 141 1998 1 101114 .
  90. 90. YaoQ.ChenJ.CaoH.JDOrth McCaffery. J. M.et al.Caveolininteractsdirectly.withdynamin-. J, Cao H, Orth JD, McCaffery JM, et al. Caveolae-Dependent Endocytosis in Viral Infection J Mol Biol 348 2005 2 491501 .
  91. 91. PelkmansL.PuntenerD.HeleniusA. Caveolae-Dependent Endocytosis in Viral Infection. Science 296 2002 5567 535539 .
  92. 92. SharmaD. K.BrownJ. C.ChoudhuryA.PetersonT. E.HolickyE.et al. Caveolae-Dependent Endocytosis in Viral Infection Mol Biol Cell 15 2004 7 31143122 .
  93. 93. LeeE.De CamilliP. Caveolae-Dependent Endocytosis in Viral Infection. Proc Natl Acad Sci U S A 99 2002 1 161166 .
  94. 94. JDOrthKrueger. E. W.CaoH.MAMc Niven Caveolae-Dependent Endocytosis in Viral Infection. Proc Natl Acad Sci U S A 99 2002 1 167172 .
  95. 95. MAMc NivenKim. L.KruegerE. W.JDOrthCao. H.et al. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 151 2000 1 187198 .
  96. 96. SchaferD. A.WeedS. A.BinnsD.KarginovA. V.ParsonsJ. T.et al. Caveolae-Dependent Endocytosis in Viral Infection Curr Biol 2002;12 21 18521857 .
  97. 97. KruegerE. W.JDOrthCao. H.MAMc NivenA.dynamin-cortactin-Arp2complexmediates.actinreorganization.ingrowth.factor-stimulatedcells. Mol Biol Cell 14 2003 3 10851096 .
  98. 98. Predescu SA, Predescu DN, Timblin BK, Stan RV, Malik AB. Caveolae-Dependent Endocytosis in Viral Infection Mol Biol Cell 14 2003 12 49975010 .
  99. 99. StahlhutM.van DeursB. Caveolae-Dependent Endocytosis in Viral Infection Mol Biol Cell 11 2000 1 325337 .
  100. 100. MADouceyBender. F. C.HessD.HofsteengeJ.BronC.Caveolininteractswith.thechaperone.complexT. C. 1 and modulates its protein folding activity. Cell Mol Life Sci 2006;63(7-8) 939 EOF948 EOF .
  101. 101. HeadB. P.PatelH. H.RothD. M.MurrayF.SwaneyJ. S.et al. Caveolae-Dependent Endocytosis in Viral Infection J Biol Chem 281 2006 36 2639126399 .
  102. 102. RavidD.ChuderlandD.LandsmanL.LavieY.ReichR.et al.FilaminA.isa.novelcaveolin-1-dependent.target-I-stimulatedin. I. G. F.cancercell.migration Exp Cell Res 314 2008 15 27622773 .
  103. 103. SverdlovM.ShininV.PlaceA. T.CastellonM.MinshallR. D.FilaminA.regulatescaveolae.internalizationtraffickingin.endothelialcells. Mol Biol Cell 20 2009 21 45314540 .
  104. 104. PelkmansL.KartenbeckJ.HeleniusA. Caveolae-Dependent Endocytosis in Viral Infection Nat Cell Biol 3 2001 5 473483 .
  105. 105. Le PU, Nabi IR. Caveolae-Dependent Endocytosis in Viral Infection J Cell Sci 2003 6) 1059 EOF71 EOF .
  106. 106. HayerA.StoeberM.RitzD.EngelS.MeyerH. H.et al.Caveolinisubiquitinated.targetedto.intralumenalvesicles.inendolysosomes.fordegradation. J Cell Biol 191 2010 3 615629 .
  107. 107. GilbertJ.BenjaminT. Caveolae-Dependent Endocytosis in Viral Infection 1 J Virol 2004;78 22 1225912267 .
  108. 108. StangE.KartenbeckJ.PartonR. G. Caveolae-Dependent Endocytosis in Viral Infection Mol Biol Cell 1997;8 1 4757 .
  109. 109. AllanderT.AndreassonK.GuptaS.BjerknerA.BogdanovicG.et al. Caveolae-Dependent Endocytosis in Viral Infection J Virol 81 2007 8 41304136 .
  110. 110. FengH.ShudaM.ChangY.MooreP. S. Caveolae-Dependent Endocytosis in Viral Infection. Science 319 2008 5866 10961100 .
  111. 111. Gaynor AM, Nissen MD, Whiley DM, Mackay IM, Lambert SB, et al. Caveolae-Dependent Endocytosis in Viral Infection. PLoS Pathog 2007 e64.
  112. 112. MacoveiA.RadulescuC.LazarC.PetrescuS.DurantelD.et al.HepatitisB.virusrequires.intactcaveolin.functionfor.productiveinfection.inHepa. R. G.cells J Virol 84 2010 1 243253 .
  113. 113. BeerC.DSAndersenRojek. A.PedersenL. Caveolae-Dependent Endocytosis in Viral Infection J Virol 79 2005 16 1077610787 .
  114. 114. StuartA. D.EustaceH. E.Mc KeeT. A.BrownT. D. A.novelcell.entrypathway.fora. D. A.F-usinghuman.enterovirusis.dependenton.lipidrafts. J Virol 76 2002 18 93079322 .
  115. 115. BrownG.CEJeffree McDonald. T.RixonH. W.JDAitkenet.al Caveolae-Dependent Endocytosis in Viral Infection. Virology 2004;327 2 175185 .
  116. 116. WerlingD.HopeJ. C.ChaplinP.CollinsR. A.TaylorG.et al. Caveolae-Dependent Endocytosis in Viral Infection J Leukoc Biol 66 1999 1 5058 .
  117. 117. CantinC.HolgueraJ.FerreiraL.VillarE.Munoz-BarrosoI. Caveolae-Dependent Endocytosis in Viral Infection J Gen Virol 2007 2) 559 EOF69 EOF .
  118. 118. MarjomakiV.PietiainenV.MatilainenH.UplaP.IvaskaJ.et al. Caveolae-Dependent Endocytosis in Viral Infection J Virol 2002;76 4 18561865 .
  119. 119. TsaiB.GilbertJ. M.StehleT.LencerW.BenjaminT. L.et al. Caveolae-Dependent Endocytosis in Viral Infection 40 Embo J 2003;22 17 43464355 .
  120. 120. EngelS.HegerT.ManciniR.HerzogF.KartenbeckJ.et al. Caveolae-Dependent Endocytosis in Viral Infection. J Virol 2011;85 9 41984211 .
  121. 121. JiangM.AbendJ. R.TsaiB.MJImperiale Caveolae-Dependent Endocytosis in Viral Infection. J Virol 83 2009 3 13501358 .
  122. 122. LieblD.DifatoF.HornikovaL.MannovaP.StokrovaJ.et al. Caveolae-Dependent Endocytosis in Viral Infection. J Virol 80 2006 9 46104622 .
  123. 123. QuerbesW.BAO’HaraWilliams. G.AtwoodW. J. Caveolae-Dependent Endocytosis in Viral Infection J Virol 80 2006 19 94029413 .
  124. 124. MannovaP.ForstovaJ. Caveolae-Dependent Endocytosis in Viral Infection J Virol 77 2003 3 16721681 .
  125. 125. QianM.CaiD.VerheyK. J.TsaiB. A.lipidreceptor.sortspolyomavirus.fromthe.endolysosometo.theendoplasmic.reticulumto.causeinfection. P.Lo PLoS Pathog 2009 e1000465.
  126. 126. LanioszV.HolthusenK. A.MenesesP. I. Caveolae-Dependent Endocytosis in Viral Infection. J Virol 2008;82 13 62886298 .
  127. 127. LanioszV.DabydeenS. A.MAHavensMeneses. P. I. Caveolae-Dependent Endocytosis in Viral Infection. J Virol 83 2009 16 82218232 .
  128. 128. SchelhaasM.MalmstromJ.PelkmansL.HaugstetterJ.EllgaardL.et al.SimianVirus. .dependson. E. R.proteinfolding.qualitycontrol.factorsfor.entryinto.hostcells. Cell 131 2007 3 516529 .
  129. 129. GeigerR.AndritschkeD.FriebeS.HerzogF.LuisoniS.etal. B. A. Caveolae-Dependent Endocytosis in Viral Infection Nat Cell Biol 2011;13 11 13051314 .
  130. 130. PithaJ.IrieT.SklarP. B.NyeJ. S. Caveolae-Dependent Endocytosis in Viral Infection Life Sci 43 1988 6 493502 .
  131. 131. Hussain KM, Leong KL, Ng MM, Chu JJ. Caveolae-Dependent Endocytosis in Viral Infection 71 J Biol Chem 286 1 309321 .
  132. 132. Van HammeE.DewerchinH. L.CornelissenE.VerhasseltB.NauwynckH. J.Clathrincaveolae-independententry.offeline.infectiousperitonitis.virusin.monocytesdepends.ondynamin.J. Caveolae-Dependent Endocytosis in Viral Infection. J Gen Virol 2008 9) 2147 EOF2158 EOF -2156.
  133. 133. MoriyamaT.MarquezJ. P.WakatsukiT.SorokinA. Caveolae-Dependent Endocytosis in Viral Infection J Virol 81 2007 16 85528562 .
  134. 134. MacoveiA.RadulescuC.LazarC.PetrescuS.DurantelD.et al.HepatitisB.virusrequires.intactcaveolin.functionfor.productiveinfection.inHepa. R. G.cells J Virol 84 841 1 243253 .
  135. 135. SpodenG.FreitagK.HusmannM.BollerK.SappM.et al.Clathrincaveolin-independententry.ofhuman.papillomavirustype.16--involvementof.tetraspanin-enrichedmicrodomains. . T. E.MsLoP. PLoS One 2008 e3313 EOF .
  136. 136. VercauterenD.VandenbrouckeR. E.JonesA. T.RejmanJ.DemeesterJ.et al. Caveolae-Dependent Endocytosis in Viral Infection. Mol Ther 18 2010 3 561569 .
  137. 137. RodalS. K.SkrettingG.GarredO.VilhardtF.van DeursB.et al. Caveolae-Dependent Endocytosis in Viral Infection Mol Biol Cell 10 1999 4 961974 .
  138. 138. KreyT.ThielH. J.RumenapfT. Caveolae-Dependent Endocytosis in Viral Infection J Virol 79 2005 7 41914200 .
  139. 139. ImelliN.MeierO.BouckeK.HemmiS.GreberU. F. Caveolae-Dependent Endocytosis in Viral Infection 2 J Virol 2004;78 6 30893098 .
  140. 140. Liao JK. Caveolae-Dependent Endocytosis in Viral Infection Curr Opin Lipidol 16 2005 6 624629 .
  141. 141. HolschermannH.SchusterD.ParvizB.HaberboschW.TillmannsH.et al.Statinsprevent. N.F-kappaB.transactivationindependently.ofthe. I. K.K-pathwayin.humanendothelial.cells Atherosclerosis 185 2006 2 240245 .
  142. 142. SunD.FernandesG. Caveolae-Dependent Endocytosis in Viral Infection Cell Immunol 223 2003 1 5262 .
  143. 143. BolardJ. How do the polyene macrolide antibiotics affect the cellular membrane properties? Biochim Biophys Acta 1986 257-304.
  144. 144. Orlandi PA, Fishman PH. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 141 1998 4 905915 .
  145. 145. SchnitzerJ. E.OhP.PinneyE.AllardJ. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Biol 127 1994 5 12171232 .
  146. 146. vonGersdorff. K.SandersN. N.VandenbrouckeR.De SmedtS. C.WagnerE.et al. Caveolae-Dependent Endocytosis in Viral Infection Mol Ther 14 2006 5 745753 .
  147. 147. Dangoria NS, Breau WC, Anderson HA, Cishek DM, Norkin LC. Caveolae-Dependent Endocytosis in Viral Infection J Gen Virol 199677 ( Pt 9) 2173 EOF82 EOF .
  148. 148. LiE.StupackD. G.BrownS. L.KlemkeR.SchlaepferD. D.et al.Associationof.p1C. A. 130CAS 130 with phosphatidylinositol-3-OH kinase mediates adenovirus cell entry. J Biol Chem 2000;275(19) 14729-14735.
  149. 149. Sharma-WaliaN.NaranattP. P.KrishnanH. H.ZengL.ChandranB. Kaposi’s sarcoma-associated herpesvirus/human herpesvirus 8 envelope glycoprotein gB induces the integrin-dependent focal adhesion kinase-Src-phosphatidylinositol 3-kinase-rho GTPase signal pathways and cytoskeletal rearrangements. J Virol 78 2004 8 42074223 .
  150. 150. CubellsL.Vila deMuga. S.TebarF.WoodP.EvansR.et al. Annexin A6-induced alterations in cholesterol transport and caveolin export from the Golgi complex. Traffic 8 2007 11 15681589 .
  151. 151. Anderson RG. Caveolae: where incoming and outgoing messengers meet. Proc Natl Acad Sci U S A 90 1993 23 1090910913 .
  152. 152. AndersonH. A.ChenY.NorkinL. C. Caveolae-Dependent Endocytosis in Viral Infection Mol Biol Cell 7 1996 11 18251834 .
  153. 153. PhoM. T.AshokA.AtwoodW. J. J. C.virusenters.humanglial.cellsby.clathrin-dependentreceptor-mediated.endocytosisJ. Caveolae-Dependent Endocytosis in Viral Infection J Virol 74 2000 5 22882292 .
  154. 154. Empig CJ, Goldsmith MA. Caveolae-Dependent Endocytosis in Viral Infection. J Virol 76 2002 10 52665270 .
  155. 155. AballayA.StahlP. D.MayorgaL. S. Caveolae-Dependent Endocytosis in Viral Infection J Cell Sci 1999 ( Pt 15) 2549 EOF57 EOF .
  156. 156. CohenP.HolmesC. F.TsukitaniY.Okadaicacid. a.newprobe.forthe.studyof.cellularregulation. Trends Biochem Sci 15 1990 3 98102 .
  157. 157. LajoieP.NabiI. R. Caveolae-Dependent Endocytosis in Viral Infection. J Cell Mol Med 11 2007 4 644653 .
  158. 158. Taylor MP, Koyuncu OO, Enquist LW. Caveolae-Dependent Endocytosis in Viral Infection. Nat Rev Microbiol 9 2011 6 427439 .
  159. 159. BreuzaL.CorbyS.ArsantoJ. P.DelgrossiM. H.ScheiffeleP.et al. Caveolae-Dependent Endocytosis in Viral Infection J Cell Sci 2002;115(Pt 23) 4457 EOF67 EOF .
  160. 160. HailstonesD.SleerL. S.PartonR. G.StanleyK. K. Caveolae-Dependent Endocytosis in Viral Infection J Lipid Res 39 1998 2 369379 .
  161. 161. HuangW. R.WangY. C.ChiP. I.WangL.WangC. Y.et al. Caveolae-Dependent Endocytosis in Viral Infection. J Biol Chem 2011;286(35) 30780-30794.
  162. 162. TrouetD.HermansD.DroogmansG.NiliusB.EggermontJ. Caveolae-Dependent Endocytosis in Viral Infection. Biochem Biophys Res Commun 284 2001 2 461465 .
  163. 163. RoyS.LuetterforstR.HardingA.ApolloniA.EtheridgeM.et al. Caveolae-Dependent Endocytosis in Viral Infection Nat Cell Biol 1 1999 2 98105 .
  164. 164. GianniT.GattaV.Campadelli-FiumeG.{alpha} Caveolae-Dependent Endocytosis in Viral Infection Proc Natl Acad Sci U S A 107 10751 51 2226022265 .
  165. 165. LakadamyaliM.MJRustZhuang. X. Caveolae-Dependent Endocytosis in Viral Infection Microbes Infect 6 2004 10 929936 .
  166. 166. ShatzM.LustigG.ReichR.LiscovitchM.CaveolinmutantsP1. 132L 132 and Y14F are dominant negative regulators of invasion, migration and aggregation in H1299 lung cancer cells. Exp Cell Res 2010;316(10) 1748-1762.
  167. 167. Conner SD, Schmid SL. Caveolae-Dependent Endocytosis in Viral Infection Nature 422 2003 6927 3744 .
  168. 168. Insel PA, Patel HH. Caveolae-Dependent Endocytosis in Viral Infection Br J Pharmacol 150 2007 3 251254 .

Written By

Norica Branza-Nichita, Alina Macovei and Catalin Lazar

Submitted: 12 April 2012 Published: 06 July 2012