HIV-1 Trans-Infection Mediated by Dendritic Cells: History
Subjects: Virology
Contributors: ,

HIV-1 cell-to-cell transmission is key for an effective viral replication that evades immunity. This highly infectious mechanism is orchestrated by different cellular targets that utilize a wide variety of processes to efficiently transfer HIV-1 particles. Dendritic cells (DCs) are the most potent antigen presenting cells that initiate antiviral immune responses, but are also the cells with highest capacity to transfer HIV-1 through cell-to-cell contacts.

  • dendritic cells (DCs)
  • HIV-1

1. Introduction

Several viruses have the ability to hijack pre-existing mechanisms of cellular communication to facilitate direct cell-to-cell viral spread [1][2][3], and the human immunodeficiency virus type 1 (HIV-1) is not an exception [1][4]. Before the definition of the precise mechanisms of cell-to-cell viral transmission, early studies highlighted the increased efficiency of HIV-1 spread by cellular contacts as compared to the diffusion-limited movement of free viral particles, suggesting that cell-to-cell dissemination might be up to 1000 times more efficient [5]. However, the first detailed description of a stable cellular junction between infected and non-infected cells to facilitate viral spread, known as virological synapse (VS), was reported for the human T cell leukaemia virus type 1 (HTLV-1), which is inefficient at infecting T cells and requires cellular contacts for effective spread [6]. Soon after this description, several studies showed co-clustering of HIV-1 proteins with their receptors CD4 and CXCR4, together with a massive viral transmission at the stable interface formed between HIV-1-infected and non-infected CD4+ T cells [7][8], thus expanding the concept of VS to HIV-1.

In  addition  to  the  VS,  there  is  another  type  of  synapse  formed  between antigen presenting cells (APCs) such as DCs and CD4+ T cells, which can even operate in the absence of productive infection of the donor APC. During antigen presentation, the formation ofcognate DC:T cell conjugates or ‘immunological synapses’ is necessary for the activation of T cells [9][10]. Once activated, T cells proliferate and differentiate into effector cells, which mediate adaptive immune responses aimed to eliminate invading viruses [11]. Intriguingly, upon HIV-1 infection, the intimate cell-to-cell contacts formed between DCs and CD4+ T cells can boost viral transmission via the formation of an ‘infectious synapse’ [12] that allows for systemic HIV-1 dissemination. 

2. Breaking the Ice: DCs Orchestrate Immune Responses against HIV-1 and Other Viruses

DCs act as pivotal players in the initiation of immunity against invading viruses [15][16], participating in both innate and adaptive immune responses. These cellular sentinels patrol distinct mucosae and, upon infection, viral sensing triggers rapid innate immune responses to contain viral spread. DC activation also elicits cellular migration towards secondary lymphoid tissues, where DCs acquire a fully mature phenotype and become competent for presenting antigens to T cells and activate them [17][18].
DCs form an integral part of innate immunity, along with other leukocytes and tissue-resident cells. DCs are present at the sites of pathogen invasion such as mucosal surfaces and the skin, and are among the first cells encountering these pathogens. DCs detect molecular patterns shared by broad groups of pathogens, termed pathogen-associated molecular patterns (PAMPs), which include viral RNA or DNA genomes, bacterial lipopolysaccharide (LPS) and yeast mannans [19][20]. DCs recognize these conserved motifs through pattern-recognition receptors (PRRs) [21]. A well-studied family of PRRs are Toll-like receptors (TLRs), which recognize a variety of ligands [22][23], each TLR having a particular sub-cellular localization and ligand specificity [24]. For instance, endosomal TLR7 and TLR8 recognize single-stranded RNA, while TLR9 binds DNA, and TLR4 recognizes LPS, an integral component of the outer membrane of gram-negative bacteria. Another group of PRRs found on DCs are C-type lectin receptors (CLRs), which include DC-SIGN (CD209), L-SIGN (CD299, Clec4M) and LSECtin (Clec4G), and recognize high mannose-containing glycans [25][26]. Within the group of I-type lectin receptors, the sialic acid-binding Ig-like lectins (Siglecs) are the best characterized members [27][28]. They are expressed by DCs, macrophages and monocytes and recognize sialic acids found on pathogens and also in host cells [29].
Viral recognition by DCs triggers the expression of genes involved in the secretion of cytokines and chemokines [30][31], which create a proinflammatory environment to eliminate or limit its replication. The main antiviral cytokines are type I interferons (IFNs), such as IFNα and IFNβ, and plasmacytoid DCs are major producers of these cytokines [32]. DCs that patrol mucosal surfaces display an immature status and can trap viruses at the entry sites, degrade them in endosomal lytic compartments and load pathogen-derived peptides onto molecules of the major histocompatibility complex (MHC). When this occurs, DCs become activated and migrate to the secondary lymphoid tissues [33], where DCs present viral-derived antigens to naïve T lymphocytes.
There are different ways of antigen presentation by DCs to T cells, depending partially on the origin of such antigens. Endogenous antigens are those expressed by the DC itself (for example viral proteins synthesized in the cytoplasm upon viral infection), and after proteasomal cleavage, the derived peptides are loaded onto MHC class I molecules and presented to CD8+ T cells [34]. In contrast, exogenous antigens are internalized by DCs through pinocytosis, phagocytosis and receptor-mediated endocytosis, processed by endosomal proteases, and the derived peptides are incorporated onto MHC class II molecules that also reach the cell surface [35]. MHC-II:peptide complexes are recognized by CD4+ T cells, which differentiate into several effector cell subtypes. In the context of viral infection these cells are mainly Th1 and T follicular helper cells [9], which prompt specific antiviral responses.
Of note, DCs have the unique capacity of presenting exogenous antigens to CD8+ T cells via MHC-I, a process known as ‘cross-presentation’ [10]. This mechanism allows antigen presentation to CD8+ T cells without productive DC infection, and is an efficient presentation pathway for viruses such as influenza A virus (IAV) [11][12] and HIV-1 [13]. Another non-classical antigen presentation pathway is that followed by endogenous peptides from measles virus [14], IAV [36] and HIV-1 [37], which are loaded onto MHC-II molecules instead of MHC-I molecules, being therefore presented to CD4+ T cells.
Despite the fined-tuned machinery for antigen presentation displayed by DCs, DC:T cell conjugates also represent a unique niche for viral transmission through the formation of infectious synapses [38], a mechanism extensively studied for HIV-1, that is also hijacked by other enveloped viruses.

3. When Immunity Is Put on Ice: DCs as Promoters of HIV-1 Cell-to-cell Transmission

Although DCs orchestrate key innate and adaptive immune antiviral responses [15][16][39], HIV-1 and other viruses have evolved strategies to evade DC surveillance [40][41][42]. Indeed, viruses exploit DC function as a way to fuel infection of target cells, colonizing distant tissues as DCs migrate (Figure 1). Landmark studies carried out in the 90s in the laboratory of Ralph Steinman showed that the efficacy of HIV-1 infection of CD4+ T cells was increased when DCs were added in co-culture as compared to the transmission of cell-free viruses [43][44]. Noteworthy, DCs are non-permissive to HIV-1 infection, as they express low levels of viral receptor and co-receptors [45], efficiently degrade incoming viruses [46][47] and express several restriction factors such as SAMHD1 that interfere with viral replication [48][49][50][51][52][53]. However, these pioneering studies demonstrated that DCs can transmit a vigorous HIV-1 infection to bystander CD4+ T cells in the absence of productive viral replication on DCs, a mechanism of viral cell-to-cell transmission known as trans-infection [25][43].
Figure 1. HIV-1 invasion is boosted by DC-mediated viral transmission in the mucosa and the migration to secondary lymphoid tissues. HIV-1 replication in the mucosa is facilitated by Siglec-1-expressing DCs that can mediate viral transmission to mucosal CD4+ T cells or migrate to secondary lymphoid tissues where the interaction with other target CD4+ T cells accelerates the settlement of systemic infection. HIV-1: human immunodeficiency virus type 1; VCC: viral containing compartment; DC: dendritic cell.
Trans-infection is one of the most potent viral transmission processes identified so far, but is only boosted when DC infection is restricted, as it is the case of HIV-1. Trans-infection was initially attributed to the activity of a DC-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), a C-type lectin receptor expressed by DCs that recognizes the HIV-1 envelope glycoprotein [25][54]. However, several studies suggested that other receptors aside from DC-SIGN operated in HIV-1 transmission [55][56][57][58][59][60][61][62]. This was suspected because DC maturation greatly increased HIV-1 trans-infection capacity while it decreased the expression of DC-SIGN [62], and because antibodies directed against DC-SIGN were not able to consistently block HIV-1 transmission [58]. Such inconsistencies led to the identification, almost a decade ago, of the sialic acid-binding immunoglobulin-like lectin 1 (Siglec-1/CD169) as the key molecule for DC-mediated HIV-1 trans-infection [63][64].
Siglec-1, also termed sialoadhesin, is an I-type lectin expressed by APCs of myeloid origin such as DCs, macrophages and monocytes [63][64][65][66][67]. At a structural level, this receptor consists of different immunoglobulin-like domains or ‘sets’, all of them extracellular. The N-terminal V-set domain contains the ligand binding activity, while the 16 extracellular C2-set domains project the V-set domain out of the cell glycocalyx, allowing for the interaction with extracellular molecules [29][68][69]. Siglec-1 has affinity for sialic acid present in both N- and O-glycans, with a higher preference for α2-3 linkages [70]. These sugars are found in a variety of complex glycolipid molecules such as gangliosides GM1a and GM3, which are components of the cell and viral membranes. In particular, these gangliosides are present in the membrane of HIV-1, allowing for viral binding to DCs via Siglec-1 and the subsequent transmission to by-stander CD4+ T cells [63][64][71][72]. Siglec-1 avidity for sialylated ligands is increased upon clustering of thousands of gangliosides in the viral membrane [29].
HIV-1 trans-infection is a dynamic process that involves viral attachment to Siglec-1, internalization within a viral containing compartment (VCC), and viral release to the intercellular space during the formation of DC:CD4+ T cell infectious synapses [73][74]. Following Siglec-1 recognition, HIV-1 particles concentrate on the surface of DCs [75][76] and are internalized into non-classical and non-acidic endosomal VCC enriched in tetraspanins, MHC-II and Siglec-1 [65][74][77]. Of note, VCCs and their content remain connected to the extracellular milieu [75][76][77], which facilitates the transmission of trapped HIV-1 particles upon the formation of DC:CD4+ T cell contacts. Although the physiological function of VCCs remains unclear, it might be related to antigen dissemination and storage, as this compartment also serves as a depot of antigen-containing extracellular vesicles that are also captured by Siglec-1 and can prime adaptive immune responses [78][79][80]. Therefore, HIV-1 exploits a pre-exiting Siglec-1-dependent antigen dissemination pathway to gain access to target CD4+ T cells.
Aside from subverting antigen presentation, HIV-1 also exploits DC migratory capacity to spread systemically. This has led to the idea that DCs can operate as ‘Trojan Horses’ and disseminate HIV-1 from the portals of viral entry to lymphoid tissues [43][81]. HIV-1 is mainly acquired through sexual transmission [82] and early events of retroviral infection have been extensively studied in non-human primate models. Following early replication at the reproductive mucosa, DCs bearing retroviruses can be found in draining lymph nodes of different non-human primate models as soon as 24 h after vaginal challenge [83][84][85][86]. Noteworthy, viral spread does not only rely on the productive infection of DCs, but also on the transference of captured viral particles via trans-infection [87][88][89][90]. Indeed, through the ex vivo culture of cells derived from human cervical tissues, we demonstrated that this mechanism relies on Siglec-1 [91]. Of note, we identified the presence of Siglec-1-enriched VCCs in the biopsy of a viremic HIV-1+ patient [91], indicating that cervical DC-mediated HIV-1 trans-infection might be a relevant process for viral acquisition in vivo. Thus, trans-infection may be key to establishing HIV-1 infection in the mucosa, leading to systemic viral dissemination thanks to the migratory capacity of DCs.
To sum up, DCs are key immune cells for the establishment of adaptive immune responses due to their migratory and antigen presentation capacities. However, pathogens such as HIV-1 have evolved strategies to subvert this immune function to efficiently spread through DC-mediated cell-to-cell contacts. Future studies should tackle this mechanism of viral dissemination, which could lead to the generation of new therapeutic strategies against HIV-1 infection.

References

  1. Dustin, M.L.; Chakraborty, A.K.; Shaw, A.S. Understanding the Structure and Function of the Immunological Synapse. Cold Spring Harb. Perspect. Biol. 2010, 2, a002311.
  2. Basu, R.; Huse, M. Mechanical Communication at the Immunological Synapse. Trends Cell Biol. 2017, 27, 241–254.
  3. Hildreth, A.D.; O’sullivan, T.E. Tissue-Resident Innate and Innate-like Lymphocyte Responses to Viral Infection. Viruses 2019, 11, 272.
  4. Piguet, V.; Steinman, R.M. The Interaction of HIV with Dendritic Cells: Outcomes and Pathways. Trends Immunol. 2007, 28, 503–510.
  5. Steinman, R.M. The Dendritic Cell System and Its Role in Immunogenicity. Annu. Rev. Immunol. 1991, 9, 271–296.
  6. Banchereau, J.; Steinman, R.M. Dendritic Cells and the Control of Immunology. Nature 1998, 392, 245–252.
  7. Jolly, C.; Kashefi, K.; Hollinshead, M.; Sattentau, Q.J. HIV-1 Cell to Cell Transfer across an Env-Induced, Actin-Dependent Synapse. J. Exp. Med. 2004, 199, 283–293.
  8. Blanco, J.; Bosch, B.; Fernández-Figueras, M.T.; Barretina, J.; Clotet, B.; Esté, J.A. High Level of Coreceptor-Independent HIV Transfer Induced by Contacts between Primary CD4 T Cells. J. Biol. Chem. 2004, 279, 51305–51314.
  9. Paludan, C.; Schmid, D.; Landthaler, M.; Vockerodt, M.; Kube, D.; Tuschl, T.; Münz, C. Endogenous MHC Class II Processing of a Viral Nuclear Antigen after Autophagy. Science 2005, 307, 593–596.
  10. Wilson, N.S.; Behrens, G.M.N.; Lundie, R.J.; Smith, C.M.; Waithman, J.; Young, L.; Forehan, S.P.; Mount, A.; Steptoe, R.J.; Shortman, K.D.; et al. Systemic Activation of Dendritic Cells by Toll-like Receptor Ligands or Malaria Infection Impairs Cross-Presentation and Antiviral Immunity. Nat. Immunol. 2006, 7, 165–172.
  11. Steinman, R.M. The Dendritic Cell System and Its Role in Immunogenicity. Annu. Rev. Immunol. 1991, 9, 271–296.
  12. Banchereau, J.; Steinman, R.M. Dendritic Cells and the Control of Immunology. Nature 1998, 392, 245–252.
  13. Janeway, C.A.; Medzhitov, R. Innate Immune Recognition. Annu. Rev. Immunol. 2002, 20, 197–216.
  14. Cui, J.; Chen, Y.; Wang, H.Y.; Wang, R.F. Mechanisms and Pathways of Innate Immune Activation and Regulation in Health and Cancer. Hum. Vaccines Immunother. 2014, 10, 3270–3285.
  15. Janeway, C.A. Pillars Article: Approaching the Asymptote? Evolution and Revolution in Immunology. Cold Spring Harb Symp Quant Biol. 1989. 54: 1-13. J. Immunol. 2013, 191, 4475–4487.
  16. Medzhitov, R.; Preston-Hurlburt, P.; Janeway, C.A. A Human Homologue of the Drosophila Toll Protein Signals Activation of Adaptive Immunity. Nature 1997, 388, 394–397.
  17. Poltorak, A.; Smirnova, I.; He, X.; Liu, M.-Y.; Van Huffel, C.; Birdwell, D.; Alejos, E.; Silva, M.; Du, X.; Thompson, P.; et al. Genetic and Physical Mapping of TheLpsLocus: Identification of the Toll-4 Receptor as a Candidate Gene in the Critical Region. Blood Cells Mol. Dis. 1998, 24, 340–355.
  18. O’Neill, L.A.J.; Golenbock, D.; Bowie, A.G. The History of Toll-like Receptors—Redefining Innate Immunity. Nat. Rev. Immunol. 2013, 13, 453–460.
  19. Geijtenbeek, T.B.H.; Kwon, D.S.; Torensma, R.; van Vliet, S.J.; van Duijnhoven, G.C.F.; Middel, J.; Cornelissen, I.L.M.H.A.; Nottet, H.S.L.M.; KewalRamani, V.N.; Littman, D.R.; et al. DC-SIGN, a Dendritic Cell-Specific HIV-1-Binding Protein That Enhances Trans-Infection of T Cells. Cell 2000, 100, 587–597.
  20. Zhang, F.; Ren, S.; Zuo, Y. DC-SIGN, DC-SIGNR and LSECtin: C-Type Lectins for Infection. Int. Rev. Immunol. 2014, 33, 54–66.
  21. Powell, L.D.; Varki, A. I-Type Lectins. J. Biol. Chem. 1995, 270, 14243–14246.
  22. Angata, T.; Brinkman-Van der Linden, E.C.M. I-Type Lectins. Biochim. Biophys. Acta (BBA)—Gen. Subj. 2002, 1572, 294–316.
  23. Crocker, P.R.; Paulson, J.C.; Varki, A. Siglecs and Their Roles in the Immune System. Nat. Rev. Immunol. 2007, 7, 255–266.
  24. Collin, M.; Bigley, V. Human Dendritic Cell Subsets: An Update. Immunology 2018, 154, 3–20.
  25. Rhodes, J.W.; Tong, O.; Harman, A.N.; Turville, S.G. Human Dendritic Cell Subsets, Ontogeny, and Impact on HIV Infection. Front. Immunol. 2019, 10, 1008.
  26. Siegal, F.P.; Kadowaki, N.; Shodell, M.; Fitzgerald-Bocarsly, P.A.; Shah, K.; Ho, S.; Antonenko, S.; Liu, Y.-J. The Nature of the Principal Type 1 Interferon-Producing Cells in Human Blood. Science 1999, 284, 1835–1837.
  27. Worbs, T.; Hammerschmidt, S.I.; Förster, R. Dendritic Cell Migration in Health and Disease. Nat. Rev. Immunol. 2017, 17, 30–48.
  28. Hashimoto, M.; Im, S.J.; Araki, K.; Ahmed, R. Cytokine-Mediated Regulation of CD8 T-Cell Responses during Acute and Chronic Viral Infection. Cold Spring Harb. Perspect. Biol. 2019, 11, a028464.
  29. Mantegazza, A.R.; Magalhaes, J.G.; Amigorena, S.; Marks, M.S. Presentation of Phagocytosed Antigens by MHC Class I and II. Traffic 2013, 14, 135–152.
  30. Huang, Q.; Hu, J.; Tang, J.; Xu, L.; Ye, L. Molecular Basis of the Differrentiation and Function of Virus Specific Follicular Helper CD4+ T Cells. Front. Immunol. 2019, 10, 249.
  31. Joffre, O.P.; Segura, E.; Savina, A.; Amigorena, S. Cross-Presentation by Dendritic Cells. Nat. Rev. Immunol. 2012, 12, 557–569.
  32. Smed-Sörensen, A.; Chalouni, C.; Chatterjee, B.; Cohn, L.; Blattmann, P.; Nakamura, N.; Delamarre, L.; Mellman, I. Influenza a Virus Infection of Human Primary Dendritic Cells Impairs Their Ability to Cross-Present Antigen to CD8 T Cells. PLoS Pathog. 2012, 8, e1002572.
  33. Bender, A.; Bui, L.K.; Feldman, M.A.; Larsson, M.; Bhardwaj, N. Inactivated Influenza Virus, When Presented on Dendritic Cells, Elicits Human CD8+ Cytolytic T Cell Responses. J. Exp. Med. 1995, 182, 1663–1671.
  34. Larsson, M.; Fonteneau, J.-F.; Lirvall, M.; Haslett, P.; Lifson, J.D.; Bhardwaj, N. Activation of HIV-1 Specific CD4 and CD8 T Cells by Human Dendritic Cells: Roles for Cross-Presentation and Non-Infectious HIV-1 Virus. AIDS 2002, 16, 1319–1329.
  35. JACOBSON, S.; SEKALY, R.P.; BELLINI, W.J.; JOHNSON, C.L.; McFARLAND, H.F.; LONG, E.O. Recognition of Intracellular Measles Virus Antigens by HLA Class II Restricted Measles Virus-Specific Cytotoxic T Lymphocytes. Ann. N. Y. Acad. Sci. 1988, 540, 352–353.
  36. Miller, M.A.; Ganesan, A.P.V.; Luckashenak, N.; Mendonca, M.; Eisenlohr, L.C. Endogenous Antigen Processing Drives the Primary CD4+ T Cell Response to Influenza. Nat. Med. 2015, 21, 1216–1222.
  37. Coulon, P.-G.; Richetta, C.; Rouers, A.; Blanchet, F.P.; Urrutia, A.; Guerbois, M.; Piguet, V.; Theodorou, I.; Bet, A.; Schwartz, O.; et al. HIV-Infected Dendritic Cells Present Endogenous MHC Class II–Restricted Antigens to HIV-Specific CD4+ T Cells. J. Immunol. 2016, 197, 517–532.
  38. Piguet, V.; Steinman, R.M. The Interaction of HIV with Dendritic Cells: Outcomes and Pathways. Trends Immunol. 2007, 28, 503–510.
  39. Norbury, C.C.; Malide, D.; Gibbs, J.S.; Bennink, J.R.; Yewdell, J.W. Visualizing Priming of Virus-Specific CD8+ T Cells by Infected Dendritic Cells in Vivo. Nat. Immunol. 2002, 3, 265–271.
  40. Pollara, G.; Kwan, A.; Newton, P.J.; Handley, M.E.; Chain, B.M.; Katz, D.R. Dendritic Cells in Viral Pathogenesis: Protective or Defective? Int. J. Exp. Pathol. 2005, 86, 187–204.
  41. Finlay, B.B.; McFadden, G. Anti-Immunology: Evasion of the Host Immune System by Bacterial and Viral Pathogens. Cell 2006, 124, 767–782.
  42. Rescigno, M. Dendritic Cell Functions: Learning from Microbial Evasion Strategies. Semin. Immunol. 2015, 27, 119–124.
  43. Cameron, P.U.; Freudenthal, P.S.; Barker, J.M.; Gezelter, S.; Inaba, K.; Steinman, R.M. Dendritic Cells Exposed to Human Immunodeficiency Virus Type-1 Transmit a Vigorous Cytopathic Infection to CD4+ T Cells. Science 1992, 257, 383–387.
  44. Pope, M.; Betjes, M.G.H.; Romani, N.; Hirmand, H.; Cameron, P.U.; Hoffman, L.; Gezelter, S.; Schuler, G.; Steinman, R.M. Conjugates of Dendritic Cells and Memory T Lymphocytes from Skin Facilitate Productive Infection with HIV-1. Cell 1994, 78, 389–398.
  45. Lee, B.; Sharron, M.; Montaner, L.J.; Weissman, D.; Doms, R.W. Quantification of CD4, CCR5, and CXCR4 Levels on Lymphocyte Subsets, Dendritic Cells, and Differentially Conditioned Monocyte-Derived Macrophages. Proc. Natl. Acad. Sci. USA 1999, 96, 5215–5220.
  46. Turville, S.G.; Santos, J.J.; Frank, I.; Cameron, P.U.; Wilkinson, J.; Miranda-Saksena, M.; Dable, J.; Stössel, H.; Romani, N.; Piatak, M.; et al. Immunodeficiency Virus Uptake, Turnover, and 2-Phase Transfer in Human Dendritic Cells. Blood 2004, 103, 2170–2179.
  47. Moris, A.; Nobile, C.; Buseyne, F.; Porrot, F.; Abastado, J.P.; Schwartz, O. DC-SIGN Promotes Exogenous MHC-I-Restricted HIV-1 Antigen Presentation. Blood 2004, 103, 2648–2654.
  48. Laguette, N.; Sobhian, B.; Casartelli, N.; Ringeard, M.; Chable-Bessia, C.; Ségéral, E.; Yatim, A.; Emiliani, S.; Schwartz, O.; Benkirane, M. SAMHD1 Is the Dendritic- and Myeloid-Cell-Specific HIV-1 Restriction Factor Counteracted by Vpx. Nature 2011, 474, 654–657.
  49. Hrecka, K.; Hao, C.; Gierszewska, M.; Swanson, S.K.; Kesik-Brodacka, M.; Srivastava, S.; Florens, L.; Washburn, M.P.; Skowronski, J. Vpx Relieves Inhibition of HIV-1 Infection of Macrophages Mediated by the SAMHD1 Protein. Nature 2011, 474, 658–661.
  50. Lahouassa, H.; Daddacha, W.; Hofmann, H.; Ayinde, D.; Logue, E.C.; Dragin, L.; Bloch, N.; Maudet, C.; Bertrand, M.; Gramberg, T.; et al. SAMHD1 Restricts the Replication of Human Immunodeficiency Virus Type 1 by Depleting the Intracellular Pool of Deoxynucleoside Triphosphates. Nat. Immunol. 2012, 13, 223–228.
  51. Sheehy, A.M.; Gaddis, N.C.; Choi, J.D.; Malim, M.H. Isolation of a Human Gene That Inhibits HIV-1 Infection and Is Suppressed by the Viral Vif Protein. Nature 2002, 418, 646–650.
  52. Mariani, R.; Chen, D.; Schröfelbauer, B.; Navarro, F.; König, R.; Bollman, B.; Münk, C.; Nymark-McMahon, H.; Landau, N.R. Species-Specific Exclusion of APOBEC3G from HIV-1 Virions by Vif. Cell 2003, 114, 21–31.
  53. Mangeat, B.; Turelli, P.; Caron, G.; Friedli, M.; Perrin, L.; Trono, D. Broad Antiretroviral Defence by Human APOBEC3G through Lethal Editing of Nascent Reverse Transcripts. Nature 2003, 424, 99–103.
  54. Geijtenbeek, T.B.H.; Tornsma, R.; van Vliet, S.J.; van Duijnhoven, G.C.F.; Adema, G.J.; van Kooyk, Y.; Figdor, C.G. Identification of DC-SIGN, a Novel Dendritic Cell-Specific ICAM-3 Receptor That Supports Primary Immune Responses. Cell 2000, 100, 575–585.
  55. Turville, S.G.; Cameron, P.U.; Handley, A.; Lin, G.; Pöhlmann, S.; Doms, R.W.; Cunningham, A.L. Diversity of Receptors Binding HIV on Dendritic Cell Subsets. Nat. Immunol. 2002, 3, 975–983.
  56. Wu, L.; Bashirova, A.A.; Martin, T.D.; Villamide, L.; Mehlhop, E.; Chertov, A.O.; Unutmaz, D.; Pope, M.; Carrington, M.; KewalRamani, V.N. Rhesus Macaque Dendritic Cells Efficiently Transmit Primate Lentiviruses Independently of DC-SIGN. Proc. Natl. Acad. Sci. USA 2002, 99, 1568–1573.
  57. Gummuluru, S.; Rogel, M.; Stamatatos, L.; Emerman, M. Binding of Human Immunodeficiency Virus Type 1 to Immature Dendritic Cells Can Occur Independently of DC-SIGN and Mannose Binding C-Type Lectin Receptors via a Cholesterol-Dependent Pathway. J. Virol. 2003, 77, 12865–12874.
  58. Trumpfheller, C.; Park, C.G.; Finke, J.; Steinman, R.M.; Granelli-Piperno, A. Cell Type-Dependent Retention and Transmission of HIV-1 by DC-SIGN. Int. Immunol. 2003, 15, 289–298.
  59. Granelli-Piperno, A.; Pritsker, A.; Pack, M.; Shimeliovich, I.; Arrighi, J.-F.; Park, C.G.; Trumpfheller, C.; Piguet, V.; Moran, T.M.; Steinman, R.M. Dendritic Cell-Specific Intercellular Adhesion Molecule 3-Grabbing Nonintegrin/CD209 Is Abundant on Macrophages in the Normal Human Lymph Node and Is Not Required for Dendritic Cell Stimulation of the Mixed Leukocyte Reaction. J. Immunol. 2005, 175, 4265–4273.
  60. Boggiano, C.; Manel, N.; Littman, D.R. Dendritic Cell-Mediated Trans-Enhancement of Human Immunodeficiency Virus Type 1 Infectivity Is Independent of DC-SIGN. J. Virol. 2007, 81, 2519–2523.
  61. Wang, J.-H.; Janas, A.M.; Olson, W.J.; Wu, L. Functionally Distinct Transmission of Human Immunodeficiency Virus Type 1 Mediated by Immature and Mature Dendritic Cells. J. Virol. 2007, 81, 8933–8943.
  62. Izquierdo-Useros, N.; Blanco, J.; Erkizia, I.; Fernández-Figueras, M.T.; Borràs, F.E.; Naranjo-Gómez, M.; Bofill, M.; Ruiz, L.; Clotet, B.; Martinez-Picado, J. Maturation of Blood-Derived Dendritic Cells Enhances Human Immunodeficiency Virus Type 1 Capture and Transmission. J. Virol. 2007, 81, 7559–7570.
  63. Izquierdo-Useros, N.; Lorizate, M.; Puertas, M.C.; Rodriguez-Plata, M.T.; Zangger, N.; Erikson, E.; Pino, M.; Erkizia, I.; Glass, B.; Clotet, B.; et al. Siglec-1 Is a Novel Dendritic Cell Receptor That Mediates HIV-1 Trans-Infection through Recognition of Viral Membrane Gangliosides. PLoS Biol. 2012, 10, e1001448.
  64. Puryear, W.B.; Akiyama, H.; Geer, S.D.; Ramirez, N.P.; Yu, X.; Reinhard, B.M.; Gummuluru, S. Interferon-Inducible Mechanism of Dendritic Cell-Mediated HIV-1 Dissemination Is Dependent on Siglec-1/CD169. PLoS Pathog. 2013, 9, e1003291.
  65. Pino, M.; Erkizia, I.; Benet, S.; Erikson, E.; Fernández-Figueras, M.T.; Guerrero, D.; Dalmau, J.; Ouchi, D.; Rausell, A.; Ciuffi, A.; et al. HIV-1 Immune Activation Induces Siglec-1 Expression and Enhances Viral Trans-Infection in Blood and Tissue Myeloid Cells. Retrovirology 2015, 12, 37.
  66. Crocker, P.R.; Gordon, S. Mouse Macrophage Hemagglutinin (Sheep Erythrocyte Receptor) with Specificity for Sialylated Glycoconjugates Characterized by a Monoclonal Antibody. J. Exp. Med. 1989, 169, 1333–1346.
  67. Rempel, H.; Calosing, C.; Sun, B.; Pulliam, L. Sialoadhesin Expressed on IFN-Induced Monocytes Binds HIV-1 and Enhances Infectivity. PLoS ONE 2008, 3, e1967.
  68. Crocker, P.R.; Mucklow, S.; Bouckson, V.; McWilliam, A.; Willis, A.C.; Gordon, S.; Milon, G.; Kelm, S.; Bradfield, P. Sialoadhesin, a Macrophage Sialic Acid Binding Receptor for Haemopoietic Cells with 17 Immunoglobulin-like Domains. EMBO J. 1994, 13, 4490–4503.
  69. Crocker, P.R.; Vinson, M.; Kelm, S.; Drickamer, K. Molecular Analysis of Sialoside Binding to Sialoadhesin by NMR and Site-Directed Mutagenesis. Biochem. J. 1999, 341, 355–361.
  70. Hartnell, A.; Steel, J.; Turley, H.; Jones, M.; Jackson, D.G.; Crocker, P.R. Characterization of Human Sialoadhesin, a Sialic Acid Binding Receptor Expressed by Resident and Inflammatory Macrophage Populations. Blood 2001, 97, 288–296.
  71. Izquierdo-Useros, N.; Lorizate, M.; Contreras, F.-X.; Rodriguez-Plata, M.T.; Glass, B.; Erkizia, I.; Prado, J.G.; Casas, J.; Fabriàs, G.; Kräusslich, H.-G.; et al. Sialyllactose in Viral Membrane Gangliosides Is a Novel Molecular Recognition Pattern for Mature Dendritic Cell Capture of HIV-1. PLoS Biol. 2012, 10, e1001315.
  72. Puryear, W.B.; Yu, X.; Ramirez, N.P.; Reinhard, B.M.; Gummuluru, S. HIV-1 Incorporation of Host-Cell-Derived Glycosphingolipid GM3 Allows for Capture by Mature Dendritic Cells. Proc. Natl. Acad. Sci. USA 2012, 109, 7475–7480.
  73. Rodriguez-Plata, M.T.; Puigdomènech, I.; Izquierdo-Useros, N.; Puertas, M.C.; Carrillo, J.; Erkizia, I.; Clotet, B.; Blanco, J.; Martinez-Picado, J. The Infectious Synapse Formed between Mature Dendritic Cells and CD4(+) T Cells Is Independent of the Presence of the HIV-1 Envelope Glycoprotein. Retrovirology 2013, 10, 42.
  74. McDonald, D.; Wu, L.; Bohks, S.M.; KewalRamani, V.N.; Unutmaz, D.; Hope, T.J. Recruitment of HIV and Its Receptors to Dendritic Cell-T Cell Junctions. Science 2003, 300, 1295–1297.
  75. Izquierdo-Useros, N.; Esteban, O.; Rodriguez-Plata, M.T.; Erkizia, I.; Prado, J.G.; Blanco, J.; García-Parajo, M.F.; Martinez-Picado, J. Dynamic Imaging of Cell-Free and Cell-Associated Viral Capture in Mature Dendritic Cells. Traffic 2011, 12, 1702–1713.
  76. Yu, H.J.; Reuter, M.A.; McDonald, D. HIV Traffics through a Specialized, Surface-Accessible Intracellular Compartment during Trans-Infection of T Cells by Mature Dendritic Cells. PLoS Pathog. 2008, 4, e1000134.
  77. Garcia, E.; Pion, M.; Pelchen-Matthews, A.; Collinson, L.; Arrighi, J.-F.F.; Blot, G.; Leuba, F.; Escola, J.-M.M.; Demaurex, N.; Marsh, M.; et al. HIV-1 Trafficking to the Dendritic Cell-T-Cell Infectious Synapse Uses a Pathway of Tetraspanin Sorting to the Immunological Synapse. Traffic 2005, 6, 488–501.
  78. Izquierdo-Useros, N.; Naranjo-Gómez, M.; Archer, J.; Hatch, S.C.; Erkizia, I.; Blanco, J.; Borràs, F.E.; Puertas, M.C.; Connor, J.H.; Fernádez-Figueras, M.T.; et al. Capture and Transfer of HIV-1 Particles by Mature Dendritic Cells Converges with the Exosome-Dissemination Pathway. Blood 2009, 113, 2732–2741.
  79. Théry, C.; Duban, L.; Segura, E.; Véron, P.; Lantz, O.; Amigorena, S. Indirect Activation of Naïve CD4+ T Cells by Dendritic Cell-Derived Exosomes. Nat. Immunol. 2002, 3, 1156–1162.
  80. Benet, S.; Gálvez, C.; Drobniewski, F.; Kontsevaya, I.; Arias, L.; Monguió-Tortajada, M.; Erkizia, I.; Urrea, V.; Ong, R.Y.; Luquin, M.; et al. Dissemination of Mycobacterium Tuberculosis Is Associated to a SIGLEC1 Null Variant That Limits Antigen Exchange via Trafficking Extracellular Vesicles. J. Extracell. Vesicles 2021, 10, e12046.
  81. Knight, S.C.; Patterson, S. Bone Marrow-Derived Dendritic Cells, Infection with Human Immunodeficiency Virus, and Immunopathology. Annu. Rev. Immunol. 1997, 15, 593–615.
  82. UNAIDS Data 2021. Available online: https://www.unaids.org/sites/default/files/media_asset/JC3032_AIDS_Data_book_2021_En.pdf (accessed on 10 November 2021).
  83. Spira, A.I.; Marx, P.A.; Patterson, B.K.; Mahoney, J.; Koup, R.A.; Wolinsky, S.M.; Ho, D.D. Cellular Targets of Infection and Route of Viral Dissemination after an Intravaginal Inoculation of Simian Immunodeficiency Virus into Rhesus Macaques. J. Exp. Med. 1996, 183, 215–225.
  84. Miller, C.J.; Li, Q.; Abel, K.; Kim, E.-Y.; Ma, Z.-M.; Wietgrefe, S.; La Franco-Scheuch, L.; Compton, L.; Duan, L.; Shore, M.D.; et al. Propagation and Dissemination of Infection after Vaginal Transmission of Simian Immunodeficiency Virus. J. Virol. 2005, 79, 9217–9227.
  85. Masurier, C.; Salomon, B.; Guettari, N.; Pioche, C.; Guigon, M.; Klatzmann, D.; Lachapelle, F. Dendritic Cells Route Human Immunodeficiency Virus to Lymph Nodes after Vaginal or Intravenous Administration to Mice. J. Virol. 1998, 72, 7822–7829.
  86. Hu, J.; Gardner, M.B.; Miller, C.J. Simian Immunodeficiency Virus Rapidly Penetrates the Cervicovaginal Mucosa after Intravaginal Inoculation and Infects Intraepithelial Dendritic Cells. J. Virol. 2000, 74, 6087–6095.
  87. Shen, R.; Kappes, J.C.; Smythies, L.E.; Richter, H.E.; Novak, L.; Smith, P.D. Vaginal Myeloid Dendritic Cells Transmit Founder HIV-1. J. Virol. 2014, 88, 7683–7688.
  88. Trifonova, R.T.; Bollman, B.; Barteneva, N.S.; Lieberman, J. Myeloid Cells in Intact Human Cervical Explants Capture HIV and Can Transmit It to CD4 T Cells. Front. Immunol. 2018, 9, 2719.
  89. Hu, Q.; Frank, I.; Williams, V.; Santos, J.J.; Watts, P.; Griffin, G.E.; Moore, J.P.; Pope, M.; Shattock, R.J. Blockade of Attachment and Fusion Receptors Inhibits HIV-1 Infection of Human Cervical Tissue. J. Exp. Med. 2004, 199, 1065–1075.
  90. Sanders, R.W.; de Jong, E.C.; Baldwin, C.E.; Schuitemaker, J.H.N.; Kapsenberg, M.L.; Berkhout, B. Differential Transmission of Human Immunodeficiency Virus Type 1 by Distinct Subsets of Effector Dendritic Cells. J. Virol. 2002, 76, 7812–7821.
  91. Perez-Zsolt, D.; Cantero-Pérez, J.; Erkizia, I.; Benet, S.; Pino, M.; Serra-Peinado, C.; Hernández-Gallego, A.; Castellví, J.; Tapia, G.; Arnau-Saz, V.; et al. Dendritic Cells from the Cervical Mucosa Capture and Transfer HIV-1 via Siglec-1. Front. Immunol. 2019, 10, 825.
More