Skip to main content
Chinese Medical Journal logoLink to Chinese Medical Journal
. 2020 Sep 15;133(23):2867–2873. doi: 10.1097/CM9.0000000000001085

Heterogeneity of HIV-1 latent reservoirs

Jia-Cong Zhao 1,2, Kai Deng 1,2
Editors: Peng Lyu, Xin Chen
PMCID: PMC10631589  PMID: 33273337

Abstract

Antiretroviral therapy (ART) can effectively inhibit human immunodeficiency virus-1 (HIV-1) replication, but is not curative due to the existence of a stable viral latent reservoir harboring replication-competent proviruses. In order to reduce or eliminate the HIV-1 latent reservoir, characteristics of the latently infected cells need to be intensively studied, and a comprehensive understanding of the heterogenous nature of the latent reservoir will be critical to develop novel therapeutic strategies. Here, we discuss the different cell types and mechanisms contributing to the complexity and heterogeneity of HIV-1 latent reservoirs, and summarize the key challenges to the development of cure strategies for acquired immunodeficiency syndrome (AIDS).

Keywords: Clonal expansion, Heterogeneity, human immunodeficiency virus-1, HIV-1, Integration sites, Latent reservoirs

Introduction

As a critical step of the retroviral life cycle, human immunodeficiency virus-1 (HIV-1) integrates its viral genome into the host genome of the infected cells, a small fraction of which can survive and persist for a long time in a resting state, avoiding cytotoxic effects or immune clearance. Since antiretroviral therapy (ART) was designed to block new infections but cannot eliminate infected cells, these latently infected cells (HIV-1 latent reservoir) can produce infectious viruses and rekindle new infections once ART is interrupted.[1,2] These latently infected cells are extremely stable, as clinical data and mathematical modeling predicted that it might take more than 73 years for them to decay to zero under continuous ART.[3,4] Therefore, HIV-1 latent reservoir has been widely considered the major barrier to viral elimination and the focal point of acquired immunodeficiency syndrome (AIDS) cure research.

The successful cases of “Berlin Patient”[5] and “London Patient”[6,7] have ignited global enthusiasm of achieving AIDS functional cure for more HIV-1 infected individuals. However, no apparent reduction of HIV-1 latent reservoir has been observed in a series of clinical trials other than the bone marrow transplantations. One of the biggest challenges underlying the endeavors to reduce HIV-1 latent reservoir is the tremendous heterogeneity of these viral genome-harboring cells, thus a comprehensive understanding of the heterogenous characteristics of latently infected cells will be critical to develop novel strategies to eliminate these cells. In this review, we discuss the different cell types and mechanisms contributing to the complexity and heterogeneity of human immunodeficiency virus-1 latent reservoirs and summarize the key challenges for developing effective therapeutic strategies for AIDS functional cure.

Distribution of HIV-1 latent reservoirs in various cell types and subpopulations

Many cell types including CD4+ T cells, macrophages, dendritic cells are susceptible to HIV-1 infection. However, only resting memory CD4+ T cells have been widely regarded and studied as the typical latent reservoirs for HIV-1, primarily due to their unique physiological state of active and resting,[8] and the dynamic process of effector-to-memory transition.[9] The earliest differentiation state of mature CD4+ T cells is called naive T cells (TN), which possess the greatest proliferative potential and a long half-life. After antigen exposure, these TN cells can further differentiate into central memory T (TCM), transitional memory T (TTM), effector memory T (TEM), and terminally differentiated T (TTD) cells, as identified by the expression of cell surface markers including CD45RA, CD45RO, CCR7 or CD62L.[10] TCM cells are considered to be the primary component of HIV-1 latent reservoirs due to its abundance and long-life span.[11] TEM cells were also shown to harbor a large portion of latent HIV-1.[12] They have a shorter life span but higher proliferation rates, which may serve as an advantage for viral persistence. TTD cells have a very small proportion of CD4+ T cell reservoirs but integrated HIV-1 DNA can also be detected.[13] Resident memory T cells (TRM) are widely distributed in tissues but only petrol in a limited area, expressing specific homing receptors and providing local surveillance and protection.[14] It is recently reported that TRM cells are also targets of HIV-1 infection and sites of viral persistence, but their roles in HIV-1 latency still needs to be further elucidated.[15]

Besides these memory T cell subsets, TN cells can also differentiate into the polarized, functional subsets, including Th1, Th2, Th17, follicular helper T (TFH), and regulatory T (Treg) cells, which are reported to also harbor replication-competent HIV-1 in patients under long time ART. Similar studies have found that CD4+ T helper cells with Th1/Th17 polarization have a preferential role as long-term HIV-1 reservoirs during ART.[1618] As Treg cells harbor high frequency of HIV-1 provirus and play an essential role in immune regulation, specific eradication of Treg as a HIV-1 reservoir will be an obstacle.[19] A new ex vivo method called QUECEL (quiescent effector cell latency) mimics the process of T cell differentiation by polarizing TN cells into Th1, Th2, Th17, and Treg cells, and then infects them with a reporter pseudovirus, which can serve as a powerful tool to study the mechanism underlying HIV-1 latency.[20] TFH cells expressing the C-X-C chemokine receptor type 5 (CXCR5) also have been identified as a major component of the HIV-1 latent reservoirs, which are infected more frequently by both productive and latent form of the virus than non-TFH cells.[2123] It is recently found that the ratio of X4-tropic provirus in peripheral T follicular helper (pTFH) cells reflects disease progression and treatment outcomes during ART.[24]

According to the definition of HIV-1 latency, if a cell is integrated with a replication-competent provirus and can persist for a relatively long time under ART, it can be considered as a part of the reservoirs. Therefore, non-T cell reservoirs can also play a role in HIV-1 latency and has become a topic of attention lately. As a key member of the innate immune system, macrophages can express low-level of CD4 molecules and the C-C chemokine receptor type 5 (CCR5), and their infection by R5-tropic HIV-1 have been firmly demonstrated for years. However, it remained controversial whether macrophages can be characterized as a classical latent reservoir of HIV-1. Recent studies had shown that macrophages carry the replication-competent HIV-1 provirus under ART in various tissues including gut-associated lymphoid tissue, lymph node, brain, lung, urethra and liver.[2530] Although infectious HIV-1 can be recovered from these macrophages, whether these viruses are kept in a low level of replicative state or are truly in a latent form still need further investigations. Since macrophages are usually long-lived, resistant to cytopathic effects of HIV-1, and can reside in various tissues, they certainly present as an important barrier to the elimination of HIV-1 reservoirs.[31] Astrocytes and microglia are considered macrophage-like cells in the central nervous system (CNS), which have been suggested to be potential viral reservoirs in the brain.[32] They can be infected by cell-to-cell contact with lymphocytes carrying virus, or up-regulating expression of CD4 and co-receptors to make them permissive. The blood-brain barrier makes them a challenge to be eliminated. The extremely low frequency of latently infected cells in patients under suppressive ART, the limited availability of tissue samples, and the lack of specific detection methods have all presented difficulties to study the distribution of HIV-1 latent reservoir, which continue to be an imperative issue if viral eradication is desired. The various cell types and subpopulations in different anatomical locations harboring latent HIV-1, and their contributions to the viral latent reservoir still need further investigations.

Multifactorial mechanisms underlying HIV-1 latency and its reversal

The transcriptionally silent state of HIV-1 provirus in ART-suppressed infected individuals is maintained through a variety of mechanisms.[33] First, the key initiation factors for HIV-1 transcription are sequestered in cytoplasm, such as nuclear factor κB (NF-κB) and nuclear factor of activated T cells (NFAT).[34] The active form of NF-κB is bound by inhibitor of NF-κB (IκB), while NFAT is phosphorylated in its inactive form. Second, the positive transcription elongation factor b (P-TEFb), a critical cofactor for HIV-1 transcriptional elongation through RNA polymerase II phosphorylation, is restricted in an inactive complex.[35] Furthermore, epigenetic modifications also maintain the proviral latency.[36] High levels of histone deacetylases (HDACs) and histone methylases (HMTs) accumulate at the long terminal repeat (LTR) of latent HIV-1, and DNA methyltransferases lead to DNA methylation on the CpG islands, resulting in transcription silencing.

The “Shock and Kill” strategy has been considered as one of the more promising strategies for achieving potential HIV-1 cure.[37] “Shock” refers to reactivate the latently infected cells to express viral mRNA and produce viral protein through latency reversal agents (LRAs). Then killing of cells with reactivated virus will be achieved by enhanced cytotoxic effect and immune clearance, or other additional interventions. A series of LRAs have been developed from in vitro and ex vivo systems, and some of them have been tested in experimental clinical trials.[38,39] HDAC inhibitors (HDACi), like vorinostat (SAHA) and panobinostat, increase acetylation of the promoter regions including HIV-1 LTR to activate the transcription of many genes without specificity and allow for binding of NF-κB and NFAT.[40] Other kinds of chromatin-modifying enzyme inhibitors, such as histone methyltransferases inhibitors (HMTi) like BIX01294 and DNA methyltransferases inhibitors (DNMTi) like decitabine, also can activate viral gene expression but will be more effective in combination with other LRAs.[4143] Protein kinase C (PKC) agonists mainly activate NF-κB by releasing it from IκB, Which results in its translocation to the nucleus and binding at the HIV-1 LTR to promote transcription. Both prostratin and bryostatin-1 are categorized to PKC agonists.[44,45] P-TEFb activator like JQ-1 can activate HIV-1 transcription independent of NF-κB via releasing active form of P-TEFb, which subsequently can be recruited to HIV-1 Tat and promote viral transcriptional elongation via RNA polymerase II.[46] What is more, recent studies have presented a wide range of LRAs by other mechanisms, including toll-like receptor (TLR) agonists GS-9620,[47] non-canonical NF-κB agonists and second mitochondrial-derived activator of caspases (SMAC) mimetic AZD5582,[48] proteasome inhibitor thiostrepton,[49] and so on.

The multifactorial mechanisms underlying HIV-1 latency and the diverse profile of latency reversing agents again highlight the complex heterogeneity of HIV-1 latent reservoir. Although a series of LRAs have shown robust activities in reactivating latent viruses in vitro or ex vivo, no apparent reduction of latent reservoir has been reported in clinical trials.[50] Even the unspecific global T cell activator phytohemagglutinin (PHA) was not sufficient to reactivate all proviruses after a single dose.[51] Therefore, other factors may also play a role in controlling viral latency and reactivation.

Diverse HIV-1 integration sites add to the heterogeneity of latent reservoir

After integration into the host genome, HIV-1 relies on the host transcriptional machinery to replicate itself, so the microenvironments around the viral integration site largely determine the transcriptional outcome of the provirus. As a result, it is entirely possible that HIV-1 integration sites significantly contribute to the heterogeneity of HIV-1 latent reservoir. Transportation and integration of HIV-1 genome are achieved by a nucleoprotein structure called pre-integration complex (PIC), which contains viral integrase, HIV-1 DNA, and other components. PIC interacts with the host chromatin-binding protein Lens epithelium-derived growth factor (LEDGF)/p75 to target the host genome.[52,53] The process of integration is not completely random, because the LEDGF/p75 promotes HIV-1 DNA to preferentially integrate into transcriptional unit of active genes.[54] But in fact, the latent and replication-competent provirus can also be found at regions with low levels of transcription.[55] The heterogeneity of the integration sites is also illustrated through that they are distributed not only in structurally relaxed euchromatin, but also in heterochromatin with great difference in chromatin accessibility.[56] After all, integration of provirus at actively transcribed sites promotes an efficient transcription of viral proteins, while integration in the site with low level of transcription would delay viral expression or result in latency.[57] Besides its location, another important factor is the orientation of the provirus relative to the host gene. Generally, when HIV-1 provirus is integrated into active genes, parallel orientations could increase the HIV-1 gene expression by >10-fold, while antiparallel orientations reduce the viral transcriptional level by 4-fold.[58]

Maldarelli et al[59] developed a method to detect HIV-1 integration sites in latently infected cells from five patients under long-term ART, and a total of 2410 different integration sites were recovered. Surprisingly, approximately 70% of the locations targeted more than once were known to be directly related to cell growth or mitosis. Another research conducted by Wagner et al[60] also found that HIV-1 integration sites were enriched in cancer-associated genes. Both studies have found many identical integration sites existing in multiple cells within each patient, suggesting that a large population of latently infected cells were generated and maintained through clonal expansion. Also, these observations indicated that the effect of integration sites in these cells could contribute to their long-term survival.

Due to the numerous diversities of HIV-1 integration sites, evaluation of latency reversal agents should not be limited to their efficacy to reactivate viral transcription, but should also consider their abilities to target proviruses with diverse integration sites [Figure 1]. The potential mechanism of HIV-1 integration sites selection still needs further exploration, and whether certain integration sites can promote latency establishment or clonal expansion of the infected cells remain a critical question that need to be solved in the future.

Figure 1.

Figure 1

The evaluation of latency reversal activities of LRAs should consider both efficacy and functional range targeting latently infected cells with diverse integration sites. (A) A schematic diagram of a latently infected cell being reactivated to produce HIV-1 transcripts and viral proteins. (B) After HIV-1 latent reservoirs are “shocked” by LRAs, some LRAs show wide functional range with lower efficacy (LRA a), while some show higher efficacy but more limited functional range (LRA b). LRA: latency reversal agent; HIV-1: human immunodeficiency virus-1.

Potential mechanisms contributing to the clonal expansion of HIV-1 latent reservoirs

Since the phenomenon of reservoir clonal expansion was first reported in 2014,[59,60] it has been confirmed by a series of studies[61,62] and widely accepted as an important mechanism maintaining the long-term stability of viral reservoirs. It has been estimated that more than half of the latently infected cells undergo clonal expansion.[62] Our recent study identified CD161+ CD4+ T cells as an important compartment of the HIV-1 latent reservoir containing a significant number of proviruses with identical sequences, also confirming the existence of clonal expansion.[63] Therefore, deciphering the elements driving clonal expansion will be significant to counteract the maintenance of the viral reservoir, and to develop novel strategies for AIDS functional cure.

So far, three possible mechanisms of the clonal expansion have been proposed: (1) antigen-driven proliferation. Evidence from infected individuals suggests that the reservoir cells can expand under the induction of tumor antigens[64] or co-infected viral antigens like cytomegalovirus, Epstein Barr virus.[65] In this situation, HIV-1-infected, antigen specific CD4+ T cells are likely to undergo expansion, but they also might be reactivated to express HIV-1 protein, resulting in cytotoxic effect or immune clearance. Therefore, the expanded clones would fluctuate over time.[66] (2) Homeostatic proliferation mediated by cytokines. Studies have found that signals from IL-7 and IL-15 in infected people can promote the homeostatic proliferation of memory CD4+ T cells with latent HIV-1 provirus.[13] The effect of cytokines driving homeostatic proliferation is relatively steady, because IL-7 and IL-15 does not induce viral antigen expression and thus expanded cells can evade immune surveillance.[67] (3) Integration site-dependent proliferation. The methods of integration sites detection identify both the integration site and the number of clonally expanded HIV-1-infected cells [Figure 2]. As reported by Maldarelli et al[59] and Wagner et al,[60] repeated integration sites tend to be localized to regions that related to cell growth or cancer-associated genes. Integration site-dependent clonal expansion of HIV-1-infected cells is likely driven by increasing transcription of a pro-proliferation gene or losing function of a tumor suppressor gene.[68,69] Integration site-dependent proliferation will gradually increase over time for it is out of immune control, but in a relatively low rate that may take years to be observed. Finally, all these mechanisms could work at the same time to drive the proliferation of HIV-1-infected cells.

Figure 2.

Figure 2

Principle of HIV-1 integration sites detection. (A) Genomic DNA of resting CD4+ T cells were extracted. (B) DNA fragments were randomly interrupted, and fragments possess both HIV-1 genome and human genome are targeted. Different cells with the same integration sites have diverse break point. (C) End repair and LTR digestion were conducted on the fragments, and several rounds of PCR were applied for targeted amplification. (D) Fragments from former step were sequenced and mapped to human genome to identify various integration sites. For the same integration sites, the variety of fragments with different length represents the number of clonally expanded HIV-1-infected cells. LTR: long terminal repeat; PCR: Polymerase chain reaction.

Conclusion and Perspective

The tremendous heterogeneity of HIV-1 latent reservoir has certainly posted huge challenges to the AIDS functional cure efforts. However, understanding and acknowledging this challenge will help us develop novel strategies to target the HIV-1 latent reservoir. This review has discussed several layers of heterogeneity of HIV-1 latent reservoir, including cell types, latency and reactivation, integration sites, and clonal expansion, all of which play important roles in the establishment and maintenance of HIV-1 latent reservoirs. The study of these characteristics also gives us some inspirations: when we aim to reactivate or silence the HIV-1 provirus, we should consider the effectiveness of the method we use on all or most latently infected cells in various types and subpopulations, and whether the effort of intervention can reach different tissues or anatomical compartments (sanctuary sites); at the same time, we can target the mechanism of HIV-1 integration and clonal expansion to destabilize or reduce the latent reservoir.

Funding

This work was supported by grants from the National Special Research Program of China for Important Infectious Diseases (No.2018ZX10302103 and No.2017ZX10202102), National Natural Science Foundation of China (No.81672024), Natural Science Foundation of Guangdong Province of China (No.2017A030306005) and Guangdong Innovative and Entrepreneurial Research Team Program (No.2016ZT06S638).

Conflicts of interest

None.

Footnotes

How to cite this article: Zhao JC, Deng K. Heterogeneity of HIV-1 latent reservoirs. Chin Med J 2020;133:2867–2873. doi: 10.1097/CM9.0000000000001085

References

  • 1.Chun T-W, Carruth L, Finzi D, Shen X, DiGiuseppe JA, Taylor H, et al. Quantification of latent tissue reservoirs and total body viral load in HIV-1 infection. Nature 1997; 387:183–188. doi: 10.1038/387183a0. [DOI] [PubMed] [Google Scholar]
  • 2.Finzi D, Hermankova M, Pierson T, Carruth LM, Buck C, Chaisson RE, et al. Identification of a reservoir for HIV-1 in patients on highly active antiretroviral therapy. Science 1997; 278:1295–1300. doi: 10.1126/science.278.5341.1295. [DOI] [PubMed] [Google Scholar]
  • 3.Siliciano JD, Kajdas J, Finzi D, Quinn TC, Chadwick K, Margolick JB, et al. Long-term follow-up studies confirm the stability of the latent reservoir for HIV-1 in resting CD4+ T cells. Nat Med 2003; 9:727–728. doi: 10.1038/nm880. [DOI] [PubMed] [Google Scholar]
  • 4.Crooks AM, Bateson R, Cope AB, Dahl NP, Griggs MK, Kuruc JD, et al. Precise Quantitation of the Latent HIV-1 Reservoir: Implications for Eradication Strategies. J Infect Dis 2015; 212:1361–1365. doi: 10.1093/infdis/jiv218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hutter G, Nowak D, Mossner M, Ganepola S, Mussig A, Allers K, et al. Long-term control of HIV by CCR5 Delta32/Delta32 stem-cell transplantation. N Engl J Med 2009; 360:692–698. doi: 10.1056/NEJMoa0802905. [DOI] [PubMed] [Google Scholar]
  • 6.Gupta RK, Abdul-Jawad S, McCoy LE, Mok HP, Peppa D, Salgado M, et al. HIV-1 remission following CCR5(32/(32 haematopoietic stem-cell transplantation. Nature 2019; 568:244–248. doi: 10.1038/s41586-019-1027-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Gupta RK, Peppa D, Hill AL, Gálvez C, Salgado M, Pace M, et al. Evidence for HIV-1 cure after CCR5(32/(32 allogeneic haemopoietic stem-cell transplantation 30 months post analytical treatment interruption: a case report. Lancet HIV 2020; 7:e340–e347. doi: 10.1016/s2352-3018(20)30069-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Chomont N, DaFonseca S, Vandergeeten C, Ancuta P, Sekaly RP. Maintenance of CD4+ T-cell memory and HIV persistence: keeping memory, keeping HIV. Curr Opin HIV AIDS 2011; 6:30–36. doi: 10.1097/COH.0b013e3283413775. [DOI] [PubMed] [Google Scholar]
  • 9.Shan L, Deng K, Gao H, Xing S, Capoferri AA, Durand CM, et al. Transcriptional reprogramming during effector-to-memory transition renders CD4(+) T Cells permissive for latent HIV-1 infection. Immunity 2017; 47:766–775. e763 doi: 10.1016/j.immuni.2017.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Mahnke YD, Brodie TM, Sallusto F, Roederer M, Lugli E. The who's who of T-cell differentiation: human memory T-cell subsets. Eur J Immunol 2013; 43:2797–2809. doi: 10.1002/eji.201343751. [DOI] [PubMed] [Google Scholar]
  • 11.Soriano-Sarabia N, Bateson RE, Dahl NP, Crooks AM, Kuruc JD, Margolis DM, et al. Quantitation of replication-competent HIV-1 in populations of resting CD4+ T cells. J Virol 2014; 88:14070–14077. doi: 10.1128/jvi.01900-14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.von Stockenstrom S, Odevall L, Lee E, Sinclair E, Bacchetti P, Killian M, et al. Longitudinal genetic characterization reveals that cell proliferation maintains a persistent HIV Type 1 DNA pool during effective HIV therapy. J Infect Dis 2015; 212:596–607. doi: 10.1093/infdis/jiv092. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Chomont N, El-Far M, Ancuta P, Trautmann L, Procopio FA, Yassine-Diab B, et al. HIV reservoir size and persistence are driven by T cell survival and homeostatic proliferation. Nat Med 2009; 15:893–900. doi: 10.1038/nm.1972. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Schenkel JM, Masopust D. Tissue-resident memory T cells. Immunity 2014; 41:886–897. doi: 10.1016/j.immuni.2014.12.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Cantero-Pérez J, Grau-Expósito J, Serra-Peinado C, Rosero DA, Luque-Ballesteros L, Astorga-Gamaza A, et al. Resident memory T cells are a cellular reservoir for HIV in the cervical mucosa. Nat Commun 2019; 10:4739.doi: 10.1038/s41467-019-12732-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Sun H, Kim D, Li X, Kiselinova M, Ouyang Z, Vandekerckhove L, et al. Th1/17 polarization of CD4 T cells supports HIV-1 persistence during antiretroviral therapy. J Virol 2015; 89:11284–11293. doi: 10.1128/jvi.01595-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Gosselin A, Wiche Salinas TR, Planas D, Wacleche VS, Zhang Y, Fromentin R, et al. HIV persists in CCR6+CD4+ T cells from colon and blood during antiretroviral therapy. AIDS (London, England) 2017; 31:35–48. doi: 10.1097/qad.0000000000001309. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Lee GQ, Orlova-Fink N, Einkauf K, Chowdhury FZ, Sun X, Harrington S, et al. Clonal expansion of genome-intact HIV-1 in functionally polarized Th1 CD4+ T cells. J Clin Invest 2017; 127:2689–2696. doi: 10.1172/jci93289. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Dunay GA, Solomatina A, Kummer S, Hüfner A, Bialek JK, Eberhard JM, et al. Assessment of the HIV-1 reservoir in CD4+ regulatory T cells by a Droplet Digital PCR based approach. Virus Res 2017; 240:107–111. doi: 10.1016/j.virusres.2017.07.008. [DOI] [PubMed] [Google Scholar]
  • 20.Dobrowolski C, Valadkhan S, Graham AC, Shukla M, Ciuffi A, Telenti A, et al. Entry of polarized effector cells into quiescence forces HIV latency. mBio 2019; 10:e00337-19.doi: 10.1128/mBio.00337-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Aid M, Dupuy FP, Moysi E, Moir S, Haddad EK, Estes JD, et al. Follicular CD4 T helper cells as a major HIV reservoir compartment: a molecular perspective. Front Immunol 2018; 9:895.doi: 10.3389/fimmu.2018.00895. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Perreau M, Savoye AL, De Crignis E, Corpataux JM, Cubas R, Haddad EK, et al. Follicular helper T cells serve as the major CD4 T cell compartment for HIV-1 infection, replication, and production. J Exp Med 2013; 210:143–156. doi: 10.1084/jem.20121932. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Lindqvist M, van Lunzen J, Soghoian DZ, Kuhl BD, Ranasinghe S, Kranias G, et al. Expansion of HIV-specific T follicular helper cells in chronic HIV infection. J Clin Invest 2012; 122:3271–3280. doi: 10.1172/jci64314. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Yu F, Li Q, Chen X, Liu J, Li L, Li B, et al. X4-tropic latent HIV-1 is enriched in peripheral follicular helper T cells and is correlated with disease progression. J Virol 2020; 94:e01219-19.doi: 10.1128/jvi.01219-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Chun TW, Nickle DC, Justement JS, Meyers JH, Roby G, Hallahan CW, et al. Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy. J Infect Dis 2008; 197:714–720. doi: 10.1086/527324. [DOI] [PubMed] [Google Scholar]
  • 26.Rose R, Lamers SL, Nolan DJ, Maidji E, Faria NR, Pybus OG, et al. HIV maintains an evolving and dispersed population in multiple tissues during suppressive combined antiretroviral therapy in individuals with cancer. J Virol 2016; 90:8984–8993. doi: 10.1128/jvi.00684-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Ko A, Kang G, Hattler JB, Galadima HI, Zhang J, Li Q, et al. Macrophages but not astrocytes harbor HIV DNA in the brains of HIV-1-infected aviremic individuals on suppressive antiretroviral therapy. JNIP 2019; 14:110–119. doi: 10.1007/s11481-018-9809-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Guadalupe M, Sankaran S, George MD, Reay E, Verhoeven D, Shacklett BL, et al. Viral suppression and immune restoration in the gastrointestinal mucosa of human immunodeficiency virus type 1-infected patients initiating therapy during primary or chronic infection. J Virol 2006; 80:8236–8247. doi: 10.1128/jvi.00120-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Ganor Y, Real F, Sennepin A, Dutertre CA, Prevedel L, Xu L, et al. HIV-1 reservoirs in urethral macrophages of patients under suppressive antiretroviral therapy. Nat Microbiol 2019; 4:633–644. doi: 10.1038/s41564-018-0335-z. [DOI] [PubMed] [Google Scholar]
  • 30.Kandathil AJ, Sugawara S, Goyal A, Durand CM, Quinn J, Sachithanandham J, et al. No recovery of replication-competent HIV-1 from human liver macrophages. J Clin Invest 2018; 128:4501–4509. doi: 10.1172/jci121678. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Kruize Z, Kootstra NA. The role of macrophages in HIV-1 persistence and pathogenesis. Front Microbiol 2019; 10:2828.doi: 10.3389/fmicb.2019.02828. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Li GH, Henderson L, Nath A. Astrocytes as an HIV reservoir: mechanism of HIV infection. Curr HIV Res 2016; 14:373–381. doi: 10.2174/1570162x14666161006121455. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Mbonye U, Karn J. The molecular basis for human immunodeficiency virus latency. Annu Rev Virol 2017; 4:261–285. doi: 10.1146/annurev-virology-101416-041646. [DOI] [PubMed] [Google Scholar]
  • 34.Bosque A, Planelles V. Induction of HIV-1 latency and reactivation in primary memory CD4+ T cells. Blood 2009; 113:58–65. doi: 10.1182/blood-2008-07-168393. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Barboric M, Yik JH, Czudnochowski N, Yang Z, Chen R, Contreras X, et al. Tat competes with HEXIM1 to increase the active pool of P-TEFb for HIV-1 transcription. Nucleic Acids Res 2007; 35:2003–2012. doi: 10.1093/nar/gkm063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Mbonye U, Karn J. Transcriptional control of HIV latency: cellular signaling pathways, epigenetics, happenstance and the hope for a cure. Virology 2014; 454–455:328–339. doi: 10.1016/j.virol.2014.02.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Deeks SG. HIV: Shock and kill. Nature 2012; 487:439–440. doi: 10.1038/487439a. [DOI] [PubMed] [Google Scholar]
  • 38.Spivak AM, Planelles V. Novel latency reversal agents for HIV-1 cure. Annu Rev Med 2018; 69:421–436. doi: 10.1146/annurev-med-052716-031710. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Bashiri K, Rezaei N, Nasi M, Cossarizza A. The role of latency reversal agents in the cure of HIV: a review of current data. Immunol Lett 2018; 196:135–139. doi: 10.1016/j.imlet.2018.02.004. [DOI] [PubMed] [Google Scholar]
  • 40.Banga R, Procopio FA, Cavassini M, Perreau M. In vitro reactivation of replication-competent and infectious HIV-1 by histone deacetylase inhibitors. J Virol 2016; 90:1858–1871. doi: 10.1128/jvi.02359-15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.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. doi: 10.1074/jbc.M110.103531. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Fernandez G, Zeichner SL. Cell line-dependent variability in HIV activation employing DNMT inhibitors. Virol J 2010; 7:266.doi: 10.1186/1743-422x-7-266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Kumar A, Darcis G, Van Lint C, Herbein G. Epigenetic control of HIV-1 post integration latency: implications for therapy. Clin Epigenetics 2015; 7:103.doi: 10.1186/s13148-015-0137-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Williams SA, Chen LF, Kwon H, Fenard D, Bisgrove D, Verdin E, et al. Prostratin antagonizes HIV latency by activating NF-kappaB. Journal Biol Chem 2004; 279:42008–42017. doi: 10.1074/jbc.M402124200. [DOI] [PubMed] [Google Scholar]
  • 45.Mehla R, Bivalkar-Mehla S, Zhang R, Handy I, Albrecht H, Giri S, et al. Bryostatin modulates latent HIV-1 infection via PKC and AMPK signaling but inhibits acute infection in a receptor independent manner. PloS One 2010; 5:e11160.doi: 10.1371/journal.pone.0011160. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Bartholomeeusen K, Xiang Y, Fujinaga K, Peterlin BM. Bromodomain and extra-terminal (BET) bromodomain inhibition activate transcription via transient release of positive transcription elongation factor b (P-TEFb) from 7SK small nuclear ribonucleoprotein. J Biol Chem 2012; 287:36609–36616. doi: 10.1074/jbc.M112.410746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Tsai A, Irrinki A, Kaur J, Cihlar T, Kukolj G, Sloan DD, et al. Toll-Like receptor 7 agonist GS-9620 induces HIV expression and HIV-specific immunity in cells from HIV-infected individuals on suppressive antiretroviral therapy. J Virol 2017; 91:e02166-16.doi: 10.1128/jvi.02166-16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nixon CC, Mavigner M, Sampey GC, Brooks AD, Spagnuolo RA, Irlbeck DM, et al. Systemic HIV and SIV latency reversal via non-canonical NF-(B signalling in vivo. Nature 2020; 578:160–165. doi: 10.1038/s41586-020-1951-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Peng W, Hong Z, Chen X, Gao H, Dai Z, Zhao J, et al. Thiostrepton reactivates latent HIV-1 through the p-TEFb and NF-(B pathways mediated by heat shock response. Antimicrob Agents Chemother 2020; 64:e02328-19.doi: 10.1128/aac.02328-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Rasmussen TA, Tolstrup M, Søgaard OS. Reversal of latency as part of a cure for HIV-1. Trends Microbiol 2016; 24:90–97. doi: 10.1016/j.tim.2015.11.003. [DOI] [PubMed] [Google Scholar]
  • 51.Ho YC, Shan L, Hosmane NN, Wang J, Laskey SB, Rosenbloom DI, et al. Replication-competent noninduced proviruses in the latent reservoir increase barrier to HIV-1 cure. Cell 2013; 155:540–551. doi: 10.1016/j.cell.2013.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Hughes SH, Coffin JM. What integration sites tell us about HIV persistence. Cell Host Microbe 2016; 19:588–598. doi: 10.1016/j.chom.2016.04.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Shun MC, Raghavendra NK, Vandegraaff N, Daigle JE, Hughes S, Kellam P, et al. LEDGF/p75 functions downstream from preintegration complex formation to effect gene-specific HIV-1 integration. Genes Dev 2007; 21:1767–1778. doi: 10.1101/gad.1565107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Ciuffi A, Llano M, Poeschla E, Hoffmann C, Leipzig J, Shinn P, et al. A role for LEDGF/p75 in targeting HIV DNA integration. Nat Med 2005; 11:1287–1289. doi: 10.1038/nm1329. [DOI] [PubMed] [Google Scholar]
  • 55.Dieudonné M, Maiuri P, Biancotto C, Knezevich A, Kula A, Lusic M, et al. Transcriptional competence of the integrated HIV-1 provirus at the nuclear periphery. EMBO J 2009; 28:2231–2243. doi: 10.1038/emboj.2009.141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Carteau S, Hoffmann C, Bushman F. Chromosome structure and human immunodeficiency virus type 1 cDNA integration: centromeric alphoid repeats are a disfavored target. J Virol 1998; 72:4005–4014. doi: 10.1128/JVI.72.5.4005-4014.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Jordan A, Defechereux P, Verdin E. The site of HIV-1 integration in the human genome determines basal transcriptional activity and response to Tat transactivation. EMBO J 2001; 20:1726–1738. doi: 10.1093/emboj/20.7.1726. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Han Y, Lin YB, An W, Xu J, Yang HC, O’Connell K, et al. Orientation-dependent regulation of integrated HIV-1 expression by host gene transcriptional readthrough. Cell Host Microbe 2008; 4:134–146. doi: 10.1016/j.chom.2008.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Maldarelli F, Wu X, Su L, Simonetti FR, Shao W, Hill S, et al. HIV latency. Specific HIV integration sites are linked to clonal expansion and persistence of infected cells. Science 2014; 345:179–183. doi: 10.1126/science.1254194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Wagner TA, McLaughlin S, Garg K, Cheung CY, Larsen BB, Styrchak S, et al. HIV latency. Proliferation of cells with HIV integrated into cancer genes contributes to persistent infection. Science 2014; 345:570–573. doi: 10.1126/science.1256304. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Cohn LB, Silva IT, Oliveira TY, Rosales RA, Parrish EH, Learn GH, et al. HIV-1 integration landscape during latent and active infection. Cell 2015; 160:420–432. doi: 10.1016/j.cell.2015.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hosmane NN, Kwon KJ, Bruner KM, Capoferri AA, Beg S, Rosenbloom DI, et al. Proliferation of latently infected CD4(+) T cells carrying replication-competent HIV-1: Potential role in latent reservoir dynamics. J Exp Med 2017; 214:959–972. doi: 10.1084/jem.20170193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Li X, Liu Z, Li Q, Hu R, Zhao L, Yang Y, et al. CD161(+) CD4(+) T cells harbor clonally expanded replication-competent HIV-1 in antiretroviral therapy-suppressed individuals. mBio 2019; 10:e02121-19.doi: 10.1128/mBio.02121-19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Simonetti FR, Sobolewski MD, Fyne E, Shao W, Spindler J, Hattori J, et al. Clonally expanded CD4+ T cells can produce infectious HIV-1 in vivo. Proc Nati Acad Sci U S A 2016; 113:1883–1888. doi: 10.1073/pnas.1522675113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Henrich TJ, Hobbs KS, Hanhauser E, Scully E, Hogan LE, Robles YP, et al. Human immunodeficiency virus type 1 persistence following systemic chemotherapy for malignancy. J Infect Dis 2017; 216:254–262. doi: 10.1093/infdis/jix265. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Wang Z, Gurule EE, Brennan TP, Gerold JM, Kwon KJ, Hosmane NN, et al. Expanded cellular clones carrying replication-competent HIV-1 persist, wax, and wane. Proc Nati Acad Sci U S A 2018; 115:E2575–E2584. doi: 10.1073/pnas.1720665115. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Vandergeeten C, Fromentin R, DaFonseca S, Lawani MB, Sereti I, Lederman MM, et al. Interleukin-7 promotes HIV persistence during antiretroviral therapy. Blood 2013; 121:4321–4329. doi: 10.1182/blood-2012-11-465625. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Liu R, Simonetti FR, Ho YC. The forces driving clonal expansion of the HIV-1 latent reservoir. Virol J 2020; 17:4.doi: 10.1186/s12985-019-1276-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Wang ND, Li TS. Factors associated with the size of HIV DNA reservoir. Chin Med J 2017; 130:224–230. doi: 10.4103/0366-6999.198009. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Chinese Medical Journal are provided here courtesy of Wolters Kluwer Health

RESOURCES