Commentary on “Exosome-mediated stable epigenetic repression of HIV-1”
Editorial Commentary

Commentary on “Exosome-mediated stable epigenetic repression of HIV-1

Joseph A. Ayariga1, Qiana L. Matthews2,3

1Biomedical Engineering Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, USA; 2Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, USA; 3Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, USA

Correspondence to: Qiana L. Matthews. Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA; Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL 36104, USA. Email: qmatthews@alasu.edu.

Comment on: Shrivastava S, Ray RM, Holguin L, et al. Exosome-mediated stable epigenetic repression of HIV-1. Nat Commun 2021;12:5541.


Received: 20 December 2021; Accepted: 14 February 2022; Published: 31 March 2022.

doi: 10.21037/exrna-21-34


Extracellular vesicles (EVs) were first described in the 1980s. An area of interest in immunology is the influence of a subset of EVs (exosomes) on the course of human disease and their centrality in the anti-human immunodeficiency virus (HIV) mitigation process. In this commentary, we dissect work published by Shrivastava et al., on exosome-mediated stable epigenetic repression of HIV-1.

While the potency of some antiretroviral agents has captured the interest of researchers and clinicians (1-3), the dangers that lurks clearly in most of these drugs are related to long-term effects and cytotoxicity. For instance, hyperlipidemia, hyperglycemia, and lipodystrophy have been shown to be induced by the well-known highly active antiretroviral therapy (HAART) (3). To navigate this pathological issue, Shrivastava et al. postulated that the exosomes derived from the body which contains cellular epigenetic arsenal like DNA methyl transferase 3 alpha (DNMT3A) could on one hand eliminate the problem of cytotoxicity and on the other hand down-regulate HIV-1 transcription (4). Undoubtedly, the progression and pathogenesis of HIV is highly favored by weakening immunity of the infected individuals, hence there is the urgent need to avoid compounding HIV patients with agents and therapies that affect immunity.

Much progress has been made since the discovery and implementation of combination antiretroviral therapy (cART) against HIV infection and control, however, given the complexity of HIV infection and disease progression, new and innovative tools must be employed to navigate HIV disease and latency. In this regard, new innovative strategies have employed the use exosomes to reduce HIV infection. Khatua and group (5) demonstrated that exosomes could release proteins and thus serve as an HIV therapy (5). HIV therapies continue to progress, including promising approaches that attempt to stop a tremendous hurdle, HIV latency and re-emergency. Along these lines, Mousseau et al. employed didehydro-cortistatin A (dCA) as a potent Tat inhibitor, reducing HIV-1 transcription and preventing viral rebound. dCA activity demonstrates the potential of the “block-and-lock” functional cure approaches. However, this approach led to dCA resistant virus (6).

Hypermethylation is a well-known genetic method used against most proviruses, for instance, CpG methylation of the human T-cell leukemia virus type 1 (HTLV-1) long terminal repeat (LTR) causes proviral latency (7), the viral 5’LTR cause latency in HIV-1 infection (8). For this reason, targeted DNA methylation of the 5’LTR is a proven method, nonetheless, this method must be rigorously studied against the background of the human body to ensure its safety. Shrivastava et al. (4) intelligently established this crucial framework in this study by utilizing a humanize mouse model to recapitulate the human body mechanisms.

The ingenious fusion of zinc finger protein (ZFP362b), which targets and binds to HIV-1 LTR, to the DNA methyl transferase ensures targeted delivery and eradicates the possibility of random methylations processes that can potentiate cascades of deleterious events in the body. Such creativity has immeasurable utility in bioengineering, cancer research and more importantly and most timely, in HIV research (Figure 1).

Figure 1 Mechanism of ZFP362b-DNMT3A exosomes payload delivery system using the EXOtic technology. ZFP, zinc finger protein; DNMT3A, DNA methyl transferase 3 alpha; EXOtic, EXOsomal transfer into cells.

The findings that ZFP362b fused to DNMT3A potently and selectively repressed HIV (4) is huge to HIV vaccinologists and researchers alike (Figure 1). To the researcher what catches the eye most was their technique. Exosomes are promiscuous, they pack all kinds of “material” in them. To avoid non-specificity, they adopted the exosome packaging technology devised by Kojima et al. (9), coined “EXOtic” (EXOsomal transfer into cells) which targets the exosome markers such as CD63, Alix, and TSG101 which allows for specific recruitment and encapsulation of mRNA into exosomes. Exosomes are released from the producer cell and enter the target cells through membrane fusion (9).

EVs were first described in the 1980s, when they were found in red blood cells (10), reticulocytes (11-13), neoplastic cell lines (14). Exosomes have been considered critical to disease mitigation and progression (15,16). Exosomes are the smallest type of EVs. These endosome-derived small membrane vesicles are approximately 50–150 nm in diameter (9,17-19). Exosome are secreted by cells and released into biological fluids in all living systems (20). An area of interest in immunology is the influence of EVs and exosomes on the course of human disease. Exosomes are central to the development of HIV mitigation and progression (21-23).

The clinical relevance of this interesting work goes beyond HIV repression alone, it represents a myriad of possibilities against several other diseases and cancers. Contrary to most therapeutic systems available which are known to exacerbate the weakening of the patient’s immunity, the EXOtic delivery of HIV repressor might significantly reduce viral load in HIV patients and at the same time cause no significant side effect since exosomes are naturally produced and present in the body.

A fundamental challenge in DNA methylation approach is how to direct the methyl transferase into the nucleus initially. The investigators solved this major huddle by embedding a nuclear localization signal into the ZFP362b-DNMT3A construct. They proved via methyl DNA immunoprecipitation that their constructs; ZFP, ZPAMt, and ZKMt were localized in the nucleus and binds to HIV-LTR region. At present, it is unclear which other regulatory factors might be affected by HIV repression using ZFP, and can this technology potentially disrupt any crucial signaling pathways that hamper development and normal tissue homeostasis? What are the long-term effects? More so, we still lack throughput data on the pharmacodynamics and pharmacokinetics of exosome encapsulated agents and hence this technology suffers similar deficiency. Nonetheless, such an excellent work is highly commendable and paves the way for research into exosomes utility in disease management and more specifically, HIV therapy.


Acknowledgments

Funding: This work was funded by the National Institutes of Health (grant No. 1R15DA045564-02) and The National Science Foundation (grant No. IOS-1900377).


Footnote

Provenance and Peer Review: This article was commissioned by the editorial office, ExRNA. The article has undergone external peer review.

Conflicts of Interest: Both authors have completed the ICMJE uniform disclosure form (available at https://exrna.amegroups.com/article/view/10.21037/exrna-21-34/coif). The authors have no conflicts of interest to declare.

Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.

Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.


References

  1. Eisfeld C, Reichelt D, Evers S, et al. CSF penetration by antiretroviral drugs. CNS Drugs 2013;27:31-55. [Crossref] [PubMed]
  2. Cenderello G, De Maria A. Discordant responses to cART in HIV-1 patients in the era of high potency antiretroviral drugs: clinical evaluation, classification, management prospects. Expert Rev Anti Infect Ther 2016;14:29-40. [Crossref] [PubMed]
  3. Fantoni M, Autore C, Del Borgo C. Drugs and cardiotoxicity in HIV and AIDS. Ann N Y Acad Sci 2001;946:179-99. [Crossref] [PubMed]
  4. Shrivastava S, Ray RM, Holguin L, et al. Exosome-mediated stable epigenetic repression of HIV-1. Nat Commun 2021;12:5541. [Crossref] [PubMed]
  5. Khatua AK, Taylor HE, Hildreth JE, et al. Exosomes packaging APOBEC3G confer human immunodeficiency virus resistance to recipient cells. J Virol 2009;83:512-21. [Crossref] [PubMed]
  6. Mousseau G, Aneja R, Clementz MA, et al. Resistance to the Tat Inhibitor Didehydro-Cortistatin A Is Mediated by Heightened Basal HIV-1 Transcription. mBio 2019;10:01750-18. [Crossref] [PubMed]
  7. Koiwa T, Hamano-Usami A, Ishida T, et al. 5'-long terminal repeat-selective CpG methylation of latent human T-cell leukemia virus type 1 provirus in vitro and in vivo. J Virol 2002;76:9389-97. [Crossref] [PubMed]
  8. Trejbalová K, Kovářová D, Blažková J, et al. Development of 5' LTR DNA methylation of latent HIV-1 provirus in cell line models and in long-term-infected individuals. Clin Epigenetics 2016;8:19. [Crossref] [PubMed]
  9. Kojima R, Bojar D, Rizzi G, et al. Designer exosomes produced by implanted cells intracerebrally deliver therapeutic cargo for Parkinson's disease treatment. Nat Commun 2018;9:1305. [Crossref] [PubMed]
  10. Allan D, Thomas P, Limbrick AR. The isolation and characterization of 60 nm vesicles ('nanovesicles') produced during ionophore A23187-induced budding of human erythrocytes. Biochem J 1980;188:881-7. [Crossref] [PubMed]
  11. Harding C, Heuser J, Stahl P. Receptor-mediated endocytosis of transferrin and recycling of the transferrin receptor in rat reticulocytes. J Cell Biol 1983;97:329-39. [Crossref] [PubMed]
  12. Johnstone RM, Adam M, Hammond JR, et al. Vesicle formation during reticulocyte maturation. Association of plasma membrane activities with released vesicles (exosomes). J Biol Chem 1987;262:9412-20. [Crossref] [PubMed]
  13. Pan BT, Teng K, Wu C, et al. Electron microscopic evidence for externalization of the transferrin receptor in vesicular form in sheep reticulocytes. J Cell Biol 1985;101:942-8. [Crossref] [PubMed]
  14. Trams EG, Lauter CJ, Salem N Jr, et al. Exfoliation of membrane ecto-enzymes in the form of micro-vesicles. Biochim Biophys Acta 1981;645:63-70. [Crossref] [PubMed]
  15. Jones LB, Kumar S, Bell CR, et al. Effects of Pseudomonas aeruginosa on Microglial-Derived Extracellular Vesicle Biogenesis and Composition. Pathogens 2019;8:297. [Crossref] [PubMed]
  16. Ellwanger JH, Veit TD, Chies JAB. Exosomes in HIV infection: A review and critical look. Infect Genet Evol 2017;53:146-54. [Crossref] [PubMed]
  17. Moldovan L, Batte K, Wang Y, et al. Analyzing the circulating microRNAs in exosomes/extracellular vesicles from serum or plasma by qRT-PCR. Methods Mol Biol 2013;1024:129-45. [Crossref] [PubMed]
  18. Bhatnagar S, Schorey JS. Exosomes released from infected macrophages contain Mycobacterium avium glycopeptidolipids and are proinflammatory. J Biol Chem 2007;282:25779-89. [Crossref] [PubMed]
  19. Jones LB, Bell CR, Bibb KE, et al. Pathogens and Their Effect on Exosome Biogenesis and Composition. Biomedicines 2018;6:79. [Crossref] [PubMed]
  20. Rilla K, Mustonen AM, Arasu UT, et al. Extracellular vesicles are integral and functional components of the extracellular matrix. Matrix Biol 2019;75-76:201-19. [Crossref] [PubMed]
  21. Chen J, Li C, Li R, et al. Exosomes in HIV infection. Curr Opin HIV AIDS 2021;16:262-70. [PubMed]
  22. Sims B, Farrow AL, Williams SD, et al. Role of TIM-4 in exosome-dependent entry of HIV-1 into human immune cells. Int J Nanomedicine 2017;12:4823-33. [Crossref] [PubMed]
  23. Sims B, Farrow AL, Williams SD, et al. Tetraspanin blockage reduces exosome-mediated HIV-1 entry. Arch Virol 2018;163:1683-9. [Crossref] [PubMed]
doi: 10.21037/exrna-21-34
Cite this article as: Ayariga JA, Matthews QL. Commentary on “Exosome-mediated stable epigenetic repression of HIV-1”. ExRNA 2022;4:8.

Download Citation