101
|
Rogée S, Grellier E, Bernard C, Loyens A, Beauvillain JC, D'halluin JC, Colin M. Intracellular trafficking of a fiber-modified adenovirus using lipid raft/caveolae endocytosis. Mol Ther 2007; 15:1963-72. [PMID: 17712334 DOI: 10.1038/sj.mt.6300283] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Most adenoviral vectors (HAdvs) elaborated for gene therapy are derived from serotype 5 viruses that use clathrin-coated vesicle endocytosis for cell entry. However, it appears that adenoviral vectors are able to take advantage of lipid raft/caveolae endocytosis to infect cells. In vivo targeting of a therapeutic gene to specific cells by vector engineering has become a major focus of gene therapy research. Yet, modification of adenoviral tropism, especially fiber gene engineering, can induce deficient intracellular trafficking of the viral particle, with a shift in subcellular localization resulting in extensive exocytosis. In this study we demonstrate that uptake of a fiber-modified adenovirus using lipid raft/caveolae endocytosis leads to non-altered intracellular trafficking without endosomal retention. Moreover, activation of lipid raft structures by this vector leads to the formation of "mega-caveosomes". These results demonstrate that, by forcing adenoviruses to take advantage of a non-clathrin, non-classical endocytic pathway, it is possible to compensate for the deficiency in endosomolysis that is associated with the use of some of the fiber-modified adenoviral constructs. Moreover, it renders such vectors ideal candidates for infecting human coxsackie and adenoviruses receptor (hCAR) negative cells.
Collapse
Affiliation(s)
- Sophie Rogée
- Institut National de la Santé et de la Recherche Médicale U837, Lille, France
| | | | | | | | | | | | | |
Collapse
|
102
|
Li PP, Nguyen AP, Qu Q, Jafri QH, Aungsumart S, Cheng RH, Kasamatsu H. Importance of calcium-binding site 2 in simian virus 40 infection. J Virol 2007; 81:6099-105. [PMID: 17360742 PMCID: PMC1900253 DOI: 10.1128/jvi.02195-06] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The exposure of molecular signals for simian virus 40 (SV40) cell entry and nuclear entry has been postulated to involve calcium coordination at two sites on the capsid made of Vp1. The role of calcium-binding site 2 in SV40 infection was examined by analyzing four single mutants of site 2, the Glu160Lys, Glu160Arg, Glu157Lys (E157K), and Glu157Arg mutants, and an E157K-E330K combination mutant. The last three mutants were nonviable. All mutants replicated viral DNA normally, and all except the last two produced particles containing all three capsid proteins and viral DNA. The defect of the site 1-site 2 E157K-E330K double mutant implies that at least one of the sites is required for particle assembly in vivo. The nonviable E157K particles, about 10% larger in diameter than the wild type, were able to enter cells but did not lead to T-antigen expression. Cell-internalized E157K DNA effectively coimmunoprecipitated with anti-Vp1 antibody, but little of the DNA did so with anti-Vp3 antibody, and none was detected in anti-importin immunoprecipitate. Yet, a substantial amount of Vp3 was present in anti-Vp1 immune complexes, suggesting that internalized E157K particles are ineffective at exposing Vp3. Our data show that E157K mutant infection is blocked at a stage prior to the interaction of the Vp3 nuclear localization signal with importins, consistent with a role for calcium-binding site 2 in postentry steps leading to the nuclear import of the infecting SV40.
Collapse
Affiliation(s)
- Peggy P Li
- Molecular Biology Institute, 456 Boyer Hall, University of California at Los Angeles, 611 East Charles E. Young Dr., Box 951570, Los Angeles, CA 90095-1570, USA
| | | | | | | | | | | | | |
Collapse
|
103
|
Nakanishi A, Itoh N, Li PP, Handa H, Liddington RC, Kasamatsu H. Minor capsid proteins of simian virus 40 are dispensable for nucleocapsid assembly and cell entry but are required for nuclear entry of the viral genome. J Virol 2007; 81:3778-85. [PMID: 17267496 PMCID: PMC1866110 DOI: 10.1128/jvi.02664-06] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
We investigated the roles of simian virus 40 capsid proteins in the viral life cycle by analyzing point mutants in Vp1 and Vp2/3, as well as a deletion mutant lacking the Vp2/3 coding sequence. The Vp1 mutants (V243E and L245E) and the Vp2/3 mutants (F157E-I158E and P164R-G165E-G166R) were previously shown to be defective in Vp1-Vp2/3 interaction and to be noninfectious or poorly infectious, respectively. Here, we show that all these point mutants form stable particles following DNA transfection into cells. The Vp2/3-mutant particles contained very low levels of Vp2/3, whereas the Vp1 mutant particles contained no detectable Vp2/3. As expected, the deletion mutant also formed particles that were noninfectious. We further characterized the two Vp1 point mutants and the deletion mutant. All three mutant particles comprised Vp1 and histone-associated viral DNA, and all were able to enter cells. However, the mutant complexes failed to associate with host importins (owing to the loss of the Vp2/3 nuclear localization signal), and the mutant viral DNAs prematurely dissociated from the Vp1s, suggesting that the nucleocapsids did not enter the nucleus. Consistently, all three mutant particles failed to express large T antigen. Together, our results demonstrate unequivocally that Vp2/3 is dispensable for the formation of nucleocapsids. Further, the nucleocapsids' ability to enter cells implies that Vp1 contains the major determinants for cell attachment and entry. We propose that the major role of Vp2/3 in infectivity is to mediate the nuclear entry of viral DNA.
Collapse
Affiliation(s)
- Akira Nakanishi
- Molecular Biology Institute and Department of Molecular, Cell, and Developmental Biology, 456 Boyer Hall, University of California, Los Angeles, 611 East Charles E. Young Dr., Box 951570, Los Angeles, CA 90095-1570, USA
| | | | | | | | | | | |
Collapse
|
104
|
Medina FA, Cohen AW, de Almeida CJ, Nagajyothi F, Braunstein VL, Teixeira MM, Tanowitz HB, Lisanti MP. Immune dysfunction in caveolin-1 null mice following infection with Trypanosoma cruzi (Tulahuen strain). Microbes Infect 2007; 9:325-33. [PMID: 17317261 PMCID: PMC2737390 DOI: 10.1016/j.micinf.2006.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 12/11/2006] [Accepted: 12/12/2006] [Indexed: 01/29/2023]
Abstract
In recent years, host cell caveolae/caveolins have emerged as potentially important targets for pathogenic microorganisms; therefore, we investigated the role of caveolin-1 (Cav-1) in T. cruzi infection using Cav-1 null mice. Cav-1 null and wild type mice were infected with the virulent Tulahuen strain. The mortality was 100% in both groups, but death was slightly delayed in wild type mice. The parasitemia in the Cav-1 null mice was significantly reduced compared with wild type littermates. Histopathologic examination of the heart revealed numerous pseudocysts, myonecrosis, and marked inflammation, which was similar in both mouse groups. Real-time PCR confirmed these observations. Infection of cultured cardiac fibroblasts obtained from Cav-1 null and wild type mice revealed no differences in infectivity. Determination of serum levels of several inflammatory mediators revealed a striking reduction in IFN-gamma, TNF-alpha and components of the nitric oxide pathway in infected Cav-1 null mice. Infection of wild type mice resulted in the expected enhancement of inflammatory mediators. The defective production of chemokines and cytokines observed in vivo is in part attributed to Cav-1 null macrophages. Despite these marked differences in the response to infection by inflammatory mediators between the two mouse strains, the final outcome was similar. These results suggest that Cav-1 may play an important role in the normal development of immune responses.
Collapse
Affiliation(s)
- Freddy A. Medina
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Alex W. Cohen
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Cecilia J. de Almeida
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
| | - Fnu Nagajyothi
- Departments of Pathology and Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Vicki L. Braunstein
- Departments of Pathology and Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
| | - Mauro Martins Teixeira
- Departamento de Bioquimica e Imunologia, Instituto de Ciencias Biologicas, Universidade Federal de Minas Gerais, Brazil 31270-901
| | - Herbert B. Tanowitz
- Departments of Pathology and Medicine, Albert Einstein College of Medicine, Bronx, NY 10461
- Corresponding Authors: Dr. Michael P. Lisanti, Department of Cancer Biology, Kimmel Cancer Center, Bluemle Life Sciences Building, Room 933, 233 S. 10th Street, Philadelphia, PA 19107. Phone: (215) 503-9295. Fax: (215) 923-1098., or . Dr. Herbert B. Tanowitz, Departments of Pathology and Medicine, 1300 Morris Park Avenue, Bronx, NY 10461. Phone: (718) 430-3342. Fax: (718) 430- 8543., E-mail:
| | - Michael P. Lisanti
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY 10461
- Department of Cancer Biology, Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107
- Muscular and Neurodegenerative Disease Unit, University of Genova and G.Gaslini Pediatric Institute, Largo Gaslini 5, 16147 Genova, Italy
- Corresponding Authors: Dr. Michael P. Lisanti, Department of Cancer Biology, Kimmel Cancer Center, Bluemle Life Sciences Building, Room 933, 233 S. 10th Street, Philadelphia, PA 19107. Phone: (215) 503-9295. Fax: (215) 923-1098., or . Dr. Herbert B. Tanowitz, Departments of Pathology and Medicine, 1300 Morris Park Avenue, Bronx, NY 10461. Phone: (718) 430-3342. Fax: (718) 430- 8543., E-mail:
| |
Collapse
|
105
|
Ning Y, Buranda T, Hudson LG. Activated epidermal growth factor receptor induces integrin alpha2 internalization via caveolae/raft-dependent endocytic pathway. J Biol Chem 2006; 282:6380-7. [PMID: 17179151 DOI: 10.1074/jbc.m610915200] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Elevated expression or activity of the epidermal growth factor (EGF) receptor is common in ovarian cancer and is associated with poor patient prognosis. Our previous studies demonstrated that expression of the constitutively active mutant form of the EGF receptor (EGFRvIII) in ovarian cancer cells led to reduction in integrin alpha2 surface expression, defects in cell spreading, and disruption of focal adhesions. Inhibition of EGFRvIII catalytic activity reversed the response, suggesting that EGF receptor activation regulates integrin alpha2. In this study we found that EGF treatment resulted in a transient loss of integrin alpha2 from the cell surface. Before EGF stimulation, integrin alpha2 and EGF receptors were associated based on biochemical and immuno-colocalization approaches. After EGF treatment, EGF receptor and integrin alpha2 were internalized and segregated into different compartments. Integrin alpha2, but not EGF receptor, was associated with caveolin-1 and GM1 (Gal_1,3GalNAc_1,4(Neu5Ac-_ 2,3)Gal_1,4Glc_1,1-ceramide) gangliosides, suggesting caveolae-mediated endocytosis. Moreover, integrin alpha2 was subsequently targeted to the Golgi apparatus and the endoplasmic reticulum. Together, these findings demonstrate that activated EGF receptor transiently modulates integrin alpha2 cell surface expression and stimulates integrin alpha2 trafficking via caveolae/raft-mediated endocytosis, representing a novel mechanism by which the EGF receptor may regulate integrin-mediated cell behavior.
Collapse
Affiliation(s)
- Yan Ning
- College of Pharmacy, University of New Mexico, Albuquerque, New Mexico 87131, USA
| | | | | |
Collapse
|
106
|
Daniels R, Rusan NM, Wadsworth P, Hebert DN. SV40 VP2 and VP3 Insertion into ER Membranes Is Controlled by the Capsid Protein VP1: Implications for DNA Translocation out of the ER. Mol Cell 2006; 24:955-66. [PMID: 17189196 DOI: 10.1016/j.molcel.2006.11.001] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2006] [Revised: 10/20/2006] [Accepted: 11/01/2006] [Indexed: 02/03/2023]
Abstract
Nonenveloped viruses such as Simian Virus 40 (SV40) exploit established cellular pathways for internalization and transport to their site of penetration. By analyzing mutant SV40 genomes that do not express VP2 or VP3, we found that these structural proteins perform essential functions that are regulated by VP1. VP2 significantly enhanced SV40 particle association with the host cell, while VP3 functioned downstream. VP2 and VP3 both integrated posttranslationally into the endoplasmic reticulum (ER) membrane. Association with VP1 pentamers prevented their ER membrane integration, indicating that VP1 controls the function of VP2 and VP3 by directing their localization between the particle and the ER membrane. These findings suggest a model in which VP2 aids in cell binding. After capsid disassembly within the ER lumen, VP3, and perhaps VP2, oligomerizes and integrates into the ER membrane, potentially creating a viroporin that aids in viral DNA transport out of the ER.
Collapse
Affiliation(s)
- Robert Daniels
- Department of Biochemistry and Molecular Biology, University of Massachusetts, Amherst, Massachusetts 01003, USA
| | | | | | | |
Collapse
|
107
|
Chinnapen DJF, Chinnapen H, Saslowsky D, Lencer WI. Rafting with cholera toxin: endocytosis and trafficking from plasma membrane to ER. FEMS Microbiol Lett 2006; 266:129-37. [PMID: 17156122 PMCID: PMC3511785 DOI: 10.1111/j.1574-6968.2006.00545.x] [Citation(s) in RCA: 183] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Cholera toxin (CT), and members of the AB(5) family of toxins enter host cells and hijack the cell's endogenous pathways to induce toxicity. CT binds to a lipid receptor on the plasma membrane (PM), ganglioside GM1, which has the ability to associate with lipid rafts. The toxin can then enter the cell by various modes of receptor-mediated endocytosis and traffic in a retrograde manner from the PM to the Golgi and the endoplasmic reticulum (ER). Once in the ER, a portion of the toxin is unfolded and retro-translocated to the cytosol so as to induce disease. GM1 is the vehicle that carries CT from PM to ER. Thus, the toxin pathway from PM to ER is a lipid-based sorting pathway, which is potentially meditated by the determinants of the GM1 ganglioside structure itself.
Collapse
Affiliation(s)
- Daniel J.-F. Chinnapen
- GI Cell Biology, Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | | | - David Saslowsky
- GI Cell Biology, Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Wayne I. Lencer
- GI Cell Biology, Children’s Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- The Harvard Digestive Diseases Center, Boston, MA, USA
| |
Collapse
|
108
|
Nakanishi A, Li PP, Qu Q, Jafri QH, Kasamatsu H. Molecular dissection of nuclear entry-competent SV40 during infection. Virus Res 2006; 124:226-30. [PMID: 17112617 PMCID: PMC1847345 DOI: 10.1016/j.virusres.2006.10.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2006] [Revised: 10/04/2006] [Accepted: 10/05/2006] [Indexed: 11/23/2022]
Abstract
To establish viral infection, SV40 must expose nuclear localization signals (NLSs) that are internal in the virion architecture in order to enter the nucleus via interaction with the host's nuclear import machinery, which includes importin alpha and importin beta. The time course for SV40 association with the importins in infected cells was examined. The viral DNA associated with importin alpha by 1.5h post infection, before associating with the importin beta nuclear import receptor, by 3h post infection. Only a small fraction of cell-internalized SV40 that contained viral DNA was bound by the two importins. This fraction, termed "nuclear entry-competent SV40," was slightly smaller than the virion but, importantly, was larger than the viral chromatin and contained both Vp1 and Vp3. Furthermore, the internalized viral DNA in either anti-importin or anti-Vp3 immune complexes was sensitive to DNase I, whereas the viral DNA in mature virions was resistant. All these results suggest that once SV40 enters the cytoplasm, it undergoes an architectural modification that exposes the virion's NLSs for nuclear entry.
Collapse
Affiliation(s)
- Akira Nakanishi
- Molecular Biology Institute and Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Peggy P. Li
- Molecular Biology Institute and Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Qiumin Qu
- Molecular Biology Institute and Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Qumber H. Jafri
- Molecular Biology Institute and Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
| | - Harumi Kasamatsu
- Molecular Biology Institute and Department of Molecular, Cell, and Developmental Biology, University of California at Los Angeles, Los Angeles, CA, 90095, USA
- *Corresponding author. Mailing address: Molecular Biology Institute, 456 Boyer Hall, University of California, Los Angeles, 611 East Charles E. Young Dr., Box 951570, Los Angeles, CA 90095-1570. Phone: (310) 825-3048. Fax: (310) 206-7286. E-mail:
| |
Collapse
|
109
|
Akita H, Hama S, Mizuguchi H, Harashima H. [Development of non-viral vector based on the quantitative comparison of intracellular trafficking with viral vector]. YAKUGAKU ZASSHI 2006; 126:1047-57. [PMID: 17077611 DOI: 10.1248/yakushi.126.1047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
For the development of efficient gene vector, intracellular processes such as cellular uptake, endosomal release and nuclear delivery must be overcome. Viruses have also evolved and have developed sophisticated mechanisms for controlling intracellular trafficking for the efficient delivery of their genomes to nuclei in host cells for symbiosis. In the light of these mechanisms, various kinds of artificial devices have been developed to overcome the intracellular barriers. However, in the majority of studies, variation of the transfection activity before and after the modification of devices was evaluated, and intracellular trafficking remained unclear. Therefore, it is understand to recognize which of the intracellular barrier should be intensively improved to enhance the transfection activity. To clarify the rate-limited process in the current non-viral vector, we compared the intracellular trafficking between adenovirus and LipofectAMINE PLUS. As a result, we found that difference of the transfection efficiency between adenovirus and LipofectAMINE PLUS was dominantly derived from the differences on transcription activity. Therefore it is essential to consider the regulation of the intranuclear events to improve the transfection activity of artificial vector.
Collapse
Affiliation(s)
- Hidetaka Akita
- Faculty of Pharmaceutical Sciences, Hokkaido University, Kita-12 Nishi-6, Kita-ku, Sapporo City 060-0812, Japan.
| | | | | | | |
Collapse
|
110
|
Abstract
Virus entry, assembly, and budding are important processes in the replication cycle of a virus. Viruses are dependent on host living cells for their replication. Viruses use the proliferative mechanism of host cells for replication of viral components. Lipid rafts, specific membrane microdomains play a critical role in virus replication because localizing and concentrating viral components in the microdomains for entry, assembly, and budding of various types of virus. In this review, we describe the involvement of membrane lipid rafts in the virus replication cycle with our current findings for understanding the role of membrane lipid rafts in virus infection.
Collapse
Affiliation(s)
- Takashi Suzuki
- COE Program in the 21st Century, Department of Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, Japan.
| | | |
Collapse
|
111
|
Heo TH, Lee SM, Bartosch B, Cosset FL, Kang CY. Hepatitis C virus E2 links soluble human CD81 and SR-B1 protein. Virus Res 2006; 121:58-64. [PMID: 16725222 DOI: 10.1016/j.virusres.2006.04.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2005] [Revised: 04/11/2006] [Accepted: 04/18/2006] [Indexed: 11/22/2022]
Abstract
Limited information is available regarding hepatitis C virus (HCV) entry events. Viral attachment and infection studies have been performed using HCV envelope glycoprotein (E2) and HCV pseudo-particle (HCVpp) models to obtain general information about the early entry events. However, the details involved in each step of viral entry into human cells are still obscure. This study provides molecular clue for the formation of a heteromultimeric complex as a possible post-attachment step. Among several putative receptors, human CD81 and scavenger receptor class B type 1 (SR-B1) have been demonstrated as considerable determinants in infectious outcome as well as attachment. In this study, we provide molecular evidence demonstrating that HCV E2 links soluble CD81 and SR-B1 protein together. This physical neighboring might explain why both CD81 and SR-B1 are indispensable factors for HCVpp infection. These data further elucidate our understanding of HCV entry and provide new insight into directing future studies identifying novel liver-specific fusion receptor(s).
Collapse
Affiliation(s)
- Tae-Hwe Heo
- Laboratory of Immunology and the Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Shillim-Dong, Kwanak-Gu, 151-742, Korea
| | | | | | | | | |
Collapse
|
112
|
Lilley BN, Gilbert JM, Ploegh HL, Benjamin TL. Murine polyomavirus requires the endoplasmic reticulum protein Derlin-2 to initiate infection. J Virol 2006; 80:8739-44. [PMID: 16912321 PMCID: PMC1563856 DOI: 10.1128/jvi.00791-06] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The pathways by which viruses enter cells are diverse, but in all cases, infection necessitates the transfer of the viral genome across a cellular membrane. Polyomavirus (Py) particles, after binding to glycolipid and glycoprotein receptors at the cell surface, are delivered to the lumen of the endoplasmic reticulum (ER). The nature and extent of virus disassembly in the ER, how the viral genome is transported to the cytosol and subsequently to the nucleus, and whether any cellular proteins are involved are not known. Here, we identify an ER-resident protein, Derlin-2, a factor implicated in the removal of misfolded proteins from the ER for cytosolic degradation, as a component of the machinery required for mouse Py to establish an infection. Inhibition of Derlin-2 function by expression of either a dominant-negative form of Derlin-2 or a short hairpin RNA that reduces Derlin-2 levels blocks Py infection by 50 to 75%. The block imposed by Derlin-2 inhibition occurs after the virus reaches the ER and can be bypassed by the introduction of Py DNA into the cytosol. These findings suggest a mode of Py entry that involves cytosolic access via the quality control machinery in the ER.
Collapse
Affiliation(s)
- Brendan N Lilley
- Department of Pathology, Havard Medical School, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
113
|
Medina FA, de Almeida CJ, Dew E, Li J, Bonuccelli G, Williams TM, Cohen AW, Pestell RG, Frank PG, Tanowitz HB, Lisanti MP. Caveolin-1-deficient mice show defects in innate immunity and inflammatory immune response during Salmonella enterica serovar Typhimurium infection. Infect Immun 2006; 74:6665-74. [PMID: 16982844 PMCID: PMC1698056 DOI: 10.1128/iai.00949-06] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
A number of studies have shown an association of pathogens with caveolae. To this date, however, there are no studies showing a role for caveolin-1 in modulating immune responses against pathogens. Interestingly, expression of caveolin-1 has been shown to occur in a regulated manner in immune cells in response to lipopolysaccharide (LPS). Here, we sought to determine the role of caveolin-1 (Cav-1) expression in Salmonella pathogenesis. Cav-1(-/-) mice displayed a significant decrease in survival when challenged with Salmonella enterica serovar Typhimurium. Spleen and tissue burdens were significantly higher in Cav-1(-/-) mice. However, infection of Cav-1(-/-) macrophages with serovar Typhimurium did not result in differences in bacterial invasion. In addition, Cav-1(-/-) mice displayed increased production of inflammatory cytokines, chemokines, and nitric oxide. Regardless of this, Cav-1(-/-) mice were unable to control the systemic infection of Salmonella. The increased chemokine production in Cav-1(-/-) mice resulted in greater infiltration of neutrophils into granulomas but did not alter the number of granulomas present. This was accompanied by increased necrosis in the liver. However, Cav-1(-/-) macrophages displayed increased inflammatory responses and increased nitric oxide production in vitro in response to Salmonella LPS. These results show that caveolin-1 plays a key role in regulating anti-inflammatory responses in macrophages. Taken together, these data suggest that the increased production of toxic mediators from macrophages lacking caveolin-1 is likely to be responsible for the marked susceptibility of caveolin-1-deficient mice to S. enterica serovar Typhimurium.
Collapse
Affiliation(s)
- Freddy A Medina
- Department of Cancer Biology, Kimmel Cancer Center, Bluemle Life Sciences Building, Room 933, 233 S. 10th Street, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Gilbert J, Ou W, Silver J, Benjamin T. Downregulation of protein disulfide isomerase inhibits infection by the mouse polyomavirus. J Virol 2006; 80:10868-70. [PMID: 16928750 PMCID: PMC1641741 DOI: 10.1128/jvi.01117-06] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Early stages of infection by the mouse polyomavirus have been studied using HeLa cells stably expressing small interfering RNA to protein disulfide isomerase (PDI). Infectibility measured by nuclear T antigen expression was reduced commensurately with the degree of PDI downregulation. Infectibility was restored by transfection with a plasmid expressing PDI but not with a control expressing catalytically inactive enzyme. Deconvolution microscopy using fluorescently labeled virus and cellular markers showed that virus reaches the endoplasmic reticulum (ER) normally in cells with reduced PDI but subsequently fails to exit the ER. Simian virus 40 infection was not inhibited in PDI-downregulated cells. The results are discussed in terms of structural differences between the two viruses and current knowledge of virus disassembly in the ER.
Collapse
Affiliation(s)
- Joanna Gilbert
- Department of Pathology, NRB-939I, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | | | | | | |
Collapse
|
115
|
Rentsendorj A, Xie J, MacVeigh M, Agadjanian H, Bass S, Kim DH, Rossi J, Hamm-Alvarez SF, Medina-Kauwe LK. Typical and atypical trafficking pathways of Ad5 penton base recombinant protein: implications for gene transfer. Gene Ther 2006; 13:821-36. [PMID: 16482205 DOI: 10.1038/sj.gt.3302729] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The adenovirus (Ad) penton base protein facilitates viral infection by binding cell surface integrins, triggering receptor-mediated endocytosis and mediating endosomal penetration. Given these multiple functions, recombinant penton base proteins have been utilized as non-viral vehicles for gene transfer by our lab and others. Although we have previously demonstrated that penton base-derived vectors undergo integrin-specific binding and cell entry, less than desirable levels of gene expression have led us to re-evaluate the recombinant penton base as an agent for gene delivery. To do so, we have examined here the intracellular trafficking of an Ad serotype 5 (Ad5) recombinant penton base protein (PB). Here, we not only observed that PB utilizes a similar, typical trafficking pathway of whole Ad, but also found that PB entered HeLa cells through pathways not yet identified as contributing to cell entry by the whole virus. We show by high-resolution confocal microscopy and biochemical methods that binding to alphav-integrins is a requirement for cell entry, but that early internalization stages did not substantially pass through clathrin-positive and early endosomal compartments. Moreover, a subpopulation of internalized protein localized with caveolin-positive compartments and Golgi markers, suggesting that a certain percentage of proteins pass through non-clathrin-mediated pathways. Similar to the virus, trafficking toward the nucleus was affected by disruption of microtubules and dynein. The majority of penton base molecules avoided the lysosome while facilitating early vesicle release of low molecular weight dextran molecules. In further support of a vesicle escape capacity, a subpopulation of internalized penton base appeared to enter the nucleus, as observed by high-resolution confocal microscopy and cell fractionation. As a confirmation of these findings, we demonstrate that a recombinant penton base facilitated cytosolic entry of an siRNA molecule as observed by RNA interference of a marker gene. Based on our findings here, we suggest that whereas soluble penton base proteins may enter cells through clathrin- and non-clathrin-mediated pathways, vesicle escape and nuclear delivery appear to be supported by a clathrin-mediated pathway. As our previous efforts have focused on utilizing recombinant penton base proteins as delivery agents for therapeutics, these findings allow us to evaluate the use of the penton base as a cell entry and intracellular trafficking agent, and may be of interest concerning the development of vectors for efficient delivery of therapeutics to cells.
Collapse
Affiliation(s)
- A Rentsendorj
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Krasteva G, Pfeil U, Drab M, Kummer W, König P. Caveolin-1 and -2 in airway epithelium: expression and in situ association as detected by FRET-CLSM. Respir Res 2006; 7:108. [PMID: 16904002 PMCID: PMC1563466 DOI: 10.1186/1465-9921-7-108] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2006] [Accepted: 08/11/2006] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Caveolae are involved in diverse cellular functions such as signal transduction, cholesterol homeostasis, endo- and transcytosis, and also may serve as entry sites for microorganisms. Hence, their occurrence in epithelium of the airways might be expected but, nonetheless, has not yet been examined. METHODS Western blotting, real-time quantitative PCR analysis of abraded tracheal epithelium and laser-assisted microdissection combined with subsequent mRNA analysis were used to examine the expression of cav-1 and cav-2, two major caveolar coat proteins, in rat tracheal epithelium. Fluorescence immunohistochemistry was performed to locate caveolae and cav-1 and -2 in the airway epithelium of rats, mice and humans. Electron-microscopic analysis was used for the identification of caveolae. CLSM-FRET analysis determined the interaction of cav-1alpha and cav-2 in situ. RESULTS Western blotting and laser-assisted microdissection identified protein and transcripts, respectively, of cav-1 and cav-2 in airway epithelium. Real-time quantitative RT-PCR analysis of abraded tracheal epithelium revealed a higher expression of cav-2 than of cav-1. Immunoreactivities for cav-1 and for cav-2 were co-localized in the cell membrane of the basal cells and basolaterally in the ciliated epithelial cells of large airways of rat and human. However, no labeling for cav-1 or cav-2 was observed in the epithelial cells of small bronchi. Using conventional double-labeling indirect immunofluorescence combined with CLSM-FRET analysis, we detected an association of cav-1alpha and -2 in epithelial cells. The presence of caveolae was confirmed by electron microscopy. In contrast to human and rat, cav-1-immunoreactivity and caveolae were confined to basal cells in mice. Epithelial caveolae were absent in cav-1-deficient mice, implicating a requirement of this caveolar protein in epithelial caveolae formation. CONCLUSION These results show that caveolae and caveolins are integral membrane components in basal and ciliated epithelial cells, indicating a crucial role in these cell types. In addition to their physiological role, they may be involved in airway infection.
Collapse
Affiliation(s)
- Gabriela Krasteva
- Institut für Anatomie und Zellbiologie, University of Giessen Lung Center, Justus-Liebig-Universität Giessen, Germany
| | - Uwe Pfeil
- Institut für Anatomie und Zellbiologie, University of Giessen Lung Center, Justus-Liebig-Universität Giessen, Germany
| | - Marek Drab
- Max-Planck Institute for Infection Biology, Department of Molecular Biology, Berlin, Germany
| | - Wolfgang Kummer
- Institut für Anatomie und Zellbiologie, University of Giessen Lung Center, Justus-Liebig-Universität Giessen, Germany
| | - Peter König
- Institut für Anatomie und Zellbiologie, University of Giessen Lung Center, Justus-Liebig-Universität Giessen, Germany
| |
Collapse
|
117
|
Abstract
BK virus (BKV) is a small, non-enveloped, double-stranded DNA virus and a member of the Polyomaviridae family. As the recently recognized etiologic agent of polyomavirus-associated nephropathy, the events involved in BKV invasion of host cells are an important area of study. Using cell culture models, the mechanism by which BKV infects permissive hosts to gain access to the replication machinery within these cells is beginning to unfold. BKV uses an N-linked glycoprotein containing an alpha(2,3)-linked sialic acid as a receptor. After this initial attachment, BKV enters cells through caveolae-mediated endocytosis. Intracellular trafficking via cellular cytoskeletal components follows this relatively slow and cholesterol-dependent internalization. BKV must reach the nucleus for viral transcription and replication to occur. Elucidating the steps of the early viral lifecycle would provide clues to help explain the infectious spread and pathology of this human pathogen.
Collapse
Affiliation(s)
- A S Dugan
- Graduate Program in Pathobiology, Brown University, Providence, RI 02903, USA
| | | | | |
Collapse
|
118
|
Abstract
The BK Virus (BKV) genome is a double-stranded, circular DNA molecule with genetic organization similar to other polyomaviruses, and high homology to JC Virus (JCV) and SV40. The archetypal form of BKV noncoding regulatory region (NCRR) is the infectious form of BKV that replicates in the urothelium and is excreted in the urine. Rearranged forms of the NCRR are found in kidney and other tissues often in association with disease. BKV strains can be assigned to genotype/serotype groups based on sequence variation in the VP1 gene. Sequencing of the complete genomes from patient samples will enhance BKV phylogenetic studies and identify genotypic differences and naturally occurring mutations in BKV that may correlate with incidence and/or severity of a disease. This chapter is a review of the molecular genetics of the BK virus in respect to BKV disease.
Collapse
Affiliation(s)
- Christopher L Cubitt
- Translational Research Laboratory, H Lee Moffitt Cancer and Research Institute, Tampa, Florida, USA
| |
Collapse
|
119
|
Randhawa P, Vats A, Shapiro R. The pathobiology of polyomavirus infection in man. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2006; 577:148-59. [PMID: 16626033 DOI: 10.1007/0-387-32957-9_10] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
This article traces the discovery of polyomaviruses and outlines investigations, which shed light on potential modes of transmission of this increasingly important group of human pathogens. The pathobiology of the virus is summarized with particular reference to interactions with host cell receptors, cell entry, cytoplasmic trafficking, and targeting of the viral genome to the nucleus. This is followed by a discussion of sites of viral latency and factors leading to viral reactivation. Finally, we present biochemical mechanisms that could potentially explain several key elements of tissue pathology characteristic of BKV mediated damage to human kidney.
Collapse
|
120
|
Carbone M, Reale A, Di Sauro A, Sthandier O, Garcia MI, Maione R, Caiafa P, Amati P. PARP-1 interaction with VP1 capsid protein regulates polyomavirus early gene expression. J Mol Biol 2006; 363:773-85. [PMID: 16979186 DOI: 10.1016/j.jmb.2006.05.077] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2006] [Revised: 05/26/2006] [Accepted: 05/30/2006] [Indexed: 11/27/2022]
Abstract
Poly(ADP-ribose)polymerases are involved in fundamental cellular events as well as they seem to be associated to some viral infection process. In this work, the poly(ADP-ribose)polymerase-1 (PARP-1) role in the polyomavirus life cycle has been investigated. Early viral transcription was reduced by competitive inhibitors of PARPs in Swiss 3T3 cells and almost abolished in PARP-1 knockout fibroblasts and in wild-type fibroblasts when PARP-1 was silenced by RNA interference. In vivo chromatin immunoprecipitation assays showed that poly(ADP-ribosyl)ation (poly(ADP-ribose)) facilitates the release of the capsid protein viral protein 1 (VP1) from the chromatin of infecting virions. In vitro experiments demonstrated that VP1 stimulates the enzymatic activity of PARP-1 and binds non-covalently both protein-free and PARP-1-bound poly(ADP-ribose). Our studies suggest that PARP-1 promotes the complete VP1 displacement from viral DNA favouring the viral early transcription.
Collapse
Affiliation(s)
- Mariarosaria Carbone
- Pasteur Institute-Fondazione Cenci Bolognetti, Department of Cellular Biotechnology and Hematology, University of Rome La Sapienza, Viale Regina Elena 324, 00161 Rome, Italy
| | | | | | | | | | | | | | | |
Collapse
|
121
|
Chromy LR, Oltman A, Estes PA, Garcea RL. Chaperone-mediated in vitro disassembly of polyoma- and papillomaviruses. J Virol 2006; 80:5086-91. [PMID: 16641302 PMCID: PMC1472060 DOI: 10.1128/jvi.80.10.5086-5091.2006] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Hsp70 chaperones play a role in polyoma- and papillomavirus assembly, as evidenced by their interaction in vivo with polyomavirus capsid proteins at late times after virus infection and by their ability to assemble viral capsomeres into capsids in vitro. We studied whether Hsp70 chaperones might also participate in the uncoating reaction. In vivo, Hsp70 co-immunoprecipitated with polyomavirus virion VP1 at 3 h after infection of mouse cells. In vitro, prokaryotic and eukaryotic Hsp70 chaperones efficiently disassembled polyoma- and papillomavirus-like particles and virions in energy-dependent reactions. These observations support a role for cell chaperones in the disassembly of these viruses.
Collapse
Affiliation(s)
- Laura R Chromy
- University of Colorado Health Sciences Center, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
122
|
Liebl D, Difato F, Horníková L, Mannová P, Stokrová J, Forstová J. Mouse polyomavirus enters early endosomes, requires their acidic pH for productive infection, and meets transferrin cargo in Rab11-positive endosomes. J Virol 2006; 80:4610-22. [PMID: 16611921 PMCID: PMC1472029 DOI: 10.1128/jvi.80.9.4610-4622.2006] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mouse polyomavirus (PyV) virions enter cells by internalization into smooth monopinocytic vesicles, which fuse under the cell membrane with larger endosomes. Caveolin-1 was detected on monopinocytic vesicles carrying PyV particles in mouse fibroblasts and epithelial cells (33). Here, we show that PyV can be efficiently internalized by Jurkat cells, which do not express caveolin-1 and lack caveolae, and that overexpression of a caveolin-1 dominant-negative mutant in mouse epithelial cells does not prevent their productive infection. Strong colocalization of VP1 with early endosome antigen 1 (EEA1) and of EEA1 with caveolin-1 in mouse fibroblasts and epithelial cells suggests that the monopinocytic vesicles carrying the virus (and vesicles containing caveolin-1) fuse with EEA1-positive early endosomes. In contrast to SV40, PyV infection is dependent on the acidic pH of endosomes. Bafilomycin A1 abolished PyV infection, and an increase in endosomal pH by NH4Cl markedly reduced its efficiency when drugs were applied during virion transport towards the cell nucleus. The block of acidification resulted in the retention of a fraction of virions in early endosomes. To monitor further trafficking of PyV, we used fluorescent resonance energy transfer (FRET) to determine mutual localization of PyV VP1 with transferrin and Rab11 GTPase at a 2- to 10-nm resolution. Positive FRET between PyV VP1 and transferrin cargo and between PyV VP1 and Rab11 suggests that during later times postinfection (1.5 to 3 h), the virus meets up with transferrin in the Rab11-positive recycling endosome. These results point to a convergence of the virus and the cargo internalized by different pathways in common transitional compartments.
Collapse
Affiliation(s)
- David Liebl
- Department of Genetics and Microbiology, Faculty of Medicine, Charles University in Prague, Vinicná 5, 128 44 Prague 2, Czech Republic
| | | | | | | | | | | |
Collapse
|
123
|
Brasset E, Taddei AR, Arnaud F, Faye B, Fausto AM, Mazzini M, Giorgi F, Vaury C. Viral particles of the endogenous retrovirus ZAM from Drosophila melanogaster use a pre-existing endosome/exosome pathway for transfer to the oocyte. Retrovirology 2006; 3:25. [PMID: 16684341 PMCID: PMC1524798 DOI: 10.1186/1742-4690-3-25] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Accepted: 05/09/2006] [Indexed: 12/27/2022] Open
Abstract
Background Retroviruses have evolved various mechanisms to optimize their transfer to new target cells via late endosomes. Here, we analyzed the transfer of ZAM, a retroelement from Drosophila melanogaster, from ovarian follicle cells to the oocyte at stage 9–10 of oogenesis, when an active yolk transfer is occurring between these two cell types. Results Combining genetic and microscopic approaches, we show that a functional secretory apparatus is required to tether ZAM to endosomal vesicles and to direct its transport to the apical side of follicle cells. There, ZAM egress requires an intact follicular epithelium communicating with the oocyte. When gap junctions are inhibited or yolk receptors mutated, ZAM particles fail to sort out the follicle cells. Conclusion Overall, our results indicate that retrotransposons do not exclusively perform intracellular replication cycles but may usurp exosomal/endosomal traffic to be routed from one cell to another.
Collapse
Affiliation(s)
- E Brasset
- INSERM, U384, Faculté de Médecine, BP38, 63001 Clermont-Ferrand, France
| | - AR Taddei
- Centre of Electron Microscopy, Department of Environmental Sciences, Tuscia, University Viterbo, Italy
| | - F Arnaud
- INSERM, U384, Faculté de Médecine, BP38, 63001 Clermont-Ferrand, France
| | - B Faye
- INSERM, U384, Faculté de Médecine, BP38, 63001 Clermont-Ferrand, France
| | - AM Fausto
- Centre of Electron Microscopy, Department of Environmental Sciences, Tuscia, University Viterbo, Italy
| | - M Mazzini
- Centre of Electron Microscopy, Department of Environmental Sciences, Tuscia, University Viterbo, Italy
| | - F Giorgi
- Centre of Electron Microscopy, Department of Environmental Sciences, Tuscia, University Viterbo, Italy
| | - C Vaury
- INSERM, U384, Faculté de Médecine, BP38, 63001 Clermont-Ferrand, France
| |
Collapse
|
124
|
Spooner RA, Smith DC, Easton AJ, Roberts LM, Lord JM. Retrograde transport pathways utilised by viruses and protein toxins. Virol J 2006; 3:26. [PMID: 16603059 PMCID: PMC1524934 DOI: 10.1186/1743-422x-3-26] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2005] [Accepted: 04/07/2006] [Indexed: 11/15/2022] Open
Abstract
A model has been presented for retrograde transport of certain toxins and viruses from the cell surface to the ER that suggests an obligatory interaction with a glycolipid receptor at the cell surface. Here we review studies on the ER trafficking cholera toxin, Shiga and Shiga-like toxins, Pseudomonas exotoxin A and ricin, and compare the retrograde routes followed by these protein toxins to those of the ER trafficking SV40 and polyoma viruses. We conclude that there is in fact no obligatory requirement for a glycolipid receptor, nor even with a protein receptor in a lipid-rich environment. Emerging data suggests instead that there is no common pathway utilised for retrograde transport by all of these pathogens, the choice of route being determined by the particular receptor utilised.
Collapse
Affiliation(s)
- Robert A Spooner
- Department of Biological Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Daniel C Smith
- Department of Biological Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Andrew J Easton
- Department of Biological Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Lynne M Roberts
- Department of Biological Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - J Michael Lord
- Department of Biological Sciences, University of Warwick, Coventry, CV4 7AL, UK
| |
Collapse
|
125
|
Low JA, Magnuson B, Tsai B, Imperiale MJ. Identification of gangliosides GD1b and GT1b as receptors for BK virus. J Virol 2006; 80:1361-6. [PMID: 16415013 PMCID: PMC1346969 DOI: 10.1128/jvi.80.3.1361-1366.2006] [Citation(s) in RCA: 147] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Gangliosides have been shown to be plasma membrane receptors for both murine polyomavirus and SV40, while JC virus uses serotonin receptors. In contrast, little is known of the membrane receptor and entry pathway for BK virus (BKV), which can cause severe disease in immunosuppressed bone marrow and renal transplant patients. Using sucrose flotation assays, we investigated BKV binding to and interaction with human erythrocyte membranes and determined that this interaction was dependent on a neuraminidase-sensitive, proteinase K-resistant molecule. BKV was found to interact with the gangliosides GT1b and GD1b. The terminal alpha2-8-linked disialic acid motif, present in both of these gangliosides, is likely to be important for this interaction. We also determined that the addition of GD1b and GT1b to LNCaP cells, which are normally resistant to BKV infection, made them susceptible to the virus. In addition, BKV interacted with membranes extracted from the endoplasmic reticulum (ER) and infection was blocked by the addition of brefeldin A, which interferes with transport from the ER to the Golgi apparatus. These data demonstrate that BKV uses the gangliosides GT1b and GD1b as receptors and passes through the ER on the way to the nucleus.
Collapse
Affiliation(s)
- Jonathan A Low
- Department of Microbiology and Immunology, University of Michigan Medical School, 1500 E. Medical Center Dr., 6304 Cancer Center, Ann Arbor, MI 48109-0942, USA
| | | | | | | |
Collapse
|
126
|
Magnuson B, Rainey EK, Benjamin T, Baryshev M, Mkrtchian S, Tsai B. ERp29 triggers a conformational change in polyomavirus to stimulate membrane binding. Mol Cell 2005; 20:289-300. [PMID: 16246730 DOI: 10.1016/j.molcel.2005.08.034] [Citation(s) in RCA: 138] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2005] [Revised: 07/28/2005] [Accepted: 08/31/2005] [Indexed: 11/20/2022]
Abstract
Membrane penetration of nonenveloped viruses is a poorly understood process. We have investigated early stages of this process by studying the conformational change experienced by polyomavirus (Py) in the lumen of the endoplasmic reticulum (ER), a step that precedes its transport into the cytosol. We show that a PDI-like protein, ERp29, exposes the C-terminal arm of Py's VP1 protein, leading to formation of a hydrophobic particle that binds to a lipid bilayer; this reaction likely mimics initiation of Py penetration across the ER membrane. Expression of a dominant-negative ERp29 decreases Py infection, indicating ERp29 facilitates viral infection. Interestingly, cholera toxin, another toxic agent that crosses the ER membrane into the cytosol, is unfolded by PDI in the ER. Our data thus identify an ER factor that mediates membrane penetration of a nonenveloped virus and suggest that PDI family members are generally involved in ER remodeling reactions.
Collapse
Affiliation(s)
- Brian Magnuson
- Department of Cell and Developmental Biology, University of Michigan Medical School, 4643 Medical Sciences II, 1335 East Catherine Street, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | |
Collapse
|
127
|
Affiliation(s)
- Akira Ono
- Virus-Cell Interaction Section, HIV Drug Resistance Program, National Cancer Institute at Frederick, National Institutes of Health, Maryland 21702, USA
| | | |
Collapse
|
128
|
Lencer WI, Saslowsky D. Raft trafficking of AB5 subunit bacterial toxins. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2005; 1746:314-21. [PMID: 16153723 DOI: 10.1016/j.bbamcr.2005.07.007] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/01/2005] [Revised: 07/15/2005] [Accepted: 07/19/2005] [Indexed: 02/07/2023]
Abstract
Cholera and the related AB(5)-subunit toxins co-opt plasma membrane (PM) glycolipids to move retrograde into the endoplasmic reticulum (ER) of the host cell where a portion of the toxin is retro-translocated to the cytosol to induce disease. Only glycolipids that associate strongly with detergent insoluble membrane microdomains can sort the toxins backwards from PM to ER. The way certain lipids and proteins are clustered in the plane of the membrane to form lipid rafts likely explains how the glycolipids can function as sorting motifs for the toxins.
Collapse
Affiliation(s)
- Wayne I Lencer
- GI Cell Biology, Enders 720, Children's Hospital Boston, the Harvard Digestive Diseases Center, and the Department of Pediatrics, Harvard Medical School, 300 Longwood Ave., Boston, MA 02115, USA.
| | | |
Collapse
|
129
|
Yanase K, Madaio MP. Nuclear localizing anti-DNA antibodies enter cells via caveoli and modulate expression of caveolin and p53. J Autoimmun 2005; 24:145-51. [PMID: 15829407 DOI: 10.1016/j.jaut.2004.11.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2004] [Revised: 09/24/2004] [Accepted: 11/15/2004] [Indexed: 11/19/2022]
Abstract
After administration to normal mice, a subset of monoclonal (m) anti-DNA antibodies (Ab) derived from MRL-lpr/lpr mice was identified that enter cells, in vivo. In the kidneys, this was associated with glomerular hypercellularity and proteinuria. In cultured cells, the same mAb bound to myosin 1 on the cell surface, prior to internalization, nuclear localization and inhibition of apoptosis. The present study focuses on the mechanisms underlying the observed functional effects. Subcellular localization studies revealed that following internalization, a prototypic, nuclear localizing, m antibody (Ab; termed H7) co-localized with myosin 1, shortly after internalization, within caveolae, near the cell membrane. Cell fractionation studies confirmed the presence of both H7 and myosin within the caveolar fraction. Since variations in caveolin protein expression have been associated with apoptotic events in cancer cells, through p53 dependent and independent pathways, modulation of caveolin by intracellular H7 was evaluated. Cellular entry of the anti-DNA Ab resulted in an increase in caveolin protein expression. Furthermore, after exposure of cells to dexamethasone to induce apoptosis, the usual increase in p53 was inhibited in the presence of intracellular H7. Taken together, the results suggest that upregulation of caveolin and inhibition of p53 induction are involved in H7-induced, inhibition of apoptosis. Furthermore, they suggest that this inhibition contributes to the glomerular hypercellularity observed in normal mice with intranuclear H7. The results also raise the possibility that inhibition of apoptotic pathways during inflammation or/and autoimmunity could influence subsequent disease events. The novel mechanism of cellular perturbation is indirect and dependent on apoptotic stimuli, and it may account for the presence of intranuclear antibodies in inflammatory and normal tissues of individuals with lupus.
Collapse
Affiliation(s)
- Kumiko Yanase
- Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104-6144, USA
| | | |
Collapse
|
130
|
Bossis I, Roden RBS, Gambhira R, Yang R, Tagaya M, Howley PM, Meneses PI. Interaction of tSNARE syntaxin 18 with the papillomavirus minor capsid protein mediates infection. J Virol 2005; 79:6723-31. [PMID: 15890910 PMCID: PMC1112158 DOI: 10.1128/jvi.79.11.6723-6731.2005] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The papillomavirus capsid mediates binding to the cell surface and passage of the virion to the perinuclear region during infection. To better understand how the virus traffics across the cell, we sought to identify cellular proteins that bind to the minor capsid protein L2. We have identified syntaxin 18 as a protein that interacts with bovine papillomavirus type 1 (BPV1) L2. Syntaxin 18 is a target membrane-associated soluble N-ethylmaleimide-sensitive factor-attachment protein receptor (tSNARE) that resides in the endoplasmic reticulum (ER). The ectopic expression of FLAG-tagged syntaxin 18, which disrupts ER trafficking, blocked BPV1 pseudovirion infection. Furthermore, the expression of FLAG-syntaxin 18 prevented the passage of BPV1 pseudovirions to the perinuclear region that is consistent with the ER. Genetic studies identified a highly conserved L2 domain, DKILK, comprising residues 40 to 44 that mediated BPV1 trafficking through the ER during infection via an interaction with the tSNARE syntaxin 18. Mutations within the DKILK motif of L2 that did not significantly impact virion morphogenesis or binding at the cell surface prevented the L2 interaction with syntaxin 18 and disrupted BPV1 infection.
Collapse
Affiliation(s)
- Ioannis Bossis
- Department of Pathology, Johns Hopkins University, Baltimore, Maryland 21205, USA
| | | | | | | | | | | | | |
Collapse
|
131
|
Colin M, Mailly L, Rogée S, D'Halluin JC. Efficient species C HAdV infectivity in plasmocytic cell lines using a clathrin-independent lipid raft/caveola endocytic route. Mol Ther 2005; 11:224-36. [PMID: 15668134 DOI: 10.1016/j.ymthe.2004.10.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2004] [Accepted: 10/05/2004] [Indexed: 10/26/2022] Open
Abstract
Hematopoietic cells are known to be refractory to species C human adenovirus (HAdV) infection; however, the reason for this has not been clearly established. We have previously demonstrated that this nonpermissivity is the consequence of inefficient HAdV particle uptake, notably in B lymphocytes. We noted that while the protein clathrin is observed in association with membranes in epithelial cells, it is found predominantly in the cytoplasm of hematopoietic cell lines. So it appears that altered clathrin-coated pit endocytosis could explain the weak HAdV uptake in B cells. In contrast, mature B cell plasmocytes are permissive to HAdV. However, this is not the result of clathrin-coated pit endocytosis since this process is also inefficient in these cells. Confocal microscopy showed colocalization between HAdV particles and caveolae/lipid rafts in plasmocytes. Moreover, inhibiting caveola endocytosis by depletion of cholesterol or expression of dominant negative caveolin-1 in these cells results in a 50-70% reduction in HAdV infectivity. It appears that caveola endocytosis and nonclathrin noncaveola endocytosis are used by HAdV to enter plasmocytes in response to a loss of the clathrin-dependent pathway. Thus targeting of caveolae by modifying the capsid of HAdV may represent an alternative approach to enhancing uptake in most hematopoietic cells for future gene therapy.
Collapse
Affiliation(s)
- Morvane Colin
- INSERM U524, IMPRT, Institut de Recherches sur le Cancer de Lille, 1 place de Verdun, 59045 Lille Cedex, France
| | | | | | | |
Collapse
|
132
|
Norkin LC, Kuksin D. The caveolae-mediated sv40 entry pathway bypasses the golgi complex en route to the endoplasmic reticulum. Virol J 2005; 2:38. [PMID: 15840166 PMCID: PMC1087894 DOI: 10.1186/1743-422x-2-38] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2005] [Accepted: 04/19/2005] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Simian virus 40 (SV40) enters cells via an atypical caveolae-mediated endocytic pathway, which delivers the virus to a new intermediary compartment, the caveosome. The virus then is believed to go directly from the caveosome to the endoplasmic reticulum. Cholera toxin likewise enters via caveolae and traffics to caveosomes. But, in contrast to SV40, cholera toxin is transported from caveosomes to the endoplasmic reticulum via the Golgi. For that reason, and because the caveosome and Golgi may have some common markers, we revisited the issue of whether SV40 might access the endoplasmic reticulum via the Golgi. RESULTS We confirmed our earlier finding that SV40 co localizes with the Golgi marker beta-COP. However, we show that the virus does not co localize with the more discriminating Golgi markers, golgin 97 and BODIPY-ceramide. CONCLUSION The caveolae-mediated SV40 entry pathway does not intersect the Golgi. SV40 is seen to co localize with beta-COP because that protein is a marker for caveosomes as well as the Golgi. Moreover, these results are consistent with the likelihood that the caveosome is a sorting organelle. In addition, there are at least two distinct but related routes by which a ligand might traffic from the caveosome to the ER; one route involving transport through the Golgi, and another pathway that does not involve the Golgi.
Collapse
Affiliation(s)
- Leonard C Norkin
- Department of Microbiology, University of Massachusetts – Amherst, MA 01003, USA
| | - Dmitry Kuksin
- Department of Microbiology, University of Massachusetts – Amherst, MA 01003, USA
| |
Collapse
|
133
|
Amer AO, Byrne BG, Swanson MS. Macrophages rapidly transfer pathogens from lipid raft vacuoles to autophagosomes. Autophagy 2005; 1:53-8. [PMID: 16874021 PMCID: PMC1584280 DOI: 10.4161/auto.1.1.1589] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Macrophages activate autophagy as an immediate response to Legionella pneumophila infection, but what marks the pathogen phagosome as a target for the autophagy machinery is not known. Because a variety of bacteria, parasites, viruses, and toxins that associate with the endoplasmic reticulum enter host cells by a cholesterol-dependent route, we tested the hypothesis that autophagy is triggered when microbes engage components of lipid raft domains. As the intracellular respiratory pathogen L. pneumophila or the extracellular uropathogen FimH(+) Escherichia coli entered macrophages by a cholesterol-sensitive mechanism, they immediatezly resided in vacuoles rich in glycosylphosphatidylinositol moieties and the autophagy enzyme Atg7. As expected for autophagosomes, the vacuoles sequentially acquired the endoplasmic reticulum protein BiP, the autophagy markers Atg8 and monodansyl-cadaverine, and the lysosomal protein LAMP-1. A robust macrophage response to the pathogens was cholesterol-dependent, since fewer Atg7-rich vacuoles were observed when macrophages were pretreated with methyl-beta-cyclodextrin or filipin. A model in which macrophages exploit autophagy to capture pathogens within the lipid raft pathway for antigen presentation prior to disposal in lysosomes is discussed.
Collapse
Affiliation(s)
| | | | - Michele S. Swanson
- *Correspondence to: M.S. Swanson; Department of Microbiology and Immunology; University of Michigan Medical School; 1150 West Medical Center Dr.; Ann Arbor, Michigan USA 48109-0620; Tel.: 734.647.7295; Fax: 734.764.3561;
| |
Collapse
|
134
|
Abstract
The conversion of exogenous and endogenous proteins into immunogenic peptides recognized by T lymphocytes involves a series of proteolytic and other enzymatic events culminating in the formation of peptides bound to MHC class I or class II molecules. Although the biochemistry of these events has been studied in detail, only in the past few years has similar information begun to emerge describing the cellular context in which these events take place. This review thus concentrates on the properties of antigen-presenting cells, especially those aspects of their overall organization, regulation, and intracellular transport that both facilitate and modulate the processing of protein antigens. Emphasis is placed on dendritic cells and the specializations that help account for their marked efficiency at antigen processing and presentation both in vitro and, importantly, in vivo. How dendritic cells handle antigens is likely to be as important a determinant of immunogenicity and tolerance as is the nature of the antigens themselves.
Collapse
Affiliation(s)
- E Sergio Trombetta
- Department of Cell Biology and Section of Immunobiology, Ludwig Institute for Cancer Research, Yale University School of Medicine, New Haven, Connecticut 06520-8002, USA.
| | | |
Collapse
|
135
|
Cordelier P, Kulkowsky JW, Ko C, Matskevitch AA, McKee HJ, Rossi JJ, Bouhamdan M, Pomerantz RJ, Kari G, Strayer DS. Protecting from R5-tropic HIV: individual and combined effectiveness of a hammerhead ribozyme and a single-chain Fv antibody that targets CCR5. Gene Ther 2005; 11:1627-37. [PMID: 15295615 DOI: 10.1038/sj.gt.3302329] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The CCR5 chemokine receptor is important for most clinical strains of HIV to establish infection. Individuals with naturally occurring polymorphisms in the CCR5 gene who have reduced or absent CCR5 are apparently otherwise healthy, but are resistant to HIV infection. With the goal of reducing CCR5 and protecting CCR5+ cells from R5-tropic HIV, we used Tag-deleted SV40-derived vectors to deliver several anti-CCR5 transgenes: 2C7, a single-chain Fv (SFv) antibody; VCKA1, a hammerhead ribozyme; and two natural CCR5 ligands, MIP-1alpha and MIP-1beta, modified to direct these chemokines, and hence their receptor to the endoplasmic reticulum. These transgenes were delivered using recombinant, Tag-deleted SV40-derived vectors to human CCR5+ cell lines and primary cells: monocyte-derived macrophages and brain microglia. All transgenes except MIP-1alpha decreased CCR5, as assayed by immunostaining, Northern blotting, and cytofluorimetry (FACS). Individually, all transgenes except MIP-1alpha protected from low challenge doses of HIV. At higher dose HIV challenges, protection provided by all transgenes diminished, the SFv and the ribozyme being most potent. Vectors carrying these two transgenes were used sequentially to deliver combination anti-CCR5 genetic therapy. This approach gave approximately additive reduction in CCR5, as measured by FACS and protected from higher dose HIV challenges. Reducing cell membrane CCR5 using anti-CCR5 transgenes, alone or in combinations, may therefore provide a degree of protection from R5-tropic strains of HIV.
Collapse
Affiliation(s)
- P Cordelier
- Department of Pathology, Jefferson Medical College, Philadelphia, PA 19107, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
136
|
Abstract
Virus entry is initiated by recognition by receptors present on the surface of host cells. Receptors can be major mediators of virus tropism, and in many cases receptor interactions occur in an apparently programmed series of events utilizing multiple receptors. After receptor interaction, both enveloped and nonenveloped viruses must deliver their genome across either the endosomal or plasma membrane for infection to proceed. Genome delivery occurs either by membrane fusion (in the case of enveloped viruses) or by pore formation or other means of permeabilizing the lipid bilayer (in the case of nonenveloped viruses). For those viruses that enter cells via endosomes, specific receptor interactions (and the signaling events that ensue) may control the particular route of endocytosis and/or the ultimate destination of the incoming virus particles. Our conception of virus entry is increasingly becoming more complex; however, the specificity involved in entry processes, once ascertained, may ultimately lead to the production of effective antiviral agents.
Collapse
Affiliation(s)
- S B Sieczkarski
- Department of Microbiology and Immunology, Cornell University, Ithaca, NY 14853, USA
| | | |
Collapse
|
137
|
Abstract
The pathway of entry of polyomavirus (Py) has been investigated with glycolipid-deficient C6 cells and added ganglioside GD1a as a specific virus receptor. Unsupplemented C6 cells show a low basal level of infection but become highly infectable by Py following preincubation with the sialic acid-containing ganglioside GD1a (38). Addition of GD1a has no effect on the overall level of virus binding but mediates the internalization and transit of virus to the endoplasmic reticulum (ER). This pathway of entry is cholesterol and caveola dependent and requires intact microtubules as well as a dynamic state of the microfilament system. In contrast to vesicular transport of other cargo via glycolipids, Py particles do not appear to pass through the Golgi apparatus. Colcemid and brefeldin A block transport of the virus to the ER in GD1a-supplemented cells and lead to accumulation of virus in a caveolin-1-containing environment. Several features distinguish the efficient GD1a-mediated pathway of virus uptake from the less-efficient pathway of basal infection in C6 cells.
Collapse
Affiliation(s)
- Joanna Gilbert
- Department of Pathology, Harvard Medical School, 77 Louis Pasteur Avenue, Boston, MA 02115, USA
| | | |
Collapse
|
138
|
Elphick GF, Querbes W, Jordan JA, Gee GV, Eash S, Manley K, Dugan A, Stanifer M, Bhatnagar A, Kroeze WK, Roth BL, Atwood WJ. The human polyomavirus, JCV, uses serotonin receptors to infect cells. Science 2004; 306:1380-3. [PMID: 15550673 DOI: 10.1126/science.1103492] [Citation(s) in RCA: 320] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The human polyomavirus, JCV, causes the fatal demyelinating disease progressive multifocal leukoencephalopathy in immunocompromised patients. We found that the serotonergic receptor 5HT2AR could act as the cellular receptor for JCV on human glial cells. The 5HT2A receptor antagonists inhibited JCV infection, and monoclonal antibodies directed at 5HT2A receptors blocked infection of glial cells by JCV, but not by SV40. Transfection of 5HT2A receptor-negative HeLa cells with a 5HT2A receptor rescued virus infection, and this infection was blocked by antibody to the 5HT2A receptor. A tagged 5HT2A receptor colocalized with labeled JCV in an endosomal compartment following internalization. Serotonin receptor antagonists may thus be useful in the treatment of progressive multifocal leukoencephalopathy.
Collapse
MESH Headings
- Antibodies, Monoclonal
- Cell Line, Transformed
- Dopamine Agonists/pharmacology
- Dopamine Antagonists/pharmacology
- Endosomes/metabolism
- Endosomes/virology
- Flow Cytometry
- Fluorescent Antibody Technique, Indirect
- HeLa Cells
- Humans
- JC Virus/physiology
- Microscopy, Confocal
- Neuroglia/physiology
- Neuroglia/virology
- Receptor, Serotonin, 5-HT2A/immunology
- Receptor, Serotonin, 5-HT2A/physiology
- Receptors, Dopamine/immunology
- Receptors, Dopamine/physiology
- Receptors, Virus/physiology
- Serotonin/pharmacology
- Serotonin 5-HT2 Receptor Antagonists
- Serotonin Antagonists/pharmacology
- Sialic Acids/physiology
- Transfection
Collapse
Affiliation(s)
- Gwendolyn F Elphick
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
139
|
Pietiäinen V, Marjomäki V, Upla P, Pelkmans L, Helenius A, Hyypiä T. Echovirus 1 endocytosis into caveosomes requires lipid rafts, dynamin II, and signaling events. Mol Biol Cell 2004; 15:4911-25. [PMID: 15356270 PMCID: PMC524743 DOI: 10.1091/mbc.e04-01-0070] [Citation(s) in RCA: 111] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2004] [Revised: 08/17/2004] [Accepted: 08/24/2004] [Indexed: 01/04/2023] Open
Abstract
Binding of echovirus 1 (EV1, a nonenveloped RNA virus) to the alpha2beta1 integrin on the cell surface is followed by endocytic internalization of the virus together with the receptor. Here, video-enhanced live microscopy revealed the rapid uptake of fluorescently labeled EV1 into mobile, intracellular structures, positive for green fluorescent protein-tagged caveolin-1. Partial colocalization of EV1 with SV40 (SV40) and cholera toxin, known to traffic via caveosomes, demonstrated that the vesicles were caveosomes. The initiation of EV1 infection was dependent on dynamin II, cholesterol, and protein phosphorylation events. Brefeldin A, a drug that prevents SV40 transport, blocked the EV1 infection cycle, whereas drugs that disrupt the cellular cytoskeleton had no effect. In situ hybridization revealed the localization of viral RNA with endocytosed viral capsid proteins in caveosomes before initiation of viral replication. Thus, both the internalization of EV1 to caveosomes and subsequent events differ clearly from caveolar endocytosis of SV40 because EV1 uptake is fast and independent of actin and EV1 is not sorted further to sER from caveosomes. These results shed further light on the cell entry of nonenveloped viral pathogens and illustrate the use of viruses as probes to dissect caveolin-associated endocytic pathways.
Collapse
Affiliation(s)
- Vilja Pietiäinen
- Department of Virology, Haartman Institute, University of Helsinki, FIN-00014 Helsinki, Finland.
| | | | | | | | | | | |
Collapse
|
140
|
Vera M, Prieto J, Strayer DS, Fortes P. Factors Influencing the Production of Recombinant SV40 Vectors. Mol Ther 2004; 10:780-91. [PMID: 15451462 DOI: 10.1016/j.ymthe.2004.06.1014] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2004] [Revised: 05/27/2004] [Accepted: 06/07/2004] [Indexed: 10/26/2022] Open
Abstract
Most gene therapy approaches employ viral vectors for gene delivery. Ideally, these vectors should be produced at high titer and purity with well-established protocols. Standardized methods to measure the quality of the vectors produced are imperative, as are techniques that allow reproducible quantitation of viral titer. We devised a series of protocols that achieve high-titer production and reproducible purification and provide for quality control and titering of recombinant simian virus 40 vectors (rSV40s). rSV40s are good candidate vehicles for gene transfer: they are easily modified to be nonreplicative and they are nonimmunogenic. Further, they infect a wide variety of cells and allow long-term transgene expression. We report here these protocols to produce rSV40 vectors in high yields, describe their purification, and characterize viral stocks using quality control techniques that monitor the presence of wild-type SV40 revertants and defective interfering particles. Several methods for reproducible titration of rSV40 viruses have been compared. We believe that these techniques can be widely applied to obtain high concentrations of high-quality rSV40 viruses reproducibly.
Collapse
Affiliation(s)
- Maria Vera
- Laboratory of Vector Development, Division of Gene Therapy, Foundation for Applied Medical Research, School of Medicine, University of Navarra, Irunlarrea 1, 31008 Pamplona, Spain
| | | | | | | |
Collapse
|
141
|
McFarland MJ, Porter AC, Rakhshan FR, Rawat DS, Gibbs RA, Barker EL. A Role for Caveolae/Lipid Rafts in the Uptake and Recycling of the Endogenous Cannabinoid Anandamide. J Biol Chem 2004; 279:41991-7. [PMID: 15292270 DOI: 10.1074/jbc.m407250200] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The mechanisms responsible for the uptake and cellular processing of the endogenous cannabinoid anandamide are not well understood. We propose that anandamide uptake may occur via a caveola/lipid raft-related endocytic process in RBL-2H3 cells. Inhibitors of caveola-related (clathrin-independent) endocytosis reduced anandamide transport by approximately 50% compared with the control. Fluorescein derived from fluorescently labeled anandamide colocalized with protein markers of caveolae at early time points following transport. In this study, we have also identified a yet unrecognized process involved in trafficking events affecting anandamide following its uptake. Following uptake of [(3)H]anandamide by RBL-2H3 cells, we found an accumulation of tritium in the caveolin-rich membranes. Inhibitors of both anandamide uptake and metabolism blocked the observed enrichment of tritium in the caveolin-rich membranes. Mass spectrometry of subcellular membrane fractions revealed that the tritium accumulation observed in the caveolin-rich membrane fraction was not representative of intact anandamide, suggesting that following metabolism by the enzyme fatty acid amide hydrolase (FAAH), anandamide metabolites are rapidly enriched in caveolae. Furthermore, HeLa cells, which do not express high levels of FAAH, showed an accumulation of tritium in the caveolin-rich membrane fraction only when transfected with FAAH cDNA. Western blot and immunocytochemistry analyses of RBL-2H3 cells revealed that FAAH was localized in intracellular compartments distinct from caveolin-1 localization. Together, these data suggest that following uptake via caveola/lipid raft-related endocytosis, anandamide is rapidly metabolized by FAAH, with the metabolites efficiently recycled to caveolin-rich membrane domains.
Collapse
Affiliation(s)
- Matthew J McFarland
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, Indiana 47907-2091, USA
| | | | | | | | | | | |
Collapse
|
142
|
Abstract
Although they were discovered more than 50 years ago, caveolae have remained enigmatic plasmalemmal organelles. With their characteristic “flasklike” shape and virtually ubiquitous tissue distribution, these interesting structures have been implicated in a wide range of cellular functions. Similar to clathrin-coated pits, caveolae function as macromolecular vesicular transporters, while their unique lipid composition classifies them as plasma membrane lipid rafts, structures enriched in a variety of signaling molecules. The caveolin proteins (caveolin-1, -2, and -3) serve as the structural components of caveolae, while also functioning as scaffolding proteins, capable of recruiting numerous signaling molecules to caveolae, as well as regulating their activity. That so many signaling molecules and signaling cascades are regulated by an interaction with the caveolins provides a paradigm by which numerous disease processes may be affected by ablation or mutation of these proteins. Indeed, studies in caveolin-deficient mice have implicated these structures in a host of human diseases, including diabetes, cancer, cardiovascular disease, atherosclerosis, pulmonary fibrosis, and a variety of degenerative muscular dystrophies. In this review, we provide an in depth summary regarding the mechanisms by which caveolae and caveolins participate in human disease processes.
Collapse
Affiliation(s)
- Alex W Cohen
- Dept. of Molecular Pharmacology and the Albert Einstein Cancer Center, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA
| | | | | | | |
Collapse
|
143
|
Yan M, Peng J, Jabbar IA, Liu X, Filgueira L, Frazer IH, Thomas R. Despite differences between dendritic cells and Langerhans cells in the mechanism of papillomavirus-like particle antigen uptake, both cells cross-prime T cells. Virology 2004; 324:297-310. [PMID: 15207617 DOI: 10.1016/j.virol.2004.03.045] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2003] [Revised: 12/11/2003] [Accepted: 03/24/2004] [Indexed: 10/26/2022]
Abstract
As human papillomavirus-like particles (HPV-VLP) represent a promising vaccine delivery vehicle, delineation of the interaction of VLP with professional APC should improve vaccine development. Differences in the capacity of VLP to signal dendritic cells (DC) and Langerhans cells (LC) have been demonstrated, and evidence has been presented for both clathrin-coated pits and proteoglycans (PG) in the uptake pathway of VLP into epithelial cells. Therefore, we compared HPV-VLP uptake mechanisms in human monocyte-derived DC and LC, and their ability to cross-present HPV VLP-associated antigen in the MHC class I pathway. DC and LC each took up virus-like particles (VLP). DC uptake of and signalling by VLP was inhibited by amiloride or cytochalasin D (CCD), but not by filipin treatment, and was blocked by several sulfated and non-sulfated polysaccharides and anti-CD16. In contrast, LC uptake was inhibited only by filipin, and VLP in LC were associated with caveolin, langerin, and CD1a. These data suggest fundamentally different routes of VLP uptake by DC and LC. Despite these differences, VLP taken up by DC and LC were each able to prime naive CD8(+) T cells and induce cytolytic effector T cells in vitro.
Collapse
Affiliation(s)
- Mengyong Yan
- Centre for Immunology and Cancer Research, Princess Alexandra Hospital, University of Queensland, Brisbane, QLD 4102, Australia
| | | | | | | | | | | | | |
Collapse
|
144
|
Manunta M, Tan PH, Sagoo P, Kashefi K, George AJT. Gene delivery by dendrimers operates via a cholesterol dependent pathway. Nucleic Acids Res 2004; 32:2730-9. [PMID: 15148360 PMCID: PMC419601 DOI: 10.1093/nar/gkh595] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Understanding the cellular uptake and intracellular trafficking of dendrimer-DNA complexes is an important prerequisite for improving the transfection efficiency of non-viral vector-mediated gene delivery. Dendrimers are synthetic polymers used for gene transfer. Although these cationic molecules show promise as versatile DNA carriers, very little is known about the mechanism of gene delivery. This paper investigates how the uptake occurs, using an endothelial cell line as model, and evaluates whether the internalization of dendriplexes takes place randomly on the cell surface or at preferential sites such as membrane rafts. Following extraction of plasma membrane cholesterol, the transfection efficiency of the gene delivered by dendrimers was drastically decreased. Replenishment of membrane cholesterol restored the gene expression. The binding and especially internalization of dendriplexes was strongly reduced by cholesterol depletion before transfection. However, cholesterol removal after transfection did not inhibit expression of the delivered gene. Fluorescent dendriplexes co-localize with the ganglioside GM1 present into membrane rafts in both an immunoprecipitation assay and confocal microscopy studies. These data strongly suggest that membrane cholesterol and raft integrity are physiologically relevant for the cellular uptake of dendrimer-DNA complexes. Hence these findings provide evidence that membrane rafts are important for the internalization of non-viral vectors in gene therapy.
Collapse
Affiliation(s)
- Maria Manunta
- Department of Immunology, Division of Medicine, Faculty of Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | | | | | | | | |
Collapse
|
145
|
Abstract
Simian virus-40 (SV40), an icosahedral papovavirus, has recently been modified to serve as a gene delivery vector. Recombinant SV40 vectors (rSV40) are good candidates for gene transfer, as they display some unique features: SV40 is a well-known virus, nonreplicative vectors are easy-to-make, and can be produced in titers of 10(12) IU/ml. They also efficiently transduce both resting and dividing cells, deliver persistent transgene expression to a wide range of cell types, and are nonimmunogenic. Present disadvantages of rSV40 vectors for gene therapy are a small cloning capacity and the possible risks related to random integration of the viral genome into the host genome. Considerable efforts have been devoted to modifing this virus and setting up protocols for viral production. Preliminary therapeutic results obtained both in tissue culture cells and in animal models for heritable and acquired diseases indicate that rSV40 vectors are promising gene transfer vehicles. This article reviews the work performed with SV40 viruses as recombinant vectors for gene transfer. A summary of the structure, genomic organization, and life cycle of wild-type SV40 viruses is presented. Furthermore, the strategies utilized for the development, production, and titering of rSV40 vectors are discussed. Last, the therapeutic applications developed to date are highlighted.
Collapse
Affiliation(s)
- Maria Vera
- School of Medicine, Foundation for Applied Medical Research, Division of Gene Therapy, Laboratory of Vectors Development, University of Navarra, Pamplona, Spain
| | | |
Collapse
|
146
|
Abstract
Our view of biological membranes has evolved dramatically over the last few decades. In the bilayer model from Singer & Nicholson (Science 1972;175:720-731), both proteins and lipids freely diffuse within the plane of the membrane. Currently, however, membranes are viewed as a mosaic of different compartments or domains maintained by an active cytoskeleton network (Ritchie et al. Mol Membr Biol 2003; 20:13-18). Due to interactions between membrane components, several types of subdomains can form with different characteristics and functions. Lipids are likely to play an important role in the formation of so-called lipid-enriched microdomains or lipid rafts, adding another order of complexity to the membrane model. Rafts represent a type of domain wherein lipids of specific chemistry may dynamically associate with each other, to form platforms important for membrane protein sorting and construction of signaling complexes (Simons & Toomre. Nat Rev Mol Cell Biol 2000;1:31-39). Currently, there are several hypotheses concerning the nature of rafts (reviewed in (Edidin. Annu Rev Biophys Biomol Struct 2003;32: 257-283; Zurzolo et al. EMBO Rep 2003;4:1117-1121)). The most commonly cited one, proposed by Kai Simons (Simons & Ikonen. Nature 1997;387:569-572; Pralle et al. J Cell Biol 2000;148:997-1008), suggests that rafts are relatively small structures ( approximately 50 nm) enriched in cholesterol and sphingolipids within which associated proteins are likely to be concentrated. Another proposal (Anderson & Jacobson. Science 2002;296:1821-1825) suggests that rafts are constructed of lipid shells. These are small dynamic assemblies wherein 'raft' proteins are preferentially associated with certain types of lipids. These 'shells' are thermodynamically stable mobile entities in the plane of the membrane that are able to target the protein they encase to preexisting rafts/caveolae domains. In this review we summarize the data suggesting a specific role for lipid domains in intracellular trafficking and sorting and present a modification of the raft model that may help explain the observed phenomena.
Collapse
Affiliation(s)
- J Bernd Helms
- Department Biochemistry & Cell Biology, Utrecht University, PO Box 80176, 3508 TD Utrecht, the Netherlands.
| | | |
Collapse
|
147
|
Arad U, Axelrod J, Ben-nun-Shaul O, Oppenheim A, Galun E. Hepatitis B virus enhances transduction of human hepatocytes by SV40-based vectors. J Hepatol 2004; 40:520-6. [PMID: 15123369 DOI: 10.1016/j.jhep.2003.11.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2003] [Revised: 11/23/2003] [Accepted: 11/25/2003] [Indexed: 12/04/2022]
Abstract
BACKGROUND/AIMS Chronic HBV infection, a world-wide epidemic, can lead to chronic hepatitis and eventually to cirrhosis and hepatocellular carcinoma. The liver poses obstacles for many available gene-transfer vectors. SV40-based vectors can transduce human hepatic and hematopoietic cells. We studied the effect of HBV on the transduction - efficiency of human hepatic cells by SV40 - based vectors. METHODS A SV40-vector carrying the luciferase gene, and wild-type SV40, were used to assess transduction efficiency of human HBV-positive and HBV-negative hepatic cells. Transduction efficiency was measured as luciferase activity or by T-antigen staining. To evaluate whether differences in transduction efficiency are due to cell recognition and/or nuclear transport, MHC-I receptors were measured by FACS analysis and SV40-DNA was extracted from the nuclei of transduced cells and quantified. RESULTS Two HBV-positive cell-lines, HepG2.2.2.15 and FLC4-A10II, were transduced significantly more efficiently than their parental HBV-negative cell-lines. Transient transfection of HuH-7 cells with the HBV genome also increased transduction efficiency. The level of MHC-I, the cellular receptor for SV40, was comparable in all the cell-lines studied. However, soon after infection with SV40, the nuclei of HepG2.2.2.15 contained >6-fold more SV40-DNA than HepG2. CONCLUSIONS HBV increases transduction by SV40-vectors. This is due to enhanced vector entry and/or transport into the nucleus. SV40-vectors appear to have a potential for gene therapy for the treatment of HBV infections.
Collapse
Affiliation(s)
- Uri Arad
- Department of Hematology and Goldyne Savad Gene Therapy Institute, The Hebrew University-Hadassah Medical School and Hadassah University Hospital, Jerusalem, Israel
| | | | | | | | | |
Collapse
|
148
|
Abstract
The biosynthesis of secretory and membrane proteins in the endoplasmic reticulum (ER) yields mostly properly folded and assembled structures with full biological activity. Such fidelity is maintained by quality control (QC) mechanisms that avoid the production of nonnative structures. QC relies on chaperone systems in the ER that monitor and assist in the folding process. When folding promotion is not sufficient, proteins are retained in the ER and eventually retranslocated to the cytosol for degradation by the ubiquitin proteasome pathway. Retention of proteins that fail QC can sometimes occur beyond the ER, and degradation can take place in lysosomes. Several diseases are associated with proteins that do not pass QC, fail to be degraded efficiently, and accumulate as aggregates. In other cases, pathology arises from the downregulation of mutated but potentially functional proteins that are retained and degraded by the QC system.
Collapse
Affiliation(s)
- E Sergio Trombetta
- Department of Cell Biology, Yale University School of Medicine, PO Box 208002, New Haven, Connecticut 06520-8002, USA.
| | | |
Collapse
|
149
|
Drachenberg CB, Papadimitriou JC, Wali R, Cubitt CL, Ramos E. BK polyoma virus allograft nephropathy: ultrastructural features from viral cell entry to lysis. Am J Transplant 2003; 3:1383-92. [PMID: 14525599 DOI: 10.1046/j.1600-6135.2003.00237.x] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
BK virions must enter the host cell and target their genome to the nucleus in order to complete their life cycle. The mechanisms by which the virions accomplish these tasks are not known. In this morphological study we found that BK virions localized beneath the host cell cytoplasmic membrane in 60-70-nm, smooth (non-coated) monopinocytotic vesicles similar to, or consistent with, caveolae. In the cytoplasm, the monopinocytotic vesicles carrying virions appeared to fuse with a system of smooth, vesicles and tubules that communicated with the rough endoplasmic reticulum and was continuous with the Golgi system. Membrane-bound single virions and large tubulo-reticular complexes loaded with virions accumulated in paranuclear locations. Occasional nuclei displayed virions within the perinuclear cisterna in association to the perinuclear viral accumulations. Tubular cells with mature productive infection had large nuclei, distended by daughter virions, whereas they lacked significant numbers of cytoplasmic virions. In addition to virally induced cell necrosis, there was extensive tubular cell damage (apoptosis and necrosis) in morphologically non-infected tubules. The observed ultrastructural interactions between the BK virions and host cells are remarkably similar to viral cell entry and nuclear targeting described for SV40 virus.
Collapse
Affiliation(s)
- Cinthia B Drachenberg
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA
| | | | | | | | | |
Collapse
|
150
|
Abstract
Clathrin-coated pits and caveolae are two of the most recognizable features of the plasma membrane of mammalian cells. While our understanding of the machinery regulating and driving clathrin-coated pit-mediated endocytosis has progressed dramatically, including the elucidation of the structure of individual components and partial in vitro reconstitution, the role of caveolae as alternative endocytic carriers still remains elusive 50 years after their discovery. However, recent work has started to provide new insights into endocytosis by caveolae and into apparently related pathways involving lipid raft domains. These pathways, distinguished by their exquisite sensitivity to cholesterol-sequestering agents, can involve caveolae but also exist in cells devoid of caveolins and caveolae. This review examines the current evidence for the involvement of rafts and caveolae in endocytosis and the molecular players involved in their regulation.
Collapse
Affiliation(s)
- Robert G Parton
- Institute for Molecular Bioscience, Centre for Microscopy and Microanalysis, and School of Biomedical Sciences, University of Queensland, Queensland, 4072, Australia.
| | | |
Collapse
|