Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Letter
  • Published:

HIV persistence in tissue macrophages of humanized myeloid-only mice during antiretroviral therapy

Abstract

Despite years of fully suppressive antiretroviral therapy (ART), HIV persists in its hosts and is never eradicated. One major barrier to eradication is that the virus infects multiple cell types that may individually contribute to HIV persistence. Tissue macrophages are critical contributors to HIV pathogenesis1,2,3; however, their specific role in HIV persistence during long-term suppressive ART has not been established4,5,6. Using humanized myeloid-only mice (MoM), we demonstrate that HIV infection of tissue macrophages is rapidly suppressed by ART, as reflected by a rapid drop in plasma viral load and a dramatic decrease in the levels of cell-associated viral RNA and DNA. No viral rebound was observed in the plasma of 67% of the ART-treated animals at 7 weeks after ART interruption, and no replication-competent virus was rescued from the tissue macrophages obtained from these animals. In contrast, in a subset of animals (33%), a delayed viral rebound was observed that is consistent with the establishment of persistent infection in tissue macrophages. These observations represent the first direct evidence, to our knowledge, of HIV persistence in tissue macrophages in vivo.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Viral suppression, persistence, and rebound induced by structured ART interruption in BLT mice.
Figure 2: ART rapidly suppresses viral replication in MoM.
Figure 3: HIV persistence in tissue macrophages during ART.

Similar content being viewed by others

Accession codes

Primary accessions

GenBank/EMBL/DDBJ

Referenced accessions

NCBI Reference Sequence

References

  1. Kumar, A. & Herbein, G. The macrophage: a therapeutic target in HIV-1 infection. Mol. Cell. Ther. 2, 10 (2014).

    Article  Google Scholar 

  2. Koppensteiner, H., Brack-Werner, R. & Schindler, M. Macrophages and their relevance in human immunodeficiency virus type I infection. Retrovirology 9, 82 (2012).

    Article  CAS  Google Scholar 

  3. Campbell, J.H., Hearps, A.C., Martin, G.E., Williams, K.C. & Crowe, S.M. The importance of monocytes and macrophages in HIV pathogenesis, treatment, and cure. AIDS 28, 2175–2187 (2014).

    Article  CAS  Google Scholar 

  4. Sattentau, Q.J. & Stevenson, M. Macrophages and HIV-1: an unhealthy constellation. Cell Host Microbe 19, 304–310 (2016).

    Article  CAS  Google Scholar 

  5. Collman, R.G., Perno, C.F., Crowe, S.M., Stevenson, M. & Montaner, L.J. HIV and cells of macrophage/dendritic lineage and other non–T cell reservoirs: new answers yield new questions. J. Leukoc. Biol. 74, 631–634 (2003).

    Article  CAS  Google Scholar 

  6. Gavegnano, C. & Schinazi, R.F. Antiretroviral therapy in macrophages: implication for HIV eradication. Antivir. Chem. Chemother. 20, 63–78 (2009).

    Article  CAS  Google Scholar 

  7. Honeycutt, J.B. et al. Macrophages sustain HIV replication in vivo independently of T cells. J. Clin. Invest. 126, 1353–1366 (2016).

    Article  Google Scholar 

  8. Denton, P.W. et al. Systemic administration of antiretrovirals prior to exposure prevents rectal and intravenous HIV-1 transmission in humanized BLT mice. PLoS One 5, e8829 (2010).

    Article  Google Scholar 

  9. Wahl, A. et al. Human breast milk and antiretrovirals dramatically reduce oral HIV-1 transmission in BLT humanized mice. PLoS Pathog. 8, e1002732 (2012).

    Article  CAS  Google Scholar 

  10. Olesen, R., Wahl, A., Denton, P.W. & Garcia, J.V. Immune reconstitution of the female reproductive tract of humanized BLT mice and their susceptibility to human immunodeficiency virus infection. J. Reprod. Immunol. 88, 195–203 (2011).

    Article  CAS  Google Scholar 

  11. Melkus, M.W. et al. Humanized mice mount specific adaptive and innate immune responses to EBV and TSST-1. Nat. Med. 12, 1316–1322 (2006).

    Article  CAS  Google Scholar 

  12. Denton, P.W. et al. Generation of HIV latency in humanized BLT mice. J. Virol. 86, 630–634 (2012).

    Article  CAS  Google Scholar 

  13. Denton, P.W. et al. Targeted cytotoxic therapy kills persisting HIV infected cells during ART. PLoS Pathog. 10, e1003872 (2014).

    Article  Google Scholar 

  14. Thompson, M.A. et al. Antiretroviral treatment of adult HIV infection: 2012 recommendations of the International Antiviral Society–USA panel. J. Am. Med. Assoc. 308, 387–402 (2012).

    CAS  Google Scholar 

  15. Perelson, A.S., Neumann, A.U., Markowitz, M., Leonard, J.M. & Ho, D.D. HIV-1 dynamics in vivo: virion clearance rate, infected cell life-span, and viral generation time. Science 271, 1582–1586 (1996).

    Article  CAS  Google Scholar 

  16. Ho, D.D. et al. Rapid turnover of plasma virions and CD4 lymphocytes in HIV-1 infection. Nature 373, 123–126 (1995).

    Article  CAS  Google Scholar 

  17. Wei, X. et al. Viral dynamics in human immunodeficiency virus type 1 infection. Nature 373, 117–122 (1995).

    Article  CAS  Google Scholar 

  18. Calin, R. et al. Treatment interruption in chronically HIV-infected patients with an ultralow HIV reservoir. AIDS 30, 761–769 (2016).

    Article  CAS  Google Scholar 

  19. Shultz, L.D., Ishikawa, F. & Greiner, D.L. Humanized mice in translational biomedical research. Nat. Rev. Immunol. 7, 118–130 (2007).

    Article  CAS  Google Scholar 

  20. Igarashi, T. et al. Macrophage are the principal reservoir and sustain high virus loads in rhesus macaques after the depletion of CD4+ T cells by a highly pathogenic simian immunodeficiency virus/HIV type 1 chimera (SHIV): implications for HIV-1 infections of humans. Proc. Natl. Acad. Sci. USA 98, 658–663 (2001).

    Article  CAS  Google Scholar 

  21. Micci, L. et al. CD4 depletion in SIV-infected macaques results in macrophage and microglia infection with rapid turnover of infected cells. PLoS Pathog. 10, e1004467 (2014).

    Article  Google Scholar 

  22. Avalos, C.R. et al. Quantitation of productively infected monocytes and macrophages of simian immunodeficiency virus–infected macaques. J. Virol. 90, 5643–5656 (2016).

    Article  CAS  Google Scholar 

  23. Hansen, S.G. et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 473, 523–527 (2011).

    Article  CAS  Google Scholar 

  24. Hansen, S.G. et al. Immune clearance of highly pathogenic SIV infection. Nature 502, 100–104 (2013).

    Article  CAS  Google Scholar 

  25. Sung, J.A. et al. Expanded cytotoxic T-cell lymphocytes target the latent HIV reservoir. J. Infect. Dis. 212, 258–263 (2015).

    Article  CAS  Google Scholar 

  26. Archin, N.M. et al. Valproic acid without intensified antiviral therapy has limited impact on persistent HIV infection of resting CD4+ T cells. AIDS 22, 1131–1135 (2008).

    Article  CAS  Google Scholar 

  27. Spina, C.A. 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. 9, e1003834 (2013).

    Article  Google Scholar 

  28. Prochazka, M., Gaskins, H.R., Shultz, L.D. & Leiter, E.H. The nonobese diabetic scid mouse: model for spontaneous thymomagenesis associated with immunodeficiency. Proc. Natl. Acad. Sci. USA 89, 3290–3294 (1992).

    Article  CAS  Google Scholar 

  29. Hill, A.L. et al. Real-time predictions of reservoir size and rebound time during antiretroviral therapy interruption trials for HIV. PLoS Pathog. 12, e1005535 (2016).

    Article  Google Scholar 

  30. Steingrover, R. et al. HIV-1 viral rebound dynamics after a single treatment interruption depends on time of initiation of highly active antiretroviral therapy. AIDS 22, 1583–1588 (2008).

    Article  CAS  Google Scholar 

  31. Persaud, D. et al. Absence of detectable HIV-1 viremia after treatment cessation in an infant. N. Engl. J. Med. 369, 1828–1835 (2013).

    Article  CAS  Google Scholar 

  32. Henrich, T.J. et al. Long-term reduction in peripheral blood HIV type 1 reservoirs following reduced-intensity conditioning allogeneic stem cell transplantation. J. Infect. Dis. 207, 1694–1702 (2013).

    Article  CAS  Google Scholar 

  33. Nath, A. & Clements, J.E. Eradication of HIV from the brain: reasons for pause. AIDS 25, 577–580 (2011).

    Article  Google Scholar 

  34. Joseph, S.B., Arrildt, K.T., Sturdevant, C.B. & Swanstrom, R. HIV-1 target cells in the CNS. J. Neurovirol. 21, 276–289 (2015).

    Article  CAS  Google Scholar 

  35. Strain, M.C. et al. Highly precise measurement of HIV DNA by droplet digital PCR. PLoS One 8, e55943 (2013).

    Article  CAS  Google Scholar 

  36. Honeycutt, J.B. et al. HIV-1 infection, response to treatment and establishment of viral latency in a novel humanized T cell–only mouse (TOM) model. Retrovirology 10, 121 (2013).

    Article  Google Scholar 

  37. Zhang, D., Fan, C., Zhang, J. & Zhang, C.H. Nonparametric methods for measurements below detection limit. Stat. Med. 28, 700–715 (2009).

    Article  Google Scholar 

  38. Lafleur, B. et al. Statistical methods for assays with limits of detection: serum bile acid as a differentiator between patients with normal colons, adenomas, and colorectal cancer. J. Carcinog. 10, 12 (2011).

    Article  Google Scholar 

  39. Gillespie, B.W. et al. Estimating population distributions when some data are below a limit of detection by using a reverse Kaplan–Meier estimator. Epidemiology 21 (Suppl. 4), S64–S70 (2010).

    Article  Google Scholar 

Download references

Acknowledgements

We thank N. Archin (University of North Carolina at Chapel Hill) for providing the CD8+-depleted feeder cells for in vitro viral outgrowth analysis; J. Kappes and C. Ochsenbauer (University of Alabama at Birmingham) for providing CH040 (11740) via the AIDS Research and Reference Reagent Program; R. Swanstrom (University of North Carolina at Chapel Hill) for providing 4013-env for use in our studies; J. Clements, R. Siliciano, and D. Margolis for their intellectual input in these studies; M. Cohen, B. Haynes, S. Lemon, A. Wahl, and M. Kovarova for their comments and suggestions regarding this manuscript; and former and current members of the Garcia laboratory and the husbandry technicians at the UNC Division of Laboratory Animal Medicine for their assistance. J.V.G. was supported by grants from the National Institute of Mental Health (MH-108179) and the National Institute of Allergy and Infectious Diseases (NIAID) (AI-111899) and by the UNC CFAR (P30 AI050410). Research reported in this publication was also supported by CARE, a Martin Delaney Collaboratory, the National Institute of Allergy and Infectious Diseases (NIAID), the National Institute of Neurological Disorders and Stroke (NINDS), the National Institute on Drug Abuse (NIDA), and the National Institute of Mental Health (NIMH) of the National Institutes of Health, grant number 1UM1AI126619-01 (J.V.G. and D.D.R.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health. J.B.H. was supported in part by the Virology Training Grant (T32 AI-007419). R.M.R. was supported by the NIAID (R01 AI-104373). D.D.R. was also supported by the BEAT-HIV Delaney Collaboration (1UM1AI126620), the UCSD CFAR (AI306214), the Department of Veterans Affairs, and the James B. Pendleton Charitable Trust. The funders had no part in study design, data collection or analysis, decision to publish, or preparation of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

J.B.H. provided the experimental design, collected and processed blood and tissue samples from mice, prepared viral stocks, performed viral inoculations, administered ART, and performed flow cytometric analyses. W.O.T. processed blood and tissue samples from mice, prepared the injectable ART formulation, and administered ART. C.E.B. provided RNA and DNA analysis, prepared the injectable ART formulation, and processed tissue samples. R.A.C. performed immunohistochemical analysis of tissues. S.M.L. and D.D.R. performed the integrated DNA analysis and analyzed the results. J.B.H., R.M.R., Y.C., and M.G.H. analyzed the data and wrote the manuscript. J.V.G. conceived the study, designed and coordinated the study, and wrote the manuscript.

Corresponding author

Correspondence to J Victor Garcia.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Figures and Tables

Supplementary Figures 1–5 & Supplementary Tables 1,2. (PDF 1885 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Honeycutt, J., Thayer, W., Baker, C. et al. HIV persistence in tissue macrophages of humanized myeloid-only mice during antiretroviral therapy. Nat Med 23, 638–643 (2017). https://doi.org/10.1038/nm.4319

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4319

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research