Untangling the role of the microbiome across the stages of HIV disease

INTRODUCTION

Anatomic sites exposed to the environment are colonized with bacteria, eukaryotes, and viruses, cumulatively referred to as the microbiome. The microbiome is diverse, and the composition widely dependent upon the unique environmental factors present across the body. A millennium of coevolution with the host has led to a predominantly symbiotic relationship, with several microbiome-produced metabolites benefitting the host [1,2▪]. Alterations to the composition of the gastrointestinal (GI) tract microbiome have been associated with several disease states [3] and are considered important contributors to progressive infection in people with HIV (PWH) and Simian Immunodeficiency Virus (SIV)-infected Asian macaque nonhuman primates (NHPs). Here, we review recent work on the role of the microbiome composition on HIV and SIV acquisition and disease progression (Fig. 1). 

FB1Box 1:

no caption available

F1FIGURE 1:

The primate microbiome is a diverse community of bacterial, eukaryotic, and viral constituents residing in the gastrointestinal lumen (as shown) and other external interfaces. Antigenic and metabolic stimulation by the microbiome may prime vaccine outcomes, alter susceptibility to lentiviral acquisition, modulate lentiviral disease progression, and fluctuate during postexposure prophylaxes. These interventions and disease states may also alter homeostasis of the healthy microbiome. A detailed understanding of the mechanisms underpinning these dynamic relationships will inform novel, improved therapeutic interventions. 1. Corley MJ, Sacdalan C, Pang APS, et al. Abrupt and altered cell-type specific DNA methylation profiles in blood during acute HIV infection persists despite prompt initiation of ART. PLoS Pathog 2021;17:e1009785.

Although experiments involving animal models contribute significantly to HIV research, questions remain regarding the usefulness of these models to microbiome-related disease outcomes. Major advantages of animal models involve longitudinal potential and the ability to control for variables such as diet, antibiotic use, and behaviorally-associated risk factors – parameters that influence the composition of the GI tract microbiome irrespective of infection. Animal models also provide the ability to analyze anatomic sites that can only rarely be obtained from humans. On the other hand, there exists species variability in the composition of the microbiome in these models as compared to human participants [4] and these models are incapable of recapitulating the complex variables that contribute to microbiome individuality across diverse human populations. These limitations suggest that animal models should complement biomedical research, providing mechanistic insights and a framework against which therapeutic interventions can be tested prior to clinical implementation.

Approaches for measurement of the microbiome generally fall into three broad categories – those that involve nucleic acid analysis, those that involve mass spectroscopic and/or proteomic analysis, and those that involve culture analysis [5] – each methodology having its own strength. Nucleic acid analysis is, by far, the most common modality. Sequencing the DNA of bacterial 16S rRNA is robust, bioinformatic pipelines for analysis are well accepted, and bacterial taxa within the microbiota are well annotated. However, 16S analysis identifies only the bacterial component of the microbiome and does not directly provide details related to the functionality. As compared to 16S analysis, metagenomic analysis of microbiome-derived DNA or RNA adds significant advantage in that it is unbiased, broadens bacterial assignment, captures nonbacterial taxa, and permits more robust functional inference. To best understand the metabolic activity of the microbiome, however, metabolomic and proteomic analyses are also required [6].

Metabolites produced by the microbiome disseminate systemically, calibrating host immunity beyond antigenic interface. One of the most commonly studied microbiome-derived metabolites is trimethylamine N-oxide (TMAO), which contributes to cardiovascular disease through currently unknown mechanisms [7,8]. Too, tryptophan catabolites have been associated with HIV disease and may be related to tryptophan metabolizing bacterial taxa [9], and short chain fatty acid (SCFA) production by bacteria represents a significant source of energy for the host [10]. A mechanistic determination of the contribution of nonhost metabolites to health and disease represents an important but underexplored area of research.

MICROBIOME-ASSOCIATED DETERMINANTS OF HIV ACQUISITION

Understanding how the microbiome contributes to the risk of sexually transmitted HIV infections is an essential next step in ending the HIV epidemic. Indeed, increased risk of HIV acquisition in heterosexual women is strongly associated with the presence of a Lactobacillus nondominant vaginal microbiome [11], and in men who have sex with men (MSM) with increased intestinal Prevotella:Bacteroides[12]. Although initial reports describing these findings were cross-sectional, recent retrospective analyses and models have strengthened and refined these associations. As described by Fulcher et al.[13▪▪], seroconversion in at-risk MSM was found to associate with unique microbiome-associated features. Compared to similarly at-risk individuals that did not seroconvert over the study period, individuals that seroconverted had decreased frequencies of several Bacteroides species and increased serum inflammatory cytokines and bioactive lipids preceding HIV acquisition. Similarly, Sui et al.[14] observed unexpected variation in cohort susceptibility to low-dose intrarectal SIV in the rhesus macaque model. Here, susceptibility to SIV acquisition was associated with frequencies of rectal CCR5+Ki67+ CD4+ T-cells and in turn, fecal Prevotella:Bacteroides ratios.

To experimentally determine whether a dysbiotic microbiome might contribute to increased susceptibility, we recently utilized the antibiotic vancomycin to induce dysbiosis in rhesus macaques [15▪▪]. As predicted, we observed that pronounced dysbiosis was associated with an increase in susceptibility to low-dose intrarectal SIV infection. Surprisingly, however, increased susceptibility was not associated with direct measures of the microbiome but rather, with measures of perturbed antimicrobial immunity– namely, reduced rectal TH17 and TH22 frequencies and increased rectal PLA2G2A, GZMB, LTF, DUOX2, and SUCNR1 transcripts. Collectively, these findings suggest that microbiome-responsive features in host immunity – more so than specific taxa – tune host susceptibility to rectal HIV infection. It remains unclear whether perturbations of the preinfection microbiome alone might influence disease progression beyond sexual acquisition. The outcome of experimental studies assessing these relationships will be important for the assessing the long-term potential of preexposure prophylaxes or repeated antibiotic exposure prior to sexual acquisition.

VACCINATING FROM WITHIN: COMMENSALS AND THE PRE-EXISTING REPERTOIRE

Although vaccine development has long harnessed adjuvants to stimulate pathogen-associated molecular pattern receptors [16], only recently have findings described the role of individuals’ microbiomes in vaccine outcomes. The administration of broad-spectrum antibiotics to individuals prior to seasonal influenza vaccination impairs the generation of antibody responses in subjects with low preexisting antibody titers in addition to enhancing inflammatory signatures [17]. These perturbations have the potential both to negate protective immunity and to alter the efficacy of functional-derived immunity. Lessons learned from studying the role of the microbiome in HIV vaccine development may lead to the identification of novel modalities and strategies.

Eliciting an improved antibody response to HIV envelope is considered the most probable target for generating protective immunity due to the early emergence of anti-Env antibodies after HIV infection. Previous work has described epitope mimicry between gp41 and commensal bacterial antigens [18,19]. A recent publication by Mayer-Blackwell et al.[20▪] reports that from a global survey of 1470 HIV Vaccine Trials Network (HVTN) participants, 58% of prevaccinated, unexposed participants had detectable anti-gp41 IgG titers – with large geographic variation – whereas anti-gp120 immunoglobulin G (IgG) was seen only in 1.5% of participants. Using prevaccination fecal samples, these authors were able to correlate several taxa of the order Eubacteriales with preexisting antigp41 IgG levels. Importantly, these authors noted a strong correlation between the magnitude of pre versus postvaccination antigp41 IgG levels, and linear epitope mapping further revealed a focusing of the gp41 response, suggestive of a recall response [18].

As naturally occurring antigp120 envelope responses expand too late in infection to effectively clear HIV [21], understanding whether and how preexisting antigp120 antibodies are generated will guide vaccine-elicited expansion. In infant rhesus macaques immunized with varied gp120 protein-based formulations, Jiang et al.[22] observed that that age was the biggest driver of antigp120 IgG plasma levels variation in their study. To determine whether the developing microbiome might prime gp120 immunity, the authors surveyed the fecal microbiome and observed that vaccination induced specific shifts in Sutterella and Rodentibacter species, which further correlated with antigp120 IgG plasma levels across modalities. From metagenomic inference analyses, the authors identified that SCFA and bile acid (BA) gene abundance increased in tandem with Suterella frequencies and correlated with antigp120 IgG, suggesting that targeting SCFA and BA pathways may improve gp120 vaccine efficacy.

Antibodies against the conserved CD4-induced (CD4i) epitope work cooperatively with CD4 binding-site antibodies to potently inhibit HIV infection in vitro[23,24]. It remains unclear whether the brief conformational exposure of CD4i is sufficient to account for estimated anti-CD4i antibody titers in vivo. To ascertain whether the antigenic identity of CD4i antibodies may be commensally derived, Biswas et al.[25] passively immunized mice with CD4i antibodies 4E9C and 916B2, and isolated antiidiotypic antibodies (aID mAbs). Using these aID to a random 12-mer phage library, these authors identified consensus peptide sequences by homology. Of aID found to target microbial-associated sequences, the authors found epitope homology with Nocardioidaceae and Vibrio vulnificus sequences. Using matching, synthesized peptides, these authors demonstrated that a single peptide recognized by Vibrio vulnificus reacted to previously identified CD4i mAbs 4E9C, 17b, and 1F4F and to several of their identified aID. Epitopes encoded by this and other cross-reactive bacterium may be useful as a mimetic to prime CD4i in vaccine strategies [24] or in the purposeful design of oral probiotics [26,27] to boost vaccine efficacy.

ANTIRETROVIRALS… ANTIVIRAL OR ANTIBACTERIAL?

Potential roles for antiretrovirals (ARVs) in modulating the intestinal microbiome in PWH have been widely described [28]. Unlike with HIV infection - where the date of acquisition is often unknown – the introduction of ARVs is a chronologically definable event and samples from stage-matched individuals show commonalities in their microbiome and microbial-translocation associated markers [29,30]. Too, the introduction of ARVs in SIV-infected rhesus macaque models induces immediate effects on the intestinal microbiome [31,32]. However, changes in the microbiome are neither uniformly observed nor categorically similar after lentiviral infection, suggesting that further research is warranted.

Defining an ‘ARV-associated microbiome’ is complicated by the use of varying ARV regimens across individuals [28]. As characterized by Pinto-Cardoso et al.[33], a cross-sectional examination of the fecal microbiomes of ARV-treated patients reveals that although individuals on long-term ART collectively have distinct diversity measures as compared to uninfected individuals, there remain modest but appreciable variations in measures of the microbiome and translocation markers between patients on differing regimens. Too, Hanttu et al.[34] reported increased alpha diversity in individuals switching from an efavirenz- to a protease inhibitor-based regimen.

Some of the confounding effects of cocktail regimens can be resolved by testing the effects of ARVs – individually or in combination – on bacterial taxa in vitro. A methodical survey of over 1000 marketed drugs against 40 gut bacterial strains revealed that 27% of nonantibiotic drugs affected the growth of at least one taxon in vitro[35]. To assess the effects of HIV ARVs more specifically on commensal taxa, Rubio-Garcia et al. cultured 16 ARVs with gut and vaginal human commensal bacteria in vitro and showed that five have antibacterial activity [36], expanding on previous reports [37]. Additionally, Wallace et al. explored the possibility that the prolonged use of ARVs may contribute to antiviral resistance in susceptible bacterial taxa [38▪]. In addition to developing antiviral resistance against the primary ARV, several taxa concerningly developed cross-resistance to other antivirals or, antibacterial resistance. It remains to be seen whether similar findings are observed in patients receiving long term ARVs as either post or preexposure prophylaxes.

Despite observed sensitivities of individual bacterial taxa to antivirals in vitro, we previously demonstrated that short courses of ARVs in healthy rhesus macaques in vivo led only to modest perturbations in the intestinal microbiome and intestinal immunity [39]. However, the administration of ARVs led to a generalizable instability of the microbiome, a measure which has conceptually been observed in other studies assessing the microbiome:ARV interface in vivo[40]. It remains unclear whether ARV-associated bacterial perturbations stem from lentiviral-associated mucosal pathologies, direct antibacterial activity, or targeting of nonbacterial components of the microbiome, as further discussed below.

HARNESSING THE POWER OF THE MICROBIOME

Therapeutics designed to improve the intestinal microbiome have gained favor as a way to reduce disease progression or to boost immune reconstitution after ARV therapy. As covered more extensively in this issue, wholesale efforts to manipulate the microbiome in HIV and SIV infection include probiotic regimens [41–45] and fecal microbial translocation [46–49]. We and others have considered that supplementation of nutrients and metabolites to stimulate or mimic commensal metabolic output may more naturally improve microbiome function, by allowing the host and microbiome to reciprocally recalibrate themselves [50–52]. In murine models, metabolite supplementation can significantly improve intestinal immune function in models of intestinal disease [53,54]. To improve immune function in immunological non-responders (INRs), Lu et al.[55] provided INR study participants with whole-protein enteral nutrition formula supplementation. With just 3 months of daily treatment, supplemented participants exhibited improved circulating T-cell counts, and reduced microbial translocation and intestinal E. coli. In a more targeted approach, we recently provided oral sodium butyrate to SIV-infected, ARV-treated macaques and assessed measures of immune reconstitution [56]. No differences were observed in measures of disease progression, the intestinal microbiome, or butyrate-responsive transcriptional pathways as compared to controls, however, suggesting that sodium-butyrate administration is insufficient to improve intestinal immune parameters in chronically SIV-infected macaques. Additional nutritional studies are needed to shed light on the metabolic regimens that best improve immune reconstitution.

NONBACTERIAL COMPONENTS OF THE MICROBIOME

Metagenomic sequencing, in theory, allows for the identification of all members of the microbiome and recent data are beginning to explore how eukaryotic and viral constituents contribute to health and disease. Indeed, nonbacterial components of the microbiome can induce significant immunological responses. For example, the recently identified Tritrichomoas muscalis has been identified as a eukaryotic member of the murine microbiome in some animal facilities and alone induces significant innate and adaptive immune responses in vivo[57]. Moreover, human infection with cytomegalovirus is sufficient to drive expansion of large frequencies of CD4+ and CD8+ T-cells in vivo[58] and the presence of viral STDs such as HPV increase susceptibility to sexually-acquired HIV acquisition [59].

Early analysis of the nonbacterial components of the GI microbiome in PWH and SIV-infected NHPs demonstrated an expansion of the virome and suggested that the expanded virome could contribute to GI tract pathology [60,61]. Subsequent analysis found that expansion of the virome was also present in plasma of PWH [62]. In PWH, the degree of virome expansion (particularly Anelloviridae, Flaviviridae, and Adenoviridae) and bacterial dysbiosis correlate with measures of immunosuppression in participants [63–66], suggesting that lack of immune responses against particular viruses may be responsible. The composition of the virome, particularly bacteriophages, is also likely influenced by administration of ARVs in PWH [67▪]. Whether and how the expanded virome contributes to disease and inflammation in PWH is an active area of research, and interventional studies aimed at reducing particular viruses may lead to reduced inflammation.

The eukaryotic component of the microbiome too is gaining attention, particularly characterization of opportunistic yeast and fungi. The complexity of the eukaryotic microbiome has been recently explored in several anatomic sites of PWH, with alterations apparent both in the GI tract and oral cavity [68–70]. Indeed, several taxa associated with fungal opportunistic infections are routinely expanded in PWH – including Candida albicans and Pneumocystis jirovecii – and correlate with the degree to which the PWH are immunocompromised. The mechanisms underlying these expansions are currently unclear [68–70]. Little exploration of the mycobiome in longitudinally obtained samples from SIV-infected Asian macaques has been performed and further work is merited.

CONCLUSION

The anatomy and functionality of the GI tract is important for nearly all facets of health and is a focus of considerable research effort in PWH and animal models of HIV. There are considerable immunological perturbations within the GI tract of both PWH and SIV-infected Asian NHPs which are associated with focal areas of damage to the epithelial barrier and accumulating studies have explored how the composition of the luminal microbiome may also contribute [71,72]. The degree to which the microbiome is ‘druggable’ is an active area of research and how individual microbiome-altering therapeutics may benefit PWH and other individuals with diseases where the microbiome may contribute will continue to benefit from placebo-controlled interventional trials in large cohorts of genetically diverse study participants.

Acknowledgements

We would like to thank the NIH Medical Arts Branch for their assistance in generating the figure in this manuscript.

Financial support and sponsorship

Funding for this study was provided in part by the Division of Intramural Research/NIAID/NIH. The content of this publication does not necessarily reflect the views or policies of DHHS, nor does the mention of trade names, commercial products, or organizations imply endorsement by the U.S. Government.

Conflicts of interest

There are no conflicts of interest.

REFERENCES AND RECOMMENDED READING

Papers of particular interest, published within the annual period of review, have been highlighted as:

▪ of special interest

▪▪ of outstanding interest

REFERENCES 1. Grasso G, Bianciotto V, Marmeisse R. Paleomicrobiology: tracking the past microbial life from single species to entire microbial communities. Microb Biotechnol 2024; 17:e14390. 2▪. Culp EJ, Goodman AL. Cross-feeding in the gut microbiome: ecology and mechanisms. Cell Host Microbe 2023; 31:485–499. 3. Gilliland A, Chan JJ, De Wolfe TJ, et al. Pathobionts in inflammatory bowel disease: origins, underlying mechanisms, and implications for clinical care. Gastroenterology 2024; 166:44–58. 4. McKenna P, Hoffmann C, Minkah N, et al. The macaque gut microbiome in health, lentiviral infection, and chronic enterocolitis. PLoS Pathog 2008; 4:e20. 5. Mills RH, Dulai PS, Vazquez-Baeza Y, et al. Multiomics analyses of the ulcerative colitis gut microbiome link Bacteroides vulgatus proteases with disease severity. Nat Microbiol 2022; 7:262–276. 6. Jadhav A, Bajaj A, Xiao Y, et al. Role of diet-microbiome interaction in gastrointestinal disorders and strategies to modulate them with microbiome-targeted therapies. Annu Rev Nutr 2023; 43:355–383. 7. Liu S, He Y, Zhang Y, et al. Targeting gut microbiota in aging-related cardiovascular dysfunction: focus on the mechanisms. Gut Microbes 2023; 15:2290331. 8. Wang Z, Klipfell E, Bennett BJ, et al. Gut flora metabolism of phosphatidylcholine promotes cardiovascular disease. Nature 2011; 472:57–63. 9. Vujkovic-Cvijin I, Swainson LA, Chu SN, et al. Gut-resident lactobacillus abundance associates with IDO1 inhibition and Th17 dynamics in SIV-infected macaques. Cell Rep 2015; 13:1589–1597. 10. He Z, Dong H. The roles of short-chain fatty acids derived from colonic bacteria fermentation of nondigestible carbohydrates and exogenous forms in ameliorating intestinal mucosal immunity of young ruminants. Front Immunol 2023; 14:1291846. 11. Gosmann C, Anahtar MN, Handley SA, et al. Lactobacillus-deficient cervicovaginal bacterial communities are associated with increased HIV acquisition in young South African women. Immunity 2017; 46:29–37. 12. Noguera-Julian M, Rocafort M, Guillen Y, et al. Gut microbiota linked to sexual preference and HIV infection. EBioMedicine 2016; 5:135–146. 13▪▪. Fulcher JA, Li F, Tobin NH, et al. Gut dysbiosis and inflammatory blood markers precede HIV with limited changes after early seroconversion. EBioMedicine 2022; 84:104286. 14. Sui Y, Dzutsev A, Venzon D, et al. Influence of gut microbiome on mucosal immune activation and SHIV viral transmission in naive macaques. Mucosal Immunol 2018; 11:1219–1229. 15▪▪. Ortiz AM, Baker PJ, Langner CA, et al. Experimental bacterial dysbiosis with consequent immune alterations increase intrarectal SIV acquisition susceptibility. Cell Rep 2023; 42:112020. 16. Pulendran B, P SA, O’Hagan DT. Emerging concepts in the science of vaccine adjuvants. Nat Rev Drug Discov 2021; 20:454–475. 17. Hagan T, Cortese M, Rouphael N, et al. Antibiotics-driven gut microbiome perturbation alters immunity to vaccines in humans. Cell 2019; 178:1313–1328. e13. 18. Trama AM, Moody MA, Alam SM, et al. HIV-1 envelope gp41 antibodies can originate from terminal ileum B cells that share cross-reactivity with commensal bacteria. Cell Host Microbe 2014; 16:215–226. 19. Williams WB, Liao HX, Moody MA, et al. Diversion of HIV-1 vaccine-induced immunity by gp41-microbiota cross-reactive antibodies. Science 2015; 349:aab1253. 20▪. Mayer-Blackwell K, Johnson AM, Potchen N, et al. Multitrial analysis of HIV-1 envelope gp41-reactive antibodies among global recipients of candidate HIV-1 vaccines. Front Immunol 2022; 13:983313. 21. Tomaras GD, Haynes BF. HIV-1-specific antibody responses during acute and chronic HIV-1 infection. Curr Opin HIV AIDS 2009; 4:373–379. 22. Jiang D, Goswami R, Dennis M, et al. Sutterella and its metabolic pathways positively correlate with vaccine-elicited antibody responses in infant rhesus macaques. Front Immunol 2023; 14:1283343. 23. Gardner MR, Fellinger CH, Prasad NR, et al. CD4-induced antibodies promote association of the HIV-1 envelope glycoprotein with CD4-binding site antibodies. J Virol 2016; 90:7822–7832. 24. Rajashekar JK, Richard J, Beloor J, et al. Modulating HIV-1 envelope glycoprotein conformation to decrease the HIV-1 reservoir. Cell Host Microbe 2021; 29:904–916. e6. 25. Biswas S, Kuwata T, Yamauchi S, et al. Idiotopes of antibodies against HIV-1 CD4-induced epitope shared with those against microorganisms. Immunology 2023; 171:534–548. 26. Wilson A, Manuzak JA, Liang H, et al. Probiotic therapy during vaccination alters antibody response to simian-human immunodeficiency virus infection but not to commensals. AIDS Res Hum Retroviruses 2023; 39:222–231. 27. Klatt NR, Broedlow C, Osborn JM, et al. Effects of persistent modulation of intestinal microbiota on SIV/HIV vaccination in rhesus macaques. NPJ Vaccines 2021; 6:34. 28. Pinto-Cardoso S, Klatt NR, Reyes-Teran G. Impact of antiretroviral drugs on the microbiome: unknown answers to important questions. Curr Opin HIV AIDS 2018; 13:53–60. 29. Sortino O, Phanuphak N, Schuetz A, et al. Impact of acute HIV infection and early antiretroviral therapy on the human gut microbiome. Open Forum Infect Dis 2020; 7:ofz367. 30. Ishizaka A, Koga M, Mizutani T, et al. Unique gut microbiome in HIV patients on antiretroviral therapy (ART) suggests association with chronic inflammation. Microbiol Spectr 2021; 9:e0070821. 31. Klase Z, Ortiz A, Deleage C, et al. Dysbiotic bacteria translocate in progressive SIV infection. Mucosal Immunol 2015; 8:1009–1020. 32. Tanes C, Walker EM, Slisarenko N, et al. Gut microbiome changes associated with epithelial barrier damage and systemic inflammation during antiretroviral therapy of chronic SIV infection. Viruses 2021; 13:1567. 33. Pinto-Cardoso S, Lozupone C, Briceno O, et al. Fecal bacterial communities in treated HIV infected individuals on two antiretroviral regimens. Sci Rep 2017; 7:43741. 34. Hanttu AM, Pekkala S, Satokari R, et al. Gut microbiota alterations after switching from a protease inhibitor or efavirenz to raltegravir in a randomized, controlled study. AIDS 2023; 37:323–332. 35. Maier L, Pruteanu M, Kuhn M, et al. Extensive impact of nonantibiotic drugs on human gut bacteria. Nature 2018; 555:623–628. 36. Rubio-Garcia E, Ferrando N, Martin N, et al. In vitro antibacterial activity of antiretroviral drugs on key commensal bacteria from the human microbiota. Front Cell Infect Microbiol 2023; 13:1306430. 37. Shilaih M, Angst DC, Marzel A, et al. Antibacterial effects of antiretrovirals, potential implications for microbiome studies in HIV. Antivir Ther 2018; 23:91–94. 38▪. Wallace VJ, Sakowski EG, Preheim SP, Prasse C. Bacteria exposed to antiviral drugs develop antibiotic cross-resistance and unique resistance profiles. Commun Biol 2023; 6:837. 39. Ortiz AM, Flynn JK, DiNapoli SR, et al. Antiretroviral therapy administration in healthy rhesus macaques is associated with transient shifts in intestinal bacterial diversity and modest immunological perturbations. J Virol 2019; 93:e00472-19. 40. Zhang Y, Andreu-Sanchez S, Vadaq N, et al. Gut dysbiosis associates with cytokine production capacity in viral-suppressed people living with HIV. Front Cell Infect Microbiol 2023; 13:1202035. 41. Ortiz AM, Klase ZA, DiNapoli SR, et al. IL-21 and probiotic therapy improve Th17 frequencies, microbial translocation, and microbiome in ARV-treated, SIV-infected macaques. Mucosal Immunol 2016; 9:458–467. 42. Klatt NR, Canary LA, Sun X, et al. Probiotic/prebiotic supplementation of antiretrovirals improves gastrointestinal immunity in SIV-infected macaques. J Clin Invest 2013; 123:903–907. 43. d’Ettorre G, Ceccarelli G, Giustini N, et al. Probiotics reduce inflammation in antiretroviral treated, HIV-infected individuals: results of the ‘Probio-HIV’ clinical trial. PLoS One 2015; 10:e0137200. 44. Blazquez-Bondia C, Parera M, Catala-Moll F, et al. Probiotic effects on immunity and microbiome in HIV-1 discordant patients. Front Immunol 2022; 13:1066036. 45. Rousseau RK, Walmsley SL, Lee T, et al. Randomized, blinded, placebo-controlled trial of de simone formulation probiotic during HIV-associated suboptimal CD4+ T cell recovery. J Acquir Immune Defic Syndr 2022; 89:199–207. 46. Vujkovic-Cvijin I, Rutishauser RL, Pao M, et al. Limited engraftment of donor microbiome via one-time fecal microbial transplantation in treated HIV-infected individuals. Gut Microbes 2017; 8:440–450. 47. Hensley-McBain T, Zevin AS, Manuzak J, et al. Effects of fecal microbial transplantation on microbiome and immunity in simian immunodeficiency virus-infected macaques. J Virol 2016; 90:4981–4989. 48. Utay NS, Monczor AN, Somasunderam A, et al. Evaluation of six weekly oral fecal microbiota transplants in people with HIV. Pathog Immun 2020; 5:364–381. 49. Serrano-Villar S, Talavera-Rodriguez A, Gosalbes MJ, et al. Fecal microbiota transplantation in HIV: a pilot placebo-controlled study. Nat Commun 2021; 12:1139. 50. Silva M, Montes CG, Canals A, et al. Role and effects of zinc supplementation in HIV-infected patients with immunovirological discordance: a randomized, double blind, case control study. PLoS One 2021; 16:e0244823. 51. Serrano-Villar S, Vazquez-Castellanos JF, Vallejo A, et al. The effects of prebiotics on microbial dysbiosis, butyrate production and immunity in HIV-infected subjects. Mucosal Immunol 2017; 10:1279–1293. 52. Guwatudde D, Wang M, Ezeamama AE, et al. The effect of standard dose multivitamin supplementation on disease progression in HIV-infected adults initiating HAART: a randomized double blind placebo-controlled trial in Uganda. BMC Infect Dis 2015; 15:348. 53. Zou J, Chassaing B, Singh V, et al. Fiber-mediated nourishment of gut microbiota protects against diet-induced obesity by restoring IL-22-mediated colonic health. Cell Host Microbe 2018; 23:41–53. e4. 54. Yang W, Yu T, Huang X, et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat Commun 2020; 11:4457. 55. Lu D, Wang YX, Geng ST, et al. Whole-protein enteral nutrition formula supplementation reduces Escherichia and improves intestinal barrier function in HIV-infected immunological nonresponders. Appl Physiol Nutr Metab 2023; 49:319–329. 56. Ortiz AM, Simpson J, Langner CA, et al. Butyrate administration is not sufficient to improve immune reconstitution in antiretroviral-treated SIV-infected macaques. Sci Rep 2022; 12:7491. 57. Chudnovskiy A, Mortha A, Kana V, et al. Host-protozoan interactions protect from mucosal infections through activation of the inflammasome. Cell 2016; 167:444–456. e14. 58. Fuhrmann S, Reus B, Frey O, et al. Marked skewing of entire T-cell memory compartment occurs only in a minority of CMV-infected individuals and is unrelated to the degree of memory subset skewing among CMV-specific T-cells. Front Immunol 2023; 14:1258339. 59. Chavez-Torres M, Gomez-Palacio-Schjetnan M, Reyes-Teran G, et al. The vaginal microbiota of women living with HIV on suppressive antiretroviral therapy and its relation to high-risk human papillomavirus infection. BMC Microbiol 2023; 23:21. 60. Handley SA, Thackray LB, Zhao G, et al. Pathogenic simian immunodeficiency virus infection is associated with expansion of the enteric virome. Cell 2012; 151:253–266. 61. Handley SA, Desai C, Zhao G, et al. SIV infection-mediated changes in gastrointestinal bacterial microbiome and virome are associated with immunodeficiency and prevented by vaccination. Cell Host Microbe 2016; 19:323–335. 62. Nganou-Makamdop K, Talla A, Sharma AA, et al. Translocated microbiome composition determines immunological outcome in treated HIV infection. Cell 2021; 184:3899–3914. e16. 63. Vujkovic-Cvijin I, Sortino O, Verheij E, et al. HIV-associated gut dysbiosis is independent of sexual practice and correlates with noncommunicable diseases. Nat Commun 2020; 11:2448. 64. Esser PL, Quintanares GHR, Langhans B, et al. Torque teno virus load is associated with CDC stage and CD4+ cell count in people living with HIV but unrelated to AIDS-defining events and Human Pegivirus load. J Infect Dis 2024; jiae014. 65. Li Y, Cao L, Ye M, et al. Plasma virome reveals blooms and transmission of anellovirus in intravenous drug users with HIV-1, HCV, and/or HBV infections. Microbiol Spectr 2022; 10:e0144722. 66. Liu K, Li Y, Xu R, et al. HIV-1 infection alters the viral composition of plasma in men who have sex with men. mSphere 2021; 6:e00081-21. 67▪. Villoslada-Blanco P, Perez-Matute P, Iniguez M, et al. Impact of HIV infection and integrase strand transfer inhibitors-based treatment on the gut virome. Sci Rep 2022; 12:21658. 68. Meng P, Zhang G, Ma X, et al. Characterization of intestinal fungal community diversity in people living with HIV/AIDS (PLWHA). AIDS Res Ther 2024; 21:10. 69. O’Connell LM, Mann AE, Osagie E, et al. Supragingival mycobiome of HIV-exposed-but-uninfected children reflects a stronger correlation with caries-free-associated taxa compared to HIV-infected or uninfected children. Microbiol Spectr 2023; 11:e0149123. 70. Gosalbes MJ, Jimenez-Hernandez N, Moreno E, et al. Interactions among the mycobiome, bacteriome, inflammation, and diet in people living with HIV. Gut Microbes 2022; 14:2089002. 71. Mudd JC, Brenchley JM. Gut mucosal barrier dysfunction, microbial dysbiosis, and their role in HIV-1 disease progression. J Infect Dis 2016; 214: (Suppl 2): S58–S66. 72. Ortiz AM, Brenchley JM. Microbial translocation: translating simian immunodeficiency virus to HIV. Curr Opin HIV AIDS 2018; 13:15–21.

Comments (0)

No login
gif