The Potential Contribution of Caveolin 1 to HIV Latent Infection
Abstract
:1. Introduction
2. Caveolin-1
3. Potential of Cav-1 in Keeping the Chromatin Open
4. Caveolin-1 and the Potential Link to HIV Latent Infection
5. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- Rerks-Ngarm, S.; Pitisuttithum, P.; Nitayaphan, S.; Kaewkungwal, J.; Chiu, J.; Paris, R.; Premsri, N.; Namwat, C.; De Souza, M.; Adams, E.; et al. Vaccination with ALVAC and AIDSVAX to Prevent HIV-1 Infection in Thailand. N. Engl. J. Med. 2009, 361, 2209–2220. [Google Scholar] [CrossRef] [PubMed]
- Blankson, J.N.; Persaud, D.; Siliciano, R.F. The Challenge of Viral Reservoirs in HIV-1 Infection. Annu. Rev. Med. 2002, 53, 557–593. [Google Scholar] [CrossRef] [PubMed]
- Finzi, D.; Blankson, J.N.; Siliciano, J.D.; Margolick, J.B.; Chadwick, K.; Pierson, T.C.; A Smith, K.; Lisziewicz, J.; Lori, F.; Flexner, C.; et al. Latent infection of CD4+ T cells provides a mechanism for lifelong persistence of HIV-1, even in patients on effective combination therapy. Nat. Med. 1999, 5, 512–517. [Google Scholar] [CrossRef] [PubMed]
- Palmer, S.; Josefsson, L.; Coffin, J.M. HIV reservoirs and the possibility of a cure for HIV infection. J. Intern. Med. 2011, 270, 550–560. [Google Scholar] [CrossRef] [PubMed]
- Smith, M.Z.; Wightman, F.; Lewin, S.R. HIV Reservoirs and Strategies for Eradication. Curr. HIV/AIDS Rep. 2012, 9, 5–15. [Google Scholar] [CrossRef] [PubMed]
- Chomont, N.; El-Far, M.; Ancuta, P.; Trautmann, L.; A Procopio, F.; Yassine-Diab, B.; Boucher, G.; Boulassel, M.-R.; Ghattas, G.; Brenchley, J.M.; et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat. Med. 2009, 15, 893–900. [Google Scholar] [CrossRef] [PubMed]
- Chun, T.-W.; Carruth, L.; Finzi, D.; Shen, X.; DiGiuseppe, J.A.; Taylor, H.; Hermankova, M.; Chadwick, K.; Margolick, J.; Quinn, T.C.; et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nat. Cell Biol. 1997, 387, 183–188. [Google Scholar] [CrossRef]
- Carter, C.C.; Onafuwa-Nuga, A.; McNamara, L.A.; Riddell, J.; Bixby, D.; Savona, M.R.; Collins, K.L. HIV-1 infects multipotent progenitor cells causing cell death and establishing latent cellular reservoirs. Nat. Med. 2010, 16, 446–451. [Google Scholar] [CrossRef] [Green Version]
- Carter, C.C.; McNamara, L.A.; Onafuwa-Nuga, A.; Shackleton, M.; Riddell, J.; Bixby, D.; Savona, M.R.; Morrison, S.J.; Collins, K.L. HIV-1 Utilizes the CXCR4 Chemokine Receptor to Infect Multipotent Hematopoietic Stem and Progenitor Cells. Cell Host Microbe 2011, 9, 223–234. [Google Scholar] [CrossRef] [Green Version]
- Bailey, J.R.; Sedaghat, A.R.; Kieffer, T.; Brennan, T.; Lee, P.K.; Wind-Rotolo, M.; Haggerty, C.M.; Kamireddi, A.R.; Liu, Y.; Lee, J.; et al. Residual Human Immunodeficiency Virus Type 1 Viremia in Some Patients on Antiretroviral Therapy Is Dominated by a Small Number of Invariant Clones Rarely Found in Circulating CD4+ T Cells. J. Virol. 2006, 80, 6441–6457. [Google Scholar] [CrossRef] [Green Version]
- Keele, B.F.; Tazi, L.; Gartner, S.; Liu, Y.; Burgon, T.B.; Estes, J.D.; Thacker, T.C.; Crandall, K.A.; McArthur, J.C.; Burton, G.F. Characterization of the Follicular Dendritic Cell Reservoir of Human Immunodeficiency Virus Type 1. J. Virol. 2008, 82, 5548–5561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhu, T.; Muthui, D.; Holte, S.; Nickle, D.; Feng, F.; Brodie, S.; Hwangbo, Y.; Mullins, J.I.; Corey, L. Evidence for Human Immunodeficiency Virus Type 1 Replication In Vivo in CD14+ Monocytes and Its Potential Role as a Source of Virus in Patients on Highly Active Antiretroviral Therapy. J. Virol. 2002, 76, 707–716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Liu, Y.; Wigdahl, B. Cellular Reservoirs of HIV-1 and their Role in Viral Persistence. Curr. HIV Res. 2008, 6, 388–400. [Google Scholar] [CrossRef] [Green Version]
- Alexaki, A.; Wigdahl, B. HIV-1 Infection of Bone Marrow Hematopoietic Progenitor Cells and Their Role in Trafficking and Viral Dissemination. PLoS Pathog. 2008, 4, e1000215. [Google Scholar] [CrossRef]
- Coleman, C.M.; Wu, L. HIV interactions with monocytes and dendritic cells: Viral latency and reservoirs. Retrovirology 2009, 6, 51. [Google Scholar] [CrossRef] [Green Version]
- Churchill, M.J.; Wesselingh, S.L.; Cowley, D.; Pardo, C.A.; McArthur, J.C.; Brew, B.J.; Gorry, P.R. Extensive astrocyte infection is prominent in human immunodeficiency virus-associated dementia. Ann. Neurol. 2009, 66, 253–258. [Google Scholar] [CrossRef] [PubMed]
- Fischer-Smith, T.; Croul, S.; Sverstiuk, A.E.; Capini, C.; L’Heureux, D.; Regulier, E.G.; Richardson, M.W.; Amini, S.; Morgello, S.; Khalili, K.; et al. CNS invasion by CD14+/CD16+ peripheral blood-derived monocytes in HIV dementia: Perivascular accumulation and reservoir of HIV infection. J. Neurovirol. 2001, 77, 528–541. [Google Scholar] [CrossRef]
- Edén, A.; Fuchs, D.; Hagberg, L.; Nilsson, S.; Spudich, S.; Svennerholm, B.; Price, R.W.; Gisslén, M. HIV-1 Viral Escape in Cerebrospinal Fluid of Subjects on Suppressive Antiretroviral Treatment. J. Infect. Dis. 2010, 202, 1819–1825. [Google Scholar] [CrossRef]
- Chun, T.; Nickle, D.C.; Justement, J.S.; Meyers, J.H.; Roby, G.; Hallahan, C.W.; Kottilil, S.; Moir, S.; Mican, J.M.; Mullins, J.I.; et al. Persistence of HIV in Gut-Associated Lymphoid Tissue despite Long-Term Antiretroviral Therapy. J. Infect. Dis. 2008, 197, 714–720. [Google Scholar] [CrossRef] [Green Version]
- Cu-Uvin, S.; Delong, A.K.; Venkatesh, K.K.; Hogan, J.W.; Ingersoll, J.; Kurpewski, J.; De Pasquale, M.P.; D’aquila, R.; Caliendo, A.M. Genital tract HIV-1 RNA shedding among women with below detectable plasma viral load. AIDS 2010, 24, 2489–2497. [Google Scholar] [CrossRef]
- Thacker, T.C.; Zhou, X.; Estes, J.D.; Jiang, Y.; Keele, B.F.; Elton, T.S.; Burton, G.F. Follicular Dendritic Cells and Human Immunodeficiency Virus Type 1 Transcription in CD4+ T Cells. J. Virol. 2008, 83, 150–158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Van Leeuwen, E.; Ter Heine, R.; Van Der Veen, F.; Repping, S.; Beijnen, J.H.; Prins, J.M. Penetration of Atazanavir in Seminal Plasma of Men Infected with Human Immunodeficiency Virus Type 1. Antimicrob. Agents Chemother. 2006, 51, 335–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yukl, S.A.; Gianella, S.; Sinclair, E.; Epling, L.; Li, Q.; Duan, L.; Choi, A.L.M.; Girling, V.; Ho, T.; Li, P.; et al. Differences in HIV Burden and Immune Activation within the Gut of HIV-Positive Patients Receiving Suppressive Antiretroviral Therapy. J. Infect. Dis. 2010, 202, 1553–1561. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yukl, S.A.; Shergill, A.K.; McQuaid, K.; Gianella, S.; Lampiris, H.; Hare, C.B.; Pandori, M.; Sinclair, E.; Günthard, H.F.; Fischer, M.; et al. Effect of raltegravir-containing intensification on HIV burden and T-cell activation in multiple gut sites of HIV-positive adults on suppressive antiretroviral therapy. AIDS 2010, 24, 2451–2460. [Google Scholar] [CrossRef]
- Luetkemeyer, A.F.; Havlir, D.V.; Currier, J.S. Complications of HIV disease and antiretroviral treatment. Top. HIV Med. Publ. Int. AIDS Soc. USA 2010, 18, 57–65. [Google Scholar]
- Gomo, Z.A.; Hakim, J.; Walker, A.S.; Tinago, W.; Mandozana, G.; Kityo, C.M.; Munderi, P.; Katabira, E.; Reid, A.; Gibb, D.M.; et al. Impact of second-line antiretroviral regimens on lipid profiles in an African setting: The DART trial sub-study. AIDS Res. Ther. 2014, 11, 32. [Google Scholar] [CrossRef] [Green Version]
- Matoga, M.; Hosseinipour, M.C.; Aga, E.; Ribaudo, H.J.; Kumarasamy, N.; Bartlett, J.; Hughes, M.D.; the ACTG A5230 Study Team. Hyperlipidaemia in HIV-infected patients on lopinavir/ritonavir monotherapy in resource-limited settings. Antivir. Ther. 2016, 22, 205–213. [Google Scholar] [CrossRef] [Green Version]
- Coiras, M.; López-Huertas, M.R.; Pérez-Olmeda, M.; Alcamí, J. Understanding HIV-1 latency provides clues for the eradication of long-term reservoirs. Nat. Rev. Genet. 2009, 7, 798–812. [Google Scholar] [CrossRef]
- Hakre, S.; Chavez, L.; Shirakawa, K.; Verdin, E. Epigenetic regulation of HIV latency. Curr. Opin. HIV AIDS 2011, 6, 19–24. [Google Scholar] [CrossRef]
- Archin, N.M.; Sung, J.M.; Garrido, C.; Soriano-Sarabia, N.; Margolis, D.M. Eradicating HIV-1 infection: Seeking to clear a persistent pathogen. Nat. Rev. Genet. 2014, 12, 750–764. [Google Scholar] [CrossRef] [Green Version]
- Taube, R.; Peterlin, B.M. Lost in Transcription: Molecular Mechanisms that Control HIV Latency. Viruses 2013, 5, 902–927. [Google Scholar] [CrossRef] [Green Version]
- Mbonye, U.; Karn, J. Transcriptional control of HIV latency: Cellular signaling pathways, epigenetics, happenstance and the hope for a cure. Virology 2014, 455, 328–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castro, S.; Colomer-Lluch, M.; Serra-Moreno, R. Barriers for HIV Cure: The Latent Reservoir. AIDS Res. Hum. Retroviruses 2018, 34. [Google Scholar] [CrossRef]
- Archin, N.M.; Liberty, A.L.; Kashuba, A.D.; Choudhary, S.K.; Kuruc, J.D.; Crooks, A.M.; Parker, D.C.; Anderson, E.M.; Kearney, M.F.; Strain, M.C.; et al. Administration of vorinostat disrupts HIV-1 latency in patients on antiretroviral therapy. Nature 2012, 487, 482–485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shan, L.; Deng, K.; Shroff, N.S.; Durand, C.M.; Rabi, S.A.; Yang, H.-C.; Zhang, H.; Margolick, J.B.; Blankson, J.N.; Siliciano, R.F. Stimulation of HIV-1-Specific Cytolytic T Lymphocytes Facilitates Elimination of Latent Viral Reservoir after Virus Reactivation. Immunity 2012, 36, 491–501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spivak, A.M.; Planelles, V. HIV-1 Eradication: Early Trials (and Tribulations). Trends Mol. Med. 2016, 22, 10–27. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Spivak, A.M.; Planelles, V. Novel Latency Reversal Agents for HIV-1 Cure. Annu. Rev. Med. 2018, 69, 421–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiang, G.; Dandekar, S. Targeting NF-kappaB signaling with protein kinase C agonists as an emerging strategy for combating HIV latency. Aids Res. Hum. Retrovir. 2015, 31, 4–12. [Google Scholar] [CrossRef]
- Spina, C.A.; Anderson, J.; Archin, N.M.; Bosque, A.; Chan, J.; Famiglietti, M.; Greene, W.C.; Kashuba, A.; Lewin, S.R.; Margolis, D.M.; et al. An In-Depth Comparison of Latent HIV-1 Reactivation in Multiple Cell Model Systems and Resting CD4+ T Cells from Aviremic Patients. PLoS Pathog. 2013, 9, e1003834. [Google Scholar] [CrossRef] [Green Version]
- Bullen, C.K.; Laird, G.M.; Durand, C.M.; Siliciano, J.D.; Siliciano, R.F. New ex vivo approaches distinguish effective and ineffective single agents for reversing HIV-1 latency in vivo. Nat. Med. 2014, 20, 425–429. [Google Scholar] [CrossRef] [Green Version]
- Laird, G.M.; Bullen, C.K.; Rosenbloom, D.I.; Martin, A.R.; Hill, A.L.; Durand, C.M.; Siliciano, J.D.; Siliciano, R.F. Ex vivo analysis identifies effective HIV-1 latency–reversing drug combinations. J. Clin. Investig. 2015, 125, 1901–1912. [Google Scholar] [CrossRef]
- Cary, D.C.; Fujinaga, K.; Peterlin, B.M. Euphorbia Kansui Reactivates Latent HIV. PLoS ONE 2016, 11, e0168027. [Google Scholar] [CrossRef] [Green Version]
- Martin, A.R.; Pollack, R.A.; Capoferri, A.; Ambinder, R.F.; Durand, C.M.; Siliciano, R.F. Rapamycin-mediated mTOR inhibition uncouples HIV-1 latency reversal from cytokine-associated toxicity. J. Clin. Investig. 2017, 127, 651–656. [Google Scholar] [CrossRef] [Green Version]
- Van Lint, C.; Emiliani, S.; Ott, M.; Verdin, E. Transcriptional activation and chromatin remodeling of the HIV-1 promoter in response to histone acetylation. EMBO J. 1996, 15, 1112–1120. [Google Scholar] [CrossRef] [PubMed]
- Archin, N.M.; Espeseth, A.; Parker, D.; Cheema, M.; Hazuda, D.; Margolis, D.M. Expression of Latent HIV Induced by the Potent HDAC Inhibitor Suberoylanilide Hydroxamic Acid. AIDS Res. Hum. Retroviruses 2009, 25, 207–212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Contreras, X.; Schweneker, M.; Chen, C.-S.; McCune, J.M.; Deeks, S.G.; Martin, J.; Peterlin, B.M. Suberoylanilide Hydroxamic Acid Reactivates HIV from Latently Infected Cells. J. Biol. Chem. 2009, 284, 6782–6789. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ke, R.; Lewin, S.R.; Elliott, J.H.; Perelson, A.S. Modeling the Effects of Vorinostat In Vivo Reveals both Transient and Delayed HIV Transcriptional Activation and Minimal Killing of Latently Infected Cells. PLoS Pathog. 2015, 11, e1005237. [Google Scholar] [CrossRef] [PubMed]
- Olesen, R.; Viganò, S.; Rasmussen, T.A.; Søgaard, O.S.; Ouyang, Z.; Buzon, M.J.; Bashirova, A.; Carrington, M.; Palmer, S.; Brinkmann, C.R.; et al. Innate Immune Activity Correlates with CD4 T Cell-Associated HIV-1 DNA Decline during Latency-Reversing Treatment with Panobinostat. J. Virol. 2015, 89, 10176–10189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Elliott, J.H.; Wightman, F.; Solomon, A.; Ghneim, K.; Ahlers, J.; Cameron, M.J.; Smith, M.Z.; Spelman, T.; McMahon, J.; Velayudham, P.; et al. Activation of HIV Transcription with Short-Course Vorinostat in HIV-Infected Patients on Suppressive Antiretroviral Therapy. PLoS Pathog. 2014, 10, e1004473. [Google Scholar] [CrossRef] [Green Version]
- Rasmussen, T.A.; Tolstrup, M.; Brinkmann, C.R.; Olesen, R.; Erikstrup, C.; Solomon, A.; Winckelmann, A.; Palmer, S.; Dinarello, C.; Buzon, M.J.; et al. Panobinostat, a histone deacetylase inhibitor, for latent-virus reactivation in HIV-infected patients on suppressive antiretroviral therapy: A phase 1/2, single group, clinical trial. Lancet HIV 2014, 1, e13–e21. [Google Scholar] [CrossRef]
- Søgaard, O.S.; Graversen, M.E.; Leth, S.; Olesen, R.; Brinkmann, C.R.; Nissen, S.K.; Kjaer, A.S.; Schleimann, M.H.; Denton, P.W.; Hey-Cunningham, W.J.; et al. The Depsipeptide Romidepsin Reverses HIV-1 Latency In Vivo. PLoS Pathog. 2015, 11, e1005142. [Google Scholar] [CrossRef] [Green Version]
- Xing, S.; Bullen, C.K.; Shroff, N.S.; Shan, L.; Yang, H.-C.; Manucci, J.L.; Bhat, S.; Zhang, H.; Margolick, J.B.; Quinn, T.C.; et al. Disulfiram Reactivates Latent HIV-1 in a Bcl-2-Transduced Primary CD4+ T Cell Model without Inducing Global T Cell Activation. J. Virol. 2011, 85, 6060–6064. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Doyon, G.; Zerbato, J.; Mellors, J.W.; Sluis-Cremer, N. Disulfiram reactivates latent HIV-1 expression through depletion of the phosphatase and tensin homolog. AIDS 2013, 27, F7–F11. [Google Scholar] [CrossRef]
- Elliott, J.H.; McMahon, J.H.; Chang, C.C.; Lee, S.A.; Hartogensis, W.; Bumpus, N.N.; Savic, R.; Roney, J.; Hoh, R.; Solomon, A.; et al. Short-term administration of disulfiram for reversal of latent HIV infection: A phase 2 dose-escalation study. Lancet HIV 2015, 2, e520–e529. [Google Scholar] [CrossRef] [Green Version]
- Spivak, A.M.; Andrade, A.; Eisele, E.; Hoh, R.; Bacchetti, P.; Bumpus, N.N.; Emad, F.; Buckheit, R., III; McCance-Katz, E.F.; Lai, J.; et al. A pilot study assessing the safety and latency-reversing activity of disulfiram in HIV-1-infected adults on antiretroviral therapy. Clin. Infect. Dis. 2014, 58, 883–890. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Madrid-Elena, N.; Garcia-Bermejo, M.L.; Serrano-Villar, S.; Diaz-de Santiago, A.; Sastre, B.; Gutierrez, C.; Dronda, C.; Díaz, M.C.; Domínguez, E.; López-Huertas, M.R.; et al. Maraviroc is associated with latent HIV-1 reactivation through NF-kappaB activation in resting CD4(+) T cells from HIV-Infected Individuals on Suppressive Antiretroviral Therapy. J. Virol. 2018, 92, e01931-17. [Google Scholar] [CrossRef] [Green Version]
- Beans, E.J.; Fournogerakis, D.; Gauntlett, C.; Heumann, L.V.; Kramer, R.; Marsden, M.D.; Murray, D.; Chun, T.-W.; Zack, J.A.; Wender, P.A. Highly potent, synthetically accessible prostratin analogs induce latent HIV expression in vitro and ex vivo. Proc. Natl. Acad. Sci. USA 2013, 110, 11698–11703. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muñoz, E.; Vo, M.Q.; Perez, M.; Calzado, M.A.; Moreno, S.; Appendino, G.; Munoz, E. Activation of Latent HIV-1 Expression by Protein Kinase C Agonists. A Novel Therapeutic Approach to Eradicate HIV-1 Reservoirs. Curr. Drug Targets 2011, 12, 348–356. [Google Scholar] [CrossRef]
- Friedman, J.; Cho, W.-K.; Chu, C.K.; Keedy, K.S.; Archin, N.M.; Margolis, D.M.; Karn, J. Epigenetic Silencing of HIV-1 by the Histone H3 Lysine 27 Methyltransferase Enhancer of Zeste 2. J. Virol. 2011, 85, 9078–9089. [Google Scholar] [CrossRef] [Green Version]
- Imai, K.; Togami, H.; Okamoto, T. Involvement of Histone H3 Lysine 9 (H3K9) Methyltransferase G9a in the Maintenance of HIV-1 Latency and Its Reactivation by BIX01294. J. Biol. Chem. 2010, 285, 16538–16545. [Google Scholar] [CrossRef] [Green Version]
- Bouchat, S.; Gatot, J.-S.; Kabeya, K.; Cardona, C.; Colin, L.; Herbein, G.; De Wit, S.; Clumeck, N.; Lambotte, O.; Rouzioux, C.; et al. Histone methyltransferase inhibitors induce HIV-1 recovery in resting CD4+ T cells from HIV-1-infected HAART-treated patients. AIDS 2012, 26, 1473–1482. [Google Scholar] [CrossRef]
- Bernhard, W.; Barreto, K.; Saunders, A.; Dahabieh, M.S.; Johnson, P.; Sadowski, I. The Suv39H1 methyltransferase inhibitor chaetocin causes induction of integrated HIV-1 without producing a T cell response. FEBS Lett. 2011, 585, 3549–3554. [Google Scholar] [CrossRef] [PubMed]
- Ebina, H.; Misawa, N.; Kanemura, Y.; Koyanagi, Y. Harnessing the CRISPR/Cas9 system to disrupt latent HIV-1 provirus. Sci. Rep. 2013, 3, srep02510. [Google Scholar] [CrossRef] [Green Version]
- Hu, W.; Kaminski, R.; Yang, F.; Zhang, Y.; Cosentino, L.; Li, F.; Luo, B.; Alvarez-Carbonell, D.; Garcia-Mesa, Y.; Karn, J.; et al. RNA-directed gene editing specifically eradicates latent and prevents new HIV-1 infection. Proc. Natl. Acad. Sci. USA 2014, 111, 11461–11466. [Google Scholar] [CrossRef] [Green Version]
- Manjunath, M.N.; Yi, G.; Dang, Y.; Shankar, P. Newer Gene Editing Technologies toward HIV Gene Therapy. Viruses 2013, 5, 2748–2766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Pan, Q.; Gendron, P.; Zhu, W.; Guo, F.; Cen, S.; Wainberg, M.A.; Liang, C. CRISPR/Cas9-Derived Mutations Both Inhibit HIV-1 Replication and Accelerate Viral Escape. Cell Rep. 2016, 15, 481–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Wang, W.; Cui, Y.C.; Pan, Q.; Zhu, W.; Gendron, P.; Guo, F.; Cen, S.; Witcher, M.; Liang, C. HIV-1 Employs Multiple Mechanisms to Resist Cas9/Single Guide RNA Targeting the Viral Primer Binding Site. J. Virol. 2018, 92, 01135-18. [Google Scholar] [CrossRef] [Green Version]
- Schönle, A.; Hartl, F.A.; Mentzel, J.; Nöltner, T.; Rauch, K.S.; Prestipino, A.; Wohlfeil, S.; Apostolova, P.; Hechinger, A.-K.; Melchinger, W.; et al. Caveolin-1 regulates TCR signal strength and regulatory T-cell differentiation into alloreactive T cells. Blood 2016, 127, 1930–1939. [Google Scholar] [CrossRef] [Green Version]
- Borger, J.G.; Morrison, V.L.; Filby, A.; Garcia, C.; Uotila, L.M.; Simbari, F.; Fagerholm, S.C.; Zamoyska, R. Caveolin-1 Influences LFA-1 Redistribution upon TCR Stimulation in CD8 T Cells. J. Immunol. 2017, 199, 874–884. [Google Scholar] [CrossRef] [Green Version]
- Tomassian, T.; Humphries, L.A.; Liu, S.D.; Silva, O.; Brooks, D.G.; Miceli, M.C. Caveolin-1 orchestrates TCR synaptic polarity, signal specificity, and function in CD8 T cells. J. Immunol. 2011, 187, 2993–3002. [Google Scholar] [CrossRef] [Green Version]
- Rothberg, K.G.; Heuser, J.E.; Donzell, W.C.; Ying, Y.-S.; Glenney, J.R.; Anderson, R.G. Caveolin, a protein component of caveolae membrane coats. Cell 1992, 68, 673–682. [Google Scholar] [CrossRef]
- Galbiati, F.; Volonte, D.; Liu, J.; Capozza, F.; Frank, P.G.; Zhu, L.; Pestell, R.G.; Lisanti, M.P. Caveolin-1 expression negatively regulates cell cycle progression by inducing G(0)/G(1) arrest via a p53/p21(WAF1/Cip1)-dependent mechanism. Mol. Biol. Cell 2001, 12, 2229–2244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fielding, P.E.; Russel, J.S.; Spencer, T.A.; Hakamata, H.; Nagao, K.; Fielding, C.J. Sterol Efflux to Apolipoprotein A-I Originates from Caveolin-Rich Microdomains and Potentiates PDGF-Dependent Protein Kinase Activity†. Biochemistry 2002, 41, 4929–4937. [Google Scholar] [CrossRef] [PubMed]
- Gargalovic, P.; Dory, L. Caveolins and macrophage lipid metabolism. J. Lipid Res. 2002, 44, 11–21. [Google Scholar] [CrossRef] [Green Version]
- Gargalovic, P.; Dory, L. Cellular apoptosis is associated with increased caveolin-1 expression in macrophages. J. Lipid Res. 2003, 44, 1622–1632. [Google Scholar] [CrossRef] [Green Version]
- Le Feuvre, R.; Guay, G.; Altschuler, Y.; Nabi, I.R. Caveolin-1 Is a Negative Regulator of Caveolae-mediated Endocytosis to the Endoplasmic Reticulum. J. Biol. Chem. 2001, 277, 3371–3379. [Google Scholar] [CrossRef] [Green Version]
- Chao, W.T.; Fan, S.S.; Chen, J.K.; Yang, V.C. Visualizing caveolin-1 and HDL in cholesterol-loaded aortic endothelial cells. J. Lipid Res. 2003, 44, 1094–1099. [Google Scholar] [CrossRef] [Green Version]
- Fu, Y.; Hoang, A.; Escher, G.; Parton, R.G.; Krozowski, Z.; Sviridov, D.; Ge, K.; Roeder, R.G.; Hankinson, O.; Wang, S. Expression of Caveolin-1 Enhances Cholesterol Efflux in Hepatic Cells. J. Biol. Chem. 2004, 279, 14140–14146. [Google Scholar] [CrossRef] [Green Version]
- Boyd, N.L.; Park, H.; Yi, H.; Boo, Y.C.; Sorescu, G.P.; Sykes, M.; Jo, H. Chronic shear induces caveolae formation and alters ERK and Akt responses in endothelial cells. Am. J. Physiol. Circ. Physiol. 2003, 285, H1113–H1122. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Muriel, O.; Echarri, A.; Hellriegel, C.; Pavon, D.M.; Beccari, L.; Del Pozo, M.A. Phosphorylated filamin A regulates actin-linked caveolae dynamics. J. Cell Sci. 2011, 124, 2763–2776. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sinha, B.; Koester, D.; Ruez, R.; Gonnord, P.; Bastiani, M.; Abankawa, D.; Stan, R.V.; Butler-Browne, G.; Vedie, B.; Johannes, L.; et al. Cells Respond to Mechanical Stress by Rapid Disassembly of Caveolae. Cell 2011, 144, 402–413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Moreno-Vicente, R.; Pavón, D.M.; Martín-Padura, I.; Català-Montoro, M.; Díez-Sánchez, A.; Quílez-Álvarez, A.; López, J.A.; Sánchez-Álvarez, M.; Vázquez, J.; Strippoli, R.; et al. Caveolin-1 Modulates Mechanotransduction Responses to Substrate Stiffness through Actin-Dependent Control of YAP. Cell Rep. 2018, 25, 1622–1635. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fiala, G.J.; Minguet, S. Caveolin-1: The Unnoticed Player in TCR and BCR Signaling. Adv. Immunol. 2018, 137, 83–133. [Google Scholar] [CrossRef]
- Lei, M.G.; Morrison, D.C. Differential Expression of Caveolin-1 in Lipopolysaccharide-Activated Murine Macrophages. Infect. Immun. 2000, 68, 5084–5089. [Google Scholar] [CrossRef] [Green Version]
- Lei, M.G.; Tan, X.; Qureshi, N.; Morrison, D.C. Regulation of Cellular Caveolin-1 Protein Expression in Murine Macrophages by Microbial Products. Infect. Immun. 2005, 73, 8136–8143. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mergia, A. The Role of Caveolin 1 in HIV Infection and Pathogenesis. Viruses 2017, 9, 129. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Palade GEFsobc. Fine structure of blood capillaries. J. Appl. Phys. 1953, 24, 1424. [Google Scholar]
- Yamada, E. The fine structure of the gall bladder epithelium of the mouse. J. Cell Biol. 1955, 1, 445–458. [Google Scholar] [CrossRef] [Green Version]
- Harris, J.; Werling, D.; Koss, M.; Monaghan, P.; Taylor, G.; Howard, C.J. Expression of caveolin by bovine lymphocytes and antigen-presenting cells. Immunology 2002, 105, 190–195. [Google Scholar] [CrossRef]
- Quest, A.F.G.; Leyton, L.; Párraga, M. Caveolins, caveolae, and lipid rafts in cellular transport, signaling, and disease. Biochem. Cell Biol. 2004, 82, 129–144. [Google Scholar] [CrossRef]
- Stan, R.V.; Tkachenko, E.; Niesman, I.R. PV1 Is a Key Structural Component for the Formation of the Stomatal and Fenestral Diaphragms. Mol. Biol. Cell 2004, 15, 3615–3630. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Stan, R.V. Structure of caveolae. Biochim. Biophys. Acta (BBA) Bioenerg. 2005, 1746, 334–348. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Parton, R.G.; Simons, K. The multiple faces of caveolae. Nat. Rev. Mol. Cell Biol. 2007, 8, 185–194. [Google Scholar] [CrossRef] [PubMed]
- Mercier, I.; Jasmin, J.-F.; Pavlides, S.; Minetti, C.; Flomenberg, N.; Pestell, R.G.; Frank, P.G.; Sotgia, F.; Lisanti, M.P. Clinical and translational implications of the caveolin gene family: Lessons from mouse models and human genetic disorders. Lab. Investig. 2009, 89, 614–623. [Google Scholar] [CrossRef] [Green Version]
- Fra, A.M.; Williamson, E.; Simons, K.; Parton, R.G. De novo formation of caveolae in lymphocytes by expression of VIP21-caveolin. Proc. Natl. Acad. Sci. USA 1995, 92, 8655–8659. [Google Scholar] [CrossRef] [Green Version]
- Hatanaka, M.; Maedab, T.; Ikemotoc, T.; Morib, H.; Seyad, T.; Shimizuac, A. Expression of Caveolin-1 in Human T Cell Leukemia Cell Lines. Biochem. Biophys. Res. Commun. 1998, 253, 382–387. [Google Scholar] [CrossRef]
- Vallejo, J.; Hardin, C.D. Expression of caveolin-1 in lymphocytes induces caveolae formation and recruitment of phosphofructokinase to the plasma membrane. FASEB J. 2005, 19, 1–19. [Google Scholar] [CrossRef]
- Medina, F.A.; Williams, T.M.; Sotgia, F.; Tanowitz, H.B.; Lisanti, M.P. A Novel Role for Caveolin-1 in B Lymphocyte Function and the Development of Thymus-Independent Immune Responses. Cell Cycle 2006, 5, 1865–1871. [Google Scholar] [CrossRef]
- Minguet, S.; Kläsener, K.; Schaffer, A.-M.; Fiala, G.J.; Osteso-Ibánez, T.; Raute, K.; Navarro-Lérida, I.; Hartl, F.A.; Seidl, M.; Reth, M.; et al. Caveolin-1-dependent nanoscale organization of the BCR regulates B cell tolerance. Nat. Immunol. 2017, 18, 1150–1159. [Google Scholar] [CrossRef] [Green Version]
- Stahlhut, M.; Van Deurs, B. Identification of Filamin as a Novel Ligand for Caveolin-1: Evidence for the Organization of Caveolin-1–associated Membrane Domains by the Actin Cytoskeleton. Mol. Biol. Cell 2000, 11, 325–337. [Google Scholar] [CrossRef] [Green Version]
- Cao, H.; Courchesne, W.E.; Mastick, C.C. A Phosphotyrosine-dependent Protein Interaction Screen Reveals a Role for Phosphorylation of Caveolin-1 on Tyrosine 14. J. Biol. Chem. 2002, 277, 8771–8774. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yao, Q.; Chen, J.; Cao, H.; Orth, J.D.; McCaffery, J.M.; Stan, R.V.; McNiven, M.A. Caveolin-1 Interacts Directly with Dynamin-2. J. Mol. Biol. 2005, 348, 491–501. [Google Scholar] [CrossRef] [PubMed]
- Schamel, W.W.; Arechaga, I.; RisueñoR, M.; Van Santen, H.M.; Cabezas, P.; Risco, C.; Valpuesta, J.M.; Alarcón, B. Coexistence of multivalent and monovalent TCRs explains high sensitivity and wide range of response. J. Exp. Med. 2005, 202, 493–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alarcón, B.; Swamy, M.; Van Santen, H.M.; Schamel, W.W. T-cell antigen-receptor stoichiometry: Pre-clustering for sensitivity. EMBO Rep. 2006, 7, 490–495. [Google Scholar] [CrossRef] [PubMed]
- Martinez-Martin, N.; Risueno, R.M.; Morreale, A.; Zaldivar, I.; Fernandez-Arenas, E.; Herranz, F.; Ortiz, A.R.; Alarcon, B. Cooperativity between T cell receptor complexes revealed by conformational mutants of CD3epsilon. Sci. Signal. 2009, 2, ra43. [Google Scholar] [CrossRef]
- Molnar, E.; Deswal, S.; Schamel, W.W. Pre-clustered TCR complexes. FEBS Lett. 2010, 584, 4832–4837. [Google Scholar] [CrossRef] [Green Version]
- Kumar, R.; Ferez, M.; Swamy, M.; Arechaga, I.; Rejas, M.T.; Valpuesta, J.; Schamel, W.W.; Alarcón, B.; Van Santen, H.M. Increased Sensitivity of Antigen-Experienced T Cells through the Enrichment of Oligomeric T Cell Receptor Complexes. Immunity 2011, 35, 375–387. [Google Scholar] [CrossRef] [Green Version]
- Schamel, W.W.; Alarcón, B. Organization of the resting TCR in nanoscale oligomers. Immunol. Rev. 2012, 251, 13–20. [Google Scholar] [CrossRef]
- Swamy, M.; Beck-Garcia, K.; Beck-Garcia, E.; Hartl, F.A.; Morath, A.; Yousefi, O.S.; Dopfer, E.P.; Molnar, E.; Schulze, A.K.; Blanco, R.; et al. A Cholesterol-Based Allostery Model of T Cell Receptor Phosphorylation. Immunity 2016, 44, 1091–1101. [Google Scholar] [CrossRef] [Green Version]
- Wang, F.; Beck-García, K.; Zorzin, C.; Schamel, W.W.A.; Davis, M.M. Inhibition of T cell receptor signaling by cholesterol sulfate, a naturally occurring derivative of membrane cholesterol. Nat. Immunol. 2016, 17, 844–850. [Google Scholar] [CrossRef] [PubMed]
- Yang, W.; Bai, Y.; Xiong, Y.; Zhang, J.; Chen, S.; Zheng, X.; Meng, X.; Li, L.; Wang, J.; Xu, C.; et al. Potentiating the antitumour response of CD8+ T cells by modulating cholesterol metabolism. Nat. Cell Biol. 2016, 531, 651–655. [Google Scholar] [CrossRef] [Green Version]
- Schamel, W.W.; Alarcon, B.; Höfer, T.; Minguet, S. The Allostery Model of TCR Regulation. J. Immunol. 2016, 198, 47–52. [Google Scholar] [CrossRef]
- Head, B.P.; Patel, H.H.; Insel, P.A. Interaction of membrane/lipid rafts with the cytoskeleton: Impact on signaling and function. Biochim. Biophys. Acta (BBA) Biomembr. 2014, 1838, 532–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.M.; Kim, H.P.; Song, R.; Choi, A.M. Caveolin-1 confers antiinflammatory effects in murine macrophages via the MKK3/p38 MAPK pathway. Am. J. Respir. Cell Mol. Biol. 2006, 34, 434–442. [Google Scholar] [CrossRef] [PubMed]
- Wang, X.M.; Nadeau, P.E.; Lin, S.; Abbott, J.E.; Mergia, A. Caveolin 1 inhibits HIV replication by transcriptional repression mediated through NF-κB Virol. J. Virol. 2011, 85, 5483–5493. [Google Scholar] [CrossRef] [Green Version]
- Fakhrzadeh, L.; Laskin, J.D.; Laskin, D.L. Regulation of caveolin-1 expression, nitric oxide production and tissue injury by tumor necrosis factor-α following ozone inhalation. Toxicol. Appl. Pharmacol. 2008, 227, 380–389. [Google Scholar] [CrossRef] [Green Version]
- Thangavel, C.; Gomes, C.M.; Zderic, S.A.; Javed, E.; Addya, S.; Singh, J.; Das, S.; Birbe, R.; Den, R.B.; Rattan, S.; et al. NF-κB and GATA-Binding Factor 6 Repress Transcription of Caveolins in Bladder Smooth Muscle Hypertrophy. Am. J. Pathol. 2019, 189, 847–867. [Google Scholar] [CrossRef] [PubMed]
- Lee, C.-Y.; Lai, T.-Y.; Tsai, M.-K.; Chang, Y.-C.; Ho, Y.-H.; Yu, I.-S.; Yeh, T.-W.; Chou, C.-C.; Lin, Y.-S.; Lawrence, T.; et al. The ubiquitin ligase ZNRF1 promotes caveolin-1 ubiquitination and degradation to modulate inflammation. Nat. Commun. 2017, 8, 15502. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zou, H.; Stoppani, E.; Volonte, D.; Galbiati, F. Caveolin-1, cellular senescence and age-related diseases. Mech. Ageing Dev. 2011, 132, 533–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.M.; Kim, H.P.; Nakahira, K.; Ryter, S.W.; Choi, A.M. The Heme Oxygenase-1/Carbon Monoxide Pathway Suppresses TLR4 Signaling by Regulating the Interaction of TLR4 with Caveolin-1. J. Immunol. 2009, 182, 3809–3818. [Google Scholar] [CrossRef] [Green Version]
- Marincowitz, C.; Genis, A.; Goswami, N.; De Boever, P.; Nawrot, T.S.; Strijdom, H. Vascular endothelial dysfunction in the wake of HIV and ART. FEBS J. 2018, 286, 1256–1270. [Google Scholar] [CrossRef] [Green Version]
- Chau-Chung, W.; Wang, S.-H.; Kuan, I.-I.; Tseng, W.-K.; Chen, M.-F.; Wu, J.-C.; Chen, Y.-L. OxLDL upregulates caveolin-1 expression in macrophages: Role for caveolin-1 in the adhesion of oxLDL-treated macrophages to endothelium. J. Cell. Biochem. 2009, 107, 460–472. [Google Scholar] [CrossRef]
- Engelman, J.A.; Lee, R.J.; Karnezis, A.; Bearss, D.; Webster, M.; Siegel, P.; Muller, W.J.; Windle, J.J.; Pestell, R.G.; Lisanti, M.P. Reciprocal Regulation of Neu Tyrosine Kinase Activity and Caveolin-1 Protein Expressionin Vitroandin Vivo. J. Biol. Chem. 1998, 273, 20448–20455. [Google Scholar] [CrossRef] [Green Version]
- Koleske, A.J.; Baltimore, D.; Lisanti, M.P. Reduction of caveolin and caveolae in oncogenically transformed cells. Proc. Natl. Acad. Sci. USA 1995, 92, 1381–1385. [Google Scholar] [CrossRef] [Green Version]
- Park, D.S.; Razani, B.; Lasorella, A.; Schreiber-Agus, N.; Pestell, R.G.; Iavarone, A.; Lisanti, M.P. Evidence that Myc isoforms transcriptionally repress caveolin-1 gene expression via an INR-dependent mechanism. Biochemistry 2001, 40, 3354–3362. [Google Scholar] [CrossRef]
- Timme, T.L.; Goltsov, A.; Tahir, S.; Li, L.; Wang, J.; Ren, C.; Johnston, R.N.; Thompson, T.C. Caveolin-1 is regulated by c-myc and suppresses c-myc-induced apoptosis. Oncogene 2000, 19, 3256–3265. [Google Scholar] [CrossRef] [Green Version]
- Porter, K.M.; Sutliff, R.L. HIV-1, reactive oxygen species, and vascular complications. Free. Radic. Biol. Med. 2012, 53, 143–159. [Google Scholar] [CrossRef] [Green Version]
- Sabin, R.J.; Anderson, R.M. Cellular Senescence - its role in cancer and the response to ionizing radiation. Genome Integr. 2011, 2, 7. [Google Scholar] [CrossRef] [Green Version]
- Sutliff, R.L.; Dikalov, S.; Weiss, D.; Parker, J.; Raidel, S.; Racine, A.K.; Russ, R.; Haase, C.P.; Taylor, W.R.; Lewis, W. Nucleoside reverse transcriptase inhibitors impair endothelium-dependent relaxation by increasing superoxide. Am. J. Physiol. Circ. Physiol. 2002, 283, H2363–H2370. [Google Scholar] [CrossRef] [Green Version]
- Kline, E.R.; Bassit, L.; Hernandez-Santiago, B.I.; Detorio, M.A.; Liang, B.; Kleinhenz, D.J.; Walp, E.R.; Dikalov, S.; Jones, D.P.; Schinazi, R.F.; et al. Long-Term Exposure to AZT, but not d4T, Increases Endothelial Cell Oxidative Stress and Mitochondrial Dysfunction. Cardiovasc. Toxicol. 2008, 9, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Pacher, P.; Gao, R.Y.; Mukhopadhyay, P.; Mohanraj, R.; Wang, H.; Horvath, B.; Yin, S. Resveratrol attenuates azidothymidine-induced cardiotoxicity by decreasing mitochondrial reactive oxygen species generation in human cardiomyocytes. Mol. Med. Rep. 2010, 4, 151–155. [Google Scholar] [CrossRef] [Green Version]
- Mak, I.T.; Nedelec, L.F.; Weglicki, W.B. Pro-oxidant properties and cytotoxicity of AZT-monophosphate and AZT. Cardiovasc. Toxicol. 2004, 4, 109–116. [Google Scholar] [CrossRef]
- Ben-Romano, R.; Rudich, A.; Etzion, S.; Potashnik, R.; Kagan, E.; Greenbaum, U.; Bashan, N. Nelfinavir induces adipocyte insulin resistance through the induction of oxidative stress: Differential protective effect of antioxidant agents. Antivir. Ther. 2006, 11, 1051. [Google Scholar]
- Mondal, D.; Pradhan, L.; Ali, M.; Agrawal, K.C. HAART Drugs Induce Oxidative Stress in Human Endothelial Cells and Increase Endothelial Recruitment of Mononuclear Cells: Exacerbation by Inflammatory Cytokines and Amelioration by Antioxidants. Cardiovasc. Toxicol. 2004, 4, 287–302. [Google Scholar] [CrossRef]
- Edeas, M. Strategies to Target Mitochondria and Oxidative Stress by Antioxidants: Key Points and Perspectives. Pharm. Res. 2011, 28, 2771–2779. [Google Scholar] [CrossRef]
- Chen, Q.; Ames, B.N. Senescence-like growth arrest induced by hydrogen peroxide in human diploid fibroblast F65 cells. Proc. Natl. Acad. Sci. USA 1994, 91, 4130–4134. [Google Scholar] [CrossRef] [Green Version]
- Chen, Q.M.; Bartholomew, J.C.; Campisi, J.; Acosta, M.; Reagan, J.D.; Ames, B.N. Molecular analysis of H2O2-induced senescent-like growth arrest in normal human fibroblasts: p53 and Rb control G1 arrest but not cell replication. Biochem. J. 1998, 332, 43–50. [Google Scholar] [CrossRef] [Green Version]
- Marnett, L.J. Oxyradicals and DNA damage. Carcinogenesis 2000, 21, 361–370. [Google Scholar] [CrossRef] [Green Version]
- Starke-Reed, P.; Yu, B. Protein Oxidation. In Methods in Aging Research; Informa UK Limited: London, UK, 1998; Volume 899, pp. 637–655. [Google Scholar]
- Yla-Herttuala, S. Oxidized LDL and atherogenesis. Ann. N. Y. Acad. Sci. 1999, 874, 134–137. [Google Scholar] [CrossRef]
- Finkel, T. Oxidant signals and oxidative stress. Curr. Opin. Cell Biol. 2003, 15, 247–254. [Google Scholar] [CrossRef]
- Frippiat, C.; Chen, Q.M.; Zdanov, S.; Magalhaes, J.-P.; Remacle, J.; Toussaint, O. Subcytotoxic H2O2Stress Triggers a Release of Transforming Growth Factor-β1, Which Induces Biomarkers of Cellular Senescence of Human Diploid Fibroblasts. J. Biol. Chem. 2000, 276, 2531–2537. [Google Scholar] [CrossRef] [Green Version]
- Frippiat, C.; Dewelle, J.; Remacle, J.; Toussaint, O. Signal transduction in H2O2-induced senescence-like phenotype in human diploid fibroblasts. Free Radic. Biol. Med. 2002, 33, 1334–1346. [Google Scholar] [CrossRef]
- Martindale, J.L.; Holbrook, N.J. Cellular response to oxidative stress: Signaling for suicide and survival. J. Cell. Physiol. 2002, 192, 1–15. [Google Scholar] [CrossRef]
- Williams, T.M.; Lisanti, M.P. Caveolin-1 in oncogenic transformation, cancer, and metastasis. Am. J. Physiol. Physiol. 2005, 288, C494–C506. [Google Scholar] [CrossRef]
- Lajoie, P.; Partridge, E.A.; Guay, G.; Goetz, J.G.; Pawling, J.; Lagana, A.; Joshi, B.; Dennis, J.W.; Nabi, I.R. Plasma membrane domain organization regulates EGFR signaling in tumor cells. J. Cell Biol. 2007, 179, 341–356. [Google Scholar] [CrossRef] [Green Version]
- Lajoie, P.; Nabi, I. Regulation of raft-dependent endocytosis. J. Cell. Mol. Med. 2007, 11, 644–653. [Google Scholar] [CrossRef] [Green Version]
- Bai, L.; Shi, G.; Zhang, L.; Guan, F.; Ma, Y.; Li, Q.; Cong, Y.-S. Cav-1 deletion impaired hematopoietic stem cell function. Cell Death Dis. 2014, 5, e1140. [Google Scholar] [CrossRef] [Green Version]
- Hulit, J.; Bash, T.; Fu, M.; Galbiati, F.; Albanese, C.; Sage, D.R.; Schlegel, A.; Zhurinsky, J.; Shtutman, M.S.; Ben-Ze’Ev, A.; et al. The Cyclin D1 Gene Is Transcriptionally Repressed by Caveolin-1. J. Biol. Chem. 2000, 275, 21203–21209. [Google Scholar] [CrossRef] [Green Version]
- Lee, S.W.; Reimer, C.L.; Oh, P.; Campbell, D.B.; E Schnitzer, J. Tumor cell growth inhibition by caveolin re-expression in human breast cancer cells. Oncogene 1998, 16, 1391–1397. [Google Scholar] [CrossRef] [Green Version]
- Zeng, W.; Tang, J.; Li, H.; Xu, H.; Lu, H.; Peng, H.; Lin, C.; Gao, R.; Lin, S.; Lin, K.; et al. Caveolin-1 deficiency protects pancreatic beta cells against palmitate-induced dysfunction and apoptosis. Cell Signal. 2018, 47, 65–78. [Google Scholar] [CrossRef]
- Park, J.; Lim, C.H.; Ham, S.; Kim, S.S.; Choi, B.-S.; Roh, T.-Y. Genome-wide analysis of histone modifications in latently HIV-1 infected T cells. AIDS 2014, 28, 1719–1728. [Google Scholar] [CrossRef] [Green Version]
- Lin, S.; Wang, X.M.; Nadeau, P.E.; Mergia, A. HIV Infection Upregulates Caveolin 1 Expression to Restrict Virus Production. J. Virol. 2010, 84, 9487–9496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lin, S.; Nadeau, P.E.; Wang, X.; Mergia, A. Caveolin-1 reduces HIV-1 infectivity by restoration of HIV Nef mediated impairment of cholesterol efflux by apoA-I. Retrovirology 2012, 9, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ménager, M.M.; Littman, D.R. Actin Dynamics Regulates Dendritic Cell-Mediated Transfer of HIV-1 to T Cells. Cell 2016, 164, 695–709. [Google Scholar] [CrossRef] [Green Version]
- Piguet, V.; Sattentau, Q. Dangerous liaisons at the virological synapse. J. Clin. Investig. 2004, 114, 605–610. [Google Scholar] [CrossRef] [Green Version]
- Jiménez-Baranda, S.; Gómez-Moutón, C.; Rojas, A.M.; Martínez-Prats, L.; Mira, E.; LaCalle, R.A.; Valencia, A.; Dimitrov, D.S.; Viola, A.; Delgado, R.; et al. Filamin-A regulates actin-dependent clustering of HIV receptors. Nat. Cell Biol. 2007, 9, 838–846. [Google Scholar] [CrossRef] [PubMed]
- Sverdlov, M.; Shinin, V.; Place, A.T.; Castellon, M.; Minshall, R.D. Filamin A Regulates Caveolae Internalization and Trafficking in Endothelial Cells. Mol. Biol. Cell 2009, 20, 4531–4540. [Google Scholar] [CrossRef] [Green Version]
- Hovanessian, A.G.; Briand, J.-P.; Said, E.A.; Svab, J.; Ferris, S.; Dali, H.; Muller, S.; Desgranges, C.; Krust, B. The Caveolin-1 Binding Domain of HIV-1 Glycoprotein gp41 Is an Efficient B Cell Epitope Vaccine Candidate against Virus Infection. Immunity 2004, 21, 617–627. [Google Scholar] [CrossRef]
- Simmons, G.E.; Jr Taylor, H.E.; Hildreth, J.E. Caveolin-1 suppresses human immunodeficiency virus-1 replication by inhibiting acetylation of NF-kappaB. Virology 2012, 432, 110–119. [Google Scholar] [CrossRef] [Green Version]
- Wang, X.M.; Nadeau, P.E.; Lo, Y.-T.; Mergia, A. Caveolin-1 Modulates HIV-1 Envelope-Induced Bystander Apoptosis through gp41. J. Virol. 2010, 84, 6515–6526. [Google Scholar] [CrossRef] [Green Version]
- Ran, X.; Xiaozhuo, R.; Trajtman, A.; Xu, W.; Kobinger, G.; Keynan, Y.; Yao, X. HIV-1 envelope glycoprotein stimulates viral transcription and increases the infectivity of the progeny virus through the manipulation of cellular machinery. Sci. Rep. 2017, 7, 1–16. [Google Scholar] [CrossRef] [PubMed]
Functions of Cav-1 | Reference |
---|---|
Component of lipid rafts | [71] |
Cell cycle regulation | [72] |
Cell signaling | [79,114,115] |
Endocytosis | [76] |
Cholesterol trafficking | [73,74,78] |
Cytoskeletal rearrangement | [113] |
Mechanosensing | [81,82] |
B cell signaling | [83] |
T cell signaling | [83] |
Cytokine production | [85,114] |
Cellular senescence | [72] |
Cell death | [74] |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2020 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
Share and Cite
Sahay, B.; Mergia, A. The Potential Contribution of Caveolin 1 to HIV Latent Infection. Pathogens 2020, 9, 896. https://doi.org/10.3390/pathogens9110896
Sahay B, Mergia A. The Potential Contribution of Caveolin 1 to HIV Latent Infection. Pathogens. 2020; 9(11):896. https://doi.org/10.3390/pathogens9110896
Chicago/Turabian StyleSahay, Bikash, and Ayalew Mergia. 2020. "The Potential Contribution of Caveolin 1 to HIV Latent Infection" Pathogens 9, no. 11: 896. https://doi.org/10.3390/pathogens9110896