Abstract

Distinct pathological events occur within the gastrointestinal (GI) tract of Asian macaques with progressive simian immunodeficiency virus (SIV) infection and humans with human immunodeficiency virus type 1 (HIV-1) infection that are critical in shaping disease course. These events include depletion and functional alteration of GI-resident CD4+ T cells, loss of antigen-presenting cells, loss of innate lymphocytes, and possible alterations to the composition of the gut microbiota. These contribute to structural damage to the GI tract and systemic translocation of GI tract microbial products. These translocated microbial products directly stimulate the immune system, and there is now overwhelming evidence that this drives chronic immune activation in HIV-1 and SIV infection. While combined antiretroviral therapy (cART) in HIV-1–infected subjects generally allows for immune reconstitution in peripheral blood, reconstitution of the GI tract occurs at a much slower pace, and both immunological and structural abnormalities persist in the GI tract. Importantly, studies of large cohorts of individuals have linked suboptimal GI reconstitution to residual inflammation and heightened morbidities in HIV-1–infected cART recipients. As a result, current era treatments aimed at augmenting restoration of the GI tract hold promise in returning cART recipients to full health.

Human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus (SIV) preferentially target and kill CD4+ leukocytes. Thus, it is not surprising that loss of CD4+ T cells and eventual development of AIDS are hallmarks of the infection [1–3]. However, viral replication within CD4+ T cells does not encompass the entire breadth of disease pathogenesis, given that immunological abnormalities in HIV-1 and SIV infection extend well beyond the CD4+ T-cell compartment and the pathology of HIV-1 infection is not completely reversed upon combined antiretroviral therapy (cART) administration [4]. Indeed, the pathogenesis of HIV-1 infection is multifaceted, and it is now widely accepted that chronic immune activation drives disease pathogenesis [1, 4]. Understanding the determinants of HIV-1-associated inflammation is an important step in mitigating some of the barriers that prevent HIV-1–infected subjects from returning fully to health.

Damage to the gastrointestinal (GI) tract occurs early and irreversibly in progressive HIV-1 and SIV infections and is closely linked to systemic inflammation [5–8]. In health, the GI tract serves as an important structural and immunological barrier against the trillions of microbial cells within the GI tract and also provides a microenvironment for their survival and their largely beneficial interactions with the host. Here, we discuss the numerous alterations observed within the GI tract in both human and simian immunodeficiency lentiviral infections and discuss novel therapeutic strategies to reduce intestinal inflammation and restore GI tract anatomy and physiology.

HEALTHY INTERACTIONS AMONG GUT COMMENSAL BACTERIA, INTESTINAL EPITHELIAL CELLS, AND IMMUNE CELLS

The GI tract is colonized by trillions of microorganisms, including bacteria, viruses, and fungi. Their interactions with the host, locally, are important in maintaining gut homeostasis [9–11]. In humans, large-scale metagenomic studies based predominantly on bacterial composition of the colonic (or fecal) biota suggest that the intestinal microbiota is enormously diverse and shaped by a number of host-genetic and environmental factors. While interindividual variation in the gut microbiome is apparent at the genus and species level, composition at the phylum level is relatively consistent among individuals. In healthy adults, gram-negative Bacteroidetes and gram-positive Firmicutes constitute a large majority of the colonic biota, with Proteobacteria, Tenericutes, and Fusobacteria represented with lower abundance [12].

Commensal bacteria of the gut are separated from the host by a single layer of epithelial cells, forming a barrier that is both physical and chemical in nature. This is accomplished in part by E-cadherin and claudins, which reinforce barrier integrity by forming tight junctions between epithelial cells [13]. Additionally, the mucosal layer of the gut separates the host from luminal bacteria through the action of antimicrobial peptides such as defensins, while also providing a nutrient-rich source to promote commensal bacteria colonization [13].

Gut barrier integrity is further maintained by specialized lymphocyte subsets that reside within GI tract tissues. Epithelial cells within the GI tract express receptors for the cytokines interleukin 17 (IL-17) and interleukin 22 (IL-22). CD4+, CD8+, and innate lymphoid subsets residing within the lamina propria of the gut are important sources of these cytokines [14–16], and they promote gut integrity through induction of epithelial cell proliferation and inducing expression of claudins, defensins, and mucin [16, 17]. Taken together, the net effect of these interactions among epithelial cells, immune cells, and the gut commensal microbiota promote a healthy GI tract.

IMMUNOLOGICAL DAMAGE TO THE GI TRACT IN HIV-1 INFECTION

Damage to the gut occurs very early after viral acquisition and is a sentinel manifestation of HIV-1 pathology. There is now overwhelming evidence that CD4+ T cells in the GI tract are lost disproportionately when compared to CD4+ T cells in peripheral blood and lymphoid tissues of humans with acute HIV-1 infection and Asian macaques with acute SIV infection [5, 18, 19]. The mechanisms underlying this loss are likely multifactorial, but it is thought that direct viral replication plays a significant role [20]. Unlike CD4+ T cells in peripheral blood, the majority of gut-resident CD4+ T cells possess an effector memory phenotype and express high levels of the HIV-1 coreceptor CCR5 [5]. Indeed, several studies in SIV-infected rhesus macaques have shown that CCR5-expressing CD4+ T cells are almost completely lost in the GI tract, and in situ hybridization has shown significant viral replication in the lamina propria of rhesus macaques with acute SIV infection [5, 19]. While a large majority of gut-resident CD4+ T cells are lost by direct viral cytopathic effects, some CD4+ T cells in the gut go on to become latently infected. Careful kinetic studies in hosts with progressive SIV infection have found abundant levels of SIV DNA in GI tissues as early as 3 days after infection [21, 22]. Importantly, the initial detection of viral DNA in gut tissues occurs within the eclipse phase of the SIV disease course and precedes systemic viremia.

While profound loss of CD4+ T cells in gut tissues is a salient event of the HIV-1 disease course, it is important to point out that these events alone cannot fully explain progression to AIDS or the broad dysfunction that exists in the GI tract. During nonprogressive SIV infection of natural hosts, moderate CD4+ T-cell depletion also occurs yet does not result in disease progression [23]. Furthermore, uninfected humans with idiopathic CD4+ lymphopenia exhibit very low numbers of CD4+ T cells in the GI tract without any evidence of systemic inflammation [24], indicating that there are other pathological phenomena that must contribute to HIV-1 pathogenesis.

Dysfunction that occurs within the GI tract during HIV-1 infection and progressive SIV infection is also associated with a number of functional alterations in resident leukocytes. Cells expressing IL-17 and/or IL-22 are significantly depleted from the GI tract and mesenteric lymph nodes as early as 10–14 days after infection and remain so throughout disease course [25, 26]. Importantly, only administration of cART during very early stages of acute HIV-1 infection is sufficient to restore frequencies of IL-17–producing cells in the GI tract [25, 27]. Given the importance of IL-17/IL-22–producing cells on GI barrier maintenance, a number of groups have hypothesized that a preferential depletion of these cells underlies gut barrier dysfunction in HIV-1 infection and progressive SIV infection [28–30]. While CD4+ T cells in the GI tract, termed T-helper type 17 (Th17) cells, are a predominant source of IL-17 in the gut, and while these cells are preferentially lost from the overall CD4+ T-cell pool, other groups of leukocytes are capable of producing IL-17 and IL-22 in the gut, and functional defects exist in these subsets as well during HIV-1 infection and progressive SIV infection. Considerable interest has recently been given to innate lymphoid cells (ILCs), particularly the ILC type 3 (ILC3) subset that provides an innate source of IL-17. Initial reports in SIV-infected rhesus macaques have found that ILC3s are depleted proportionally in gut tissues very early in the disease course and remain so throughout the chronic phase [31, 32]. Reports in HIV-1–infected humans have supported these findings, as early and sustained ILC depletion in blood has also been observed [33]. In addition, IL-17–producing CD8+ mucosal-associated invariant T (MAIT) cells, which uniformly express the semi-invariant Vα7.2 T-cell receptor, follow similar dynamics of early and nonreversible loss in SIV/HIV-1-infected subjects [34, 35], implicating a dysfunction of IL-17–producing leukocytes well beyond the Th17 subset in HIV-1 infection.

Given that the loss of IL-17–producing gut leukocytes is linked to GI damage, the mechanisms underlying the loss of these cells are thus of considerable interest. The surface chemokine receptor CCR6 marks Th17 polarization, and some studies have noted a preferential infection of the CCR6-expressing CD4+ T cells in vitro [28, 30]. Yet it is likely that characterizing Th17 cells on the basis of this phenotype overestimates Th17 cell frequency in comparison to functional Th17 assessment. Indeed, we have previously mitogenically stimulated CD4+ T cells from SIV-infected rhesus macaques and flow cytometrically sorted Th1 and Th17 cells to determine levels of SIV DNA and found equal frequencies of SIV infection among Th1 and Th17 cells [36], indicating that preferential infection of Th17 cells may not occur systemically in vivo.

Intriguingly, ILC3s and MAIT cells do not express HIV-1 receptors and are not permissive to SIV or HIV-1 infection, yet they are depleted concurrently with Th17 cells in HIV-1 or SIV infection [32, 34]. Precise mechanisms behind the loss of these cells are just beginning to be explored, although inflammatory mediators may play a role, as a recent report in humanized-mouse models of HIV-1 infection have implicated interferon α as a potential determinant of ILC3 loss in this setting [37]. Additionally, several groups have investigated cell types that are known to maintain IL-17–producing cells, such as gut-resident dendritic cells (DCs), and have found that they too are altered both functionally and proportionally in SIV or HIV-1 infection. The enzyme indoleamine 2,3-dioxegenase 1 (IDO1) catabolizes the amino acid tryptophan and suppresses T-cell function in a number of disease states [38, 39]. IDO also suppresses Th17 cell function in vitro, and in HIV-1 infection upregulation of IDO activity in DCs is associated with lower frequencies of Th17 cells [39]. Moreover, we have shown in SIV-infected rhesus macaques that increased IDO activity is associated with loss of CD103+ DCs in the gut, which are a source of factors important for Th17 polarization [16]. Taken together, alterations in the landscape of gut-resident APCs may also be important in IL-17–producing cell dysfunction during HIV-1 or SIV infection.

STRUCTURAL DAMAGE TO THE GI TRACT IN HIV-1 INFECTION

Both inflammation and immunological abnormalities that occur in the gut during the HIV-1 or SIV disease course are tightly linked to structural damage to the GI tract. Even in the early days of the HIV-1 epidemic, several structural abnormalities were observed at the tissue level in the GI tract of subjects with chronic HIV-1 infection, such as focal epithelial cell degeneration, malabsorption, and crypt hyperplasia [40]. As histological techniques have advanced, several groups have corroborated these findings at the cellular level, observing massive enterocyte apoptosis, decreased expression of tight junction proteins, and increased intestinal permeability in both chronic HIV-1 and SIV infection [7, 26, 41]. The net effect of these abnormalities results in focal breaches to the gut epithelial barrier, and this is associated directly with the degree of microbial translocation both locally and systemically (discussed below) [42, 43].

HIV-1 and SIV-associated structural damage that occurs to the gut is likely due to a variety of mechanisms. As barrier integrity of the GI epithelium is progressively compromised, sterile anatomical niches of the gut become exposed to lumenal bacterial products. Translocation of bacterial products regulate inflammatory processes locally within the gut, and it is believed that inflammation is, at least in part, a key initiator of GI barrier dysfunction in HIV-1 and SIV infection. Indeed, levels of transcripts encoding inflammatory cytokines such as tumor necrosis factor (TNF), interleukin 6 (IL-6), interleukin 10, and interferon γ (IFN-γ) are significantly elevated in the colonic mucosa of HIV-1–infected patients and remain so after suppressive cART [44]. Additionally, given that the amount of damage to the GI tract in HIV-1 and SIV infection is inversely related to frequencies of IL-17–producing lymphocytes in the gut [16], it is also likely that, after initial inflammatory processes occur, damage is further perpetuated by alterations in lymphocyte subsets involved in maintenance and repair of the GI tract.

CONSEQUENCES OF GI BARRIER DYSFUNCTION IN HIV-1 AND SIV INFECTIONS

Immunological and structural damage to the GI tract promote translocation of commensal bacteria locally and systemically. In both HIV-1 and progressive SIV infection, focal breaches of the epithelial barrier can be found in spatial juxtaposition to infiltrating microbial products in vivo [7, 42]. There is overwhelming evidence of persistent endotoxemia during chronic HIV-1 and SIV infection. Since the first description of this phenomenon in 2006 [6], >50 groups have corroborated the existence of microbial products in plasma from HIV-1-infected humans and Asian macaques with progressively SIV infection [2, 22]. Importantly, there is clear indication that these microbial products are bioactive in vivo and exacerbate immune activation. For example, monocyte subsets that directly respond to microbial products through Toll-like receptors (TLRs) are systemically activated in HIV-1-infected humans and SIV-infected Asian macaques [45, 46], and while microbial products may stimulate the adaptive immune system more indirectly, higher levels of lipopolysaccharide (LPS) have been linked with elevated indices of immune activation on CD8+ T cells in HIV-1-infected subjects [2, 6].

While it is clear that translocated microbial products contribute to HIV-1-associated immune activation, the degree to which they contribute is less certain. Because HIV-1 and SIV replication alone each induce inflammation and chronic immune stimulation, researchers have turned to settings that uncouple these factors, to assess how GI damage and resulting microbial translocation independently influence HIV-1 and SIV disease pathology. This has been highlighted recently in work by Hao et al, in which gut damage induced by dextran-sodium sulfate administration to healthy, SIV-uninfected, rhesus macaques recapitulated many pathologic features of HIV-1 and SIV infection [47]. Given that microbial translocation in patients with inflammatory bowel diseases is also associated with systemic inflammation and immune activation [48, 49], these data provide evidence that microbial translocation independently contributes to HIV-1 and SIV disease pathology.

The degree to which microbial translocation, as opposed to HIV-1 and SIV replication, influences immune activation can be further understood in HIV-1–infected cART recipients. In the current era of treatment, cART is highly successful at suppressing viremia to undetectable levels over prolonged periods [50]. Nevertheless, treated HIV-1–infected individuals exhibit higher incidences of non–AIDS-related morbidities and mortalities when compared to the general population. These morbidities include cardiovascular disease, cancer, osteoporosis, and neurocognitive dysfunction [4]. Importantly, several studies point to residual inflammation as an independent predictor of these morbidities [4]. Indeed, levels inflammatory proteins such as IL-6, C-reactive protein, and the coagulation marker D-dimer remain elevated in plasma of virally suppressed HIV-1-infected subjects, and they are particularly elevated in a subset of patients who display suboptimal CD4+ T-cell reconstitution in response to cART [51, 52].

As cART recipients are, for the most part, virally suppressed, it is important to consider mechanisms other than viral replication that may contribute to residual inflammation in treated HIV-1-infected subjects. Much effort has focused on assessing immune reconstitution in mucosal and lymphoid tissues after ART administration. Shortly after the first description of microbial translocation in HIV-1 and SIV infection, in 2006, it was found that significant CD4+ T-cell depletion remained in the GI tract of cART-treated patients, some of whom had had virological suppression for up to 7 years [8]. Several follow-up studies have corroborated these results, and it is now known that despite immune reconstitution in peripheral blood, reconstitution of CD4+ T cells in the GI tract occurs at a much slower pace [53, 54]. Defects in structural components of the GI tract persist as well in HIV-1–infected subjects who received long-term cART, as one study reported that expression of tight junction proteins remained significantly lower in colonic tissue of HIV-1–infected patients receiving cART [55]. Interestingly, these defects were not observed in the terminal ileum, indicating that cART may be more effective at reconstituting some anatomical sites of the GI tract than at others, with immunological improvements more pronounced in the duodenum as compared to the rectum [56]. While gut reconstitution may be compartmentalized, it remains clear that, overall, the GI tract of cART recipients does not completely normalize despite a general improvement in the health of these subjects. Moreover, GI damage, residual inflammation, microbial translocation, and non–AIDS-related morbidities all appear to be linked to one another. This link is most significantly illustrated in the findings of multiple groups, showing that levels of soluble CD14, a molecule cleaved from the cell surface of monocytes in response to activation by LPS, are elevated in plasma of HIV-1–infected cART recipients and predict earlier mortality [57–60]. These associations are also seen with surrogate plasma markers of gut epithelial integrity, such as zonulin-1 and intestinal fatty-acid binding protein, which independently predict mortality in cART recipients with a diagnosis of AIDS [58].

Exactly why reconstitution of the GI tract is suboptimal following cART is uncertain, although an altered immunological microenvironment in the GI tract that persists long after cART administration is likely a contributing factor. Indeed, levels of inflammatory cytokines such as TNF, IL-6, and IFN-γ remain elevated in gut tissues of cART recipients [44]. Local inflammation within the gut promotes collagen deposition and damage to the architecture of gut-associated lymphoid tissues (GALT) that support a large majority of GI tract–resident CD4+ T cells [61]. Importantly, the degree of collagen deposition has shown to be inversely related to the extent of CD4+ T-cell depletion in the GALT and of GALT CD4+ T-cell reconstitution after cART [62]. While cART can successfully reconstitute CD4+ T cells in the peripheral blood of many HIV-1–infected subjects, functional abnormalities in gut-resident CD4+ T cells oftentimes persist as well, particularly in the Th17 cell subset [27, 55, 63]. Although the mechanisms are unclear, it appears that immunological abnormalities in the gut influence the size of the viral reservoir. CD4+ T cells harboring viral DNA are enriched in gut tissues of HIV-1–infected cART recipients, compared with CD4+ T cells in peripheral blood [64], and in SIV-infected rhesus macaques receiving ART regimens, functional defects in Th17 cells correlate inversely with levels of viral DNA in gut tissues of these animals [27], suggesting a link between residual GI dysfunction and persistence of the viral reservoir.

DYSBIOSIS IN LENTIVIRAL IMMUNODEFICIENCY INFECTIONS

Diseases associated with GI inflammation are not limited to HIV-1 and SIV infection, as inflammatory bowel diseases have several similarities [65]. In these settings, significant shifts in the gut microflora composition (or dysbiosis) occur, and these are linked to inflammation [66]. Because chronic gut inflammation is also characteristic of HIV-1 and SIV infection, researchers have begun to examine whether dysbiosis occurs in HIV-1 and SIV infection and how this influences disease pathology. Indeed, significant dysbiosis is clearly evident in both the rectal and fecal biota of untreated HIV-1–infected subjects as compared to uninfected individuals [67–69]. While it is important to note that the composition of the microbiome is not uniform across the GI tract, significant dysbiosis is clearly evident in both the rectal and fecal biota of untreated HIV-1–infected subjects as compared to uninfected individuals [67–69]. Moreover, cART might alter the microbiota composition, but the resulting composition bears more resemblance to the microbiome of untreated HIV-1–infected subjects than that of healthy uninfected individuals [69, 70]. The microbiota of HIV-1–infected subjects are profoundly depleted of Bacteroides (which are associated with limiting inflammation) and enriched for Proteobacteria (which promote inflammation) [67–69]. While it is clear that the studies noted above have uncovered potential common features of HIV-1-associated dysbiosis, some inconsistencies exist, highlighting a growing need to identify non–HIV-1-related variables that may affect the microbiome. For example, contradictory findings have been reported in associations between HIV-1 serostatus and shifts in the fecal composition of the Prevotella genus. While some studies observed a significant enrichment of Prevotella in chronic HIV-1 infection [70, 71], these findings are contradicted by studies controlling for HIV-1 risk groups [67, 72]. Intriguingly, a recent report stratified sexual preference and studied the fecal microbial composition among HIV-1–infected and uninfected control subjects, finding that same-sex-attracted men had microbiomes enriched for Prevotella [73]. Moreover, no specific signature of dysbiosis at the genera level was apparent in the HIV-1–infected group when controlling for sexual preference, with the only HIV-1-infected difference being decreased microbiome richness [74, 75]. In line with these findings, lower bacterial richness has reported to be more pronounced in immunological nonresponders displaying suboptimal CD4+ T-cell reconstitution during cART [76].

Nonhuman primate SIV models of HIV-1 infection have allowed researchers to longitudinally assess the GI tract microbiome relative to SIV infection while also controlling for factors such as diet. In 3 initial reports, all animals infected with SIV progressed to AIDS, yet disease progression was not associated with any significant changes to composition of the bacterial flora [77–79]. While bacterial community composition may remain relatively stable during SIV infection, disease course may be associated with emergence of specific groups of bacteria, as bacterial families that frequently harbor enteropathogens are expanded in SIV-infected animals with low blood CD4+ T-cell percentages [78]. Notably, these findings appear to be concordant with those from studies in HIV-1–infected humans, as well [80]. It is unclear why other aspects of microbial dysbiosis in HIV-1–infected infected humans are not recapitulated in nonhuman primate models of HIV-1 infection. However, it is clear that, in both species, HIV-1 and progressive SIV infection are associated with a significant increase in both abundance and diversity of the enteric virome [78]. In SIV-infected Asian macaques, those viruses that were expanded included potentially pathogenic adenoviruses, which were found to be in close proximity to lesions in the intestinal epithelium [78]. Separate studies in a cohort of HIV-1–infected Ugandan patients similarly found an expansion of the fecal virome [80], and this was particularly apparent in subjects with low peripheral CD4+ T-cell counts and was independent of cART [80].

Unraveling the precise host-microbiota interactions that drive pathogenic features of HIV-1 and SIV infection has proved difficult to date, given that many microbial species of the gut are anaerobic and difficult to culture in vitro. Nevertheless, some researchers have used metagenomic approaches to uncover metabolic gene pathways enriched in gut microbial communities of HIV-1–infected subjects, with one study noting an increase in pathways involved in synthesis of molecules such as LPS [71]. We also have examined metabolic properties of the GI tract microbiome, finding that Proteobacteria, while constituting only a small component of the microbiome, are the most metabolically active and preferentially translocate in SIV-infected Asian macaques [81]. While these studies begin to shed light on the functional properties of potentially pathogenic bacteria, a better indication of the role of dysbiosis in HIV-1 and SIV pathogenesis has come from correlative studies that link dysbiosis to important features of HIV-1 and SIV disease progression. For example, 2 studies have found that the overall microbial composition in rectosigmoid biopsy specimens correlates directly with CD4+ and CD8+ T-cell activation in the blood and gut mucosa of HIV-1–infected subjects [67–69]. A greater and more reliable predictor of gut microbial health may be an overall measurement of species diversity, and 2 groups have found that decreased diversity is associated with low CD4+ T-cell counts in HIV-1–infected subjects [76, 80]. Interestingly, 2 groups have reported that overall bacterial diversity in the stool of cART recipients is decreased when compared to both untreated HIV-1–infected and healthy control subjects [69, 70]. Given that up to 40% of ART recipients experience mild-to-severe diarrhea [82], these results raise the possibility that antiretroviral drugs alone may affect the gut microbiota. In line with this hypothesis, we have also seen that administration of ART is marked by transient dysbiosis in the stool of SIV-infected rhesus macaques [81]. Taken together, these studies outline the increasing importance of the microbiome in shaping HIV-1 pathogenesis and leave open the possibility of therapeutic strategies aimed at targeting the microbiome to improve the health of HIV-1–infected subjects.

TREATMENTS

GI dysfunction and resultant inflammation clearly persist in HIV-1–infected subjects despite long-term suppressive cART. Importantly, when cART is given in the first 10–20 days after HIV-1 transmission, mucosal and systemic inflammation tend to be fully reversed (or prevented) [25, 83]. Because HIV-1 diagnosis and treatment at such early stages in the disease course is challenging, supplementation of cART with therapies aimed at augmenting gut mucosal reconstitution and reducing microbial translocation have been considered. Two studies have focused on interfering with translocating microbial products directly through administration of the LPS-sequestering, phosphate-binding drug sevelamer, and these have had conflicting results. While sevelamer drastically reduced indices of immune activation and inflammation systemically in untreated SIVagm-infected pigtail macaques [84], the drug had no effect on any of these parameters in cART-naive HIV-1–infected humans [85]. Other approaches to cART supplementation, one involving rifaximin, a nonabsorbable antibiotic that decreases microbial translocation in cirrhotics, and another involving mesalamine, an antiinflammatory drug used in treatment of inflammatory bowel disease, have yielded negligible improvements in inflammatory or immune parameters in treated HIV-1–infected subjects [86, 87]. It is notable that rifaximin treatment with the addition of sulfasalazine (an antiinflammatory agent) in pigtail macaques with acute SIV infection transiently reduced inflammation and coagulation markers [88], suggesting that combinatorial approaches blocking inflammation/microbial translocation could warrant more-promising results when compared to the monotherapy studies in HIV-1–infected humans noted above.

While the studies described have centered on blockade of microbial translocation, a therapeutic strategy gaining recent attention has been dietary supplementation with probiotic bacteria. Probiotic bacteria are thought to confer benefit to the host via a variety of mechanisms, such as production of short-chain fatty acids and competition with potentially pathogenic bacteria, all of which promote an antiinflammatory state. Given the importance of the microbiome in gut homeostasis and that many immune abnormalities in the treatment era appear to be related to residual gut damage, a number of recent studies have investigated supplementation of cART with probiotics. Each of these studies found some reduction in indices of inflammation, coagulation, or immune activation associated with probiotics when compared to cART alone [89–91]. In related approaches, unique oligosaccharide mixtures thought to stimulate the growth of probiotic species (prebiotics) have been used alone or in combination with probiotic supplementation (synbiotics). In these studies, prebiotic or synbiotic therapy in HIV-1–infected subjects was associated with reductions in markers of inflammation and marginally significant increases in CD4+ T-cell counts [92, 93]. Thus, most of these studies show that probiotic supplementation was associated with some degree of immunological improvement. It is important to note, however, that the mechanisms by which probiotics exert these effects are unclear. There is also some uncertainty as to which formulations of probiotic species are the most beneficial in HIV-1–infected cART recipients or whether the same probiotic or prebiotic mixtures would work for HIV-1–infected individuals across all demographic characteristics. Two of the most widely used probiotic formulations, Lactobacillus GG and VSL-3, were recently shown to confer an immunological benefit in SIV-infected Asian macaques receiving cART, reducing fibrosis of GI tract lymphoid follicles and reconstituting gut CD4+ T cells to near healthy levels [79]. These results suggest that the largest effects of probiotic treatment may be within the GI tract itself and that some of the systemic benefits conferred by probiotics in the human studies noted above may be related to GI tract reconstitution. It is notable that all of these studies relied on relatively small sample sizes and thus will need to be confirmed within larger cohorts. Yet, given the relative safety of probiotics when compared to that of other therapies, these studies may warrant probiotics as promising supplements to cART in HIV-1–infected subjects.

Fecal microbiota transplantation (FMT), which involves the transfer of microbial flora from a recipient to a donor, has recently been considered as a potential therapy in HIV-1–infected subjects. In comparison to probiotics, the fecal microbiota is more diverse and tailored to the colonic niche, thus having the potential to mediate more-durable effects. Indeed, a recent study found that a single transfer of donor microbial strains persisted within the recipient for up to 3 months [94]. One FMT trial is currently underway in HIV-1–infected cART recipients. While clinical end points have yet to be reported, FMT appeared to be safe and engraftment of donor microbial flora was seen in all 6 subjects enrolled in the study [95]. FMT has recently been performed in SIV-infected Asian macaques receiving cART [96]. Treatment was well tolerated and reduced indices of immune activation after FMT.

Last, several approaches have sought to boost immune reconstitution in HIV-1–infected cART recipients with exogenous homeostatic cytokines. For example, interleukin 21 (IL-21) is a pleiotropic cytokine that can promote gut homeostasis through enhancement of Th17 cell function. Recombinant IL-21 has recently been given to SIV-infected Asian macaques receiving cART, where it improved Th17 restoration and promoted more-effective reduction of immune activation in blood and gut mucosal tissues when compared to cART alone [97]. In line with a model whereby incomplete gut reconstitution can promote persistence of the HIV-1 reservoir, cART-recipient animals receiving IL-21 in this study also exhibited lower levels of proviral DNA in gut tissues [97], thus potentially providing a rational for clinical studies of IL-21 therapy in HIV-1–infected cART recipients. In other studies, encouraging results have been obtained with a separate homeostatic cytokine, interleukin 7 (IL-7), which is currently in phase 2 clinical trials in HIV-1–infected cART recipients [98]. IL-7 is important in promoting homeostatic division of naive T cells and enhancing T-cell homing to the gut. When compared to cART alone, IL-7 therapy achieved more-sustained CD4+ T-cell restoration in the majority of participants [98]. Taken together, the novel therapeutic strategies outlined above may promote effective immune reconstitution by restoring structural and immunological components of the GI tract.

CONCLUDING REMARKS

Structural and immunological damage to the GI tract have become salient features of HIV-1 and SIV disease (Figure 1). There have been significant strides in understanding the mechanisms underlying damage to the GI tract, although significant questions remain. These include the following: (1) how early after infection do these begin? (2) what mechanisms underlie microbial dysbiosis, and is this an important consideration for HIV-1-infected individuals? (3) are particular bacterial taxa actually mediating pathology in HIV-1 and SIV infection? and (4) will cART recipients benefit from therapies aimed at restoring GI tract health? The therapeutic strategies outlined above not only provide promising directions for research, but also underscore proof-of-principle observations that reconstitution of the gut mucosa will likely be important in returning HIV-1–infected subjects fully to health.

Gastrointestinal (GI) damage is not fully reversed during human immunodeficiency virus type 1 (HIV-1) infection, despite immune reconstitution in peripheral blood. Viral replication, chronic inflammation, and loss of interleukin 17 (IL-17)–producing cells (Th17/Tc17) cause irreparable damage to the GI tract, inducing the translocation of commensal microbes from the lumen into the systemic circulation. The GI microenvironment does not fully normalize with combined antiretroviral therapy (cART), despite apparent immune reconstitution in peripheral blood. Abbreviation: ILC, innate lymphoid cell.
Figure 1.

Gastrointestinal (GI) damage is not fully reversed during human immunodeficiency virus type 1 (HIV-1) infection, despite immune reconstitution in peripheral blood. Viral replication, chronic inflammation, and loss of interleukin 17 (IL-17)–producing cells (Th17/Tc17) cause irreparable damage to the GI tract, inducing the translocation of commensal microbes from the lumen into the systemic circulation. The GI microenvironment does not fully normalize with combined antiretroviral therapy (cART), despite apparent immune reconstitution in peripheral blood. Abbreviation: ILC, innate lymphoid cell.

Notes

Disclaimer. The content of this publication does not necessarily reflect the views or policies of the Department of Health and Human Services, nor does the mention of trade names, commercial products, or organizations imply endorsement by the US government.

Supplement sponsorship. This article appears as part of the supplement “Persistent Inflammation in Treated HIV Disease,” sponsored by Case Western Reserve University and supported by funds from the James B. Pendleton Charitable Trust.

Financial support. This work was supported by the Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health.

Potential conflict of interest. Both authors: No reported conflicts. Both authors have submitted the ICMJE Form for Disclosure of Potential Conflicts of Interest. Conflicts that the editors consider relevant to the content of the manuscript have been disclosed.

References

1

Giorgi
JV
,
Hultin
LE
,
McKeating
JA
et al. .
Shorter survival in advanced human immunodeficiency virus type 1 infection is more closely associated with T lymphocyte activation than with plasma virus burden or virus chemokine coreceptor usage
.
J Infect Dis
1999
;
179
:
859
70
.

2

Hunt
PW
,
Brenchley
J
,
Sinclair
E
et al. .
Relationship between T cell activation and CD4+ T cell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy
.
J Infect Dis
2008
;
197
:
126
33
.

3

Pantaleo
G
,
Graziosi
C
,
Fauci
AS
.
New concepts in the immunopathogenesis of human immunodeficiency virus infection
.
N Engl J Med
1993
;
328
:
327
35
.

4

Lederman
MM
,
Funderburg
NT
,
Sekaly
RP
,
Klatt
NR
,
Hunt
PW
.
Residual immune dysregulation syndrome in treated HIV infection
.
Adv Immunol
2013
;
119
:
51
83
.

5

Brenchley
JM
,
Schacker
TW
,
Ruff
LE
et al. .
CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract
.
J Exp Med
2004
;
200
:
749
59
.

6

Brenchley
JM
,
Price
DA
,
Schacker
TW
et al. .
Microbial translocation is a cause of systemic immune activation in chronic HIV infection
.
Nat Med
2006
;
12
:
1365
71
.

7

Estes
JD
,
Harris
LD
,
Klatt
NR
et al. .
Damaged intestinal epithelial integrity linked to microbial translocation in pathogenic simian immunodeficiency virus infections
.
PLoS Pathog
2010
;
6
:
e1001052
.

8

Mehandru
S
,
Poles
MA
,
Tenner-Racz
K
et al. .
Lack of mucosal immune reconstitution during prolonged treatment of acute and early HIV-1 infection
.
PLoS Med
2006
;
3
:
e484
.

9

O'Hara
AM
,
Shanahan
F
.
The gut flora as a forgotten organ
.
EMBO Rep
2006
;
7
:
688
93
.

10

Reyes
A
,
Haynes
M
,
Hanson
N
et al. .
Viruses in the faecal microbiota of monozygotic twins and their mothers
.
Nature
2010
;
466
:
334
8
.

11

Underhill
DM
,
Iliev
ID
.
The mycobiota: interactions between commensal fungi and the host immune system
.
Nat Rev Immunol
2014
;
14
:
405
16
.

12

Gill
SR
,
Pop
M
,
Deboy
RT
et al. .
Metagenomic analysis of the human distal gut microbiome
.
Science
2006
;
312
:
1355
9
.

13

Turner
JR
.
Intestinal mucosal barrier function in health and disease
.
Nat Rev Immunol
2009
;
9
:
799
809
.

14

Aujla
SJ
,
Kolls
JK
.
IL-22: a critical mediator in mucosal host defense
.
J Mol Med (Berl)
2009
;
87
:
451
4
.

15

Cella
M
,
Fuchs
A
,
Vermi
W
et al. .
A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity
.
Nature
2009
;
457
:
722
5
.

16

Klatt
NR
,
Estes
JD
,
Sun
X
et al. .
Loss of mucosal CD103+ DCs and IL-17+ and IL-22+ lymphocytes is associated with mucosal damage in SIV infection
.
Mucosal Immunol
2012
;
5
:
646
57
.

17

Sugimoto
K
,
Ogawa
A
,
Mizoguchi
E
et al. .
IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis
.
J Clin Invest
2008
;
118
:
534
44
.

18

Rodgers
VD
,
Fassett
R
,
Kagnoff
MF
.
Abnormalities in intestinal mucosal T cells in homosexual populations including those with the lymphadenopathy syndrome and acquired immunodeficiency syndrome
.
Gastroenterology
1986
;
90
:
552
8
.

19

Li
Q
,
Duan
L
,
Estes
JD
et al. .
Peak SIV replication in resting memory CD4+ T cells depletes gut lamina propria CD4+ T cells
.
Nature
2005
;
434
:
1148
52
.

20

Mattapallil
JJ
,
Douek
DC
,
Hill
B
,
Nishimura
Y
,
Martin
M
,
Roederer
M
.
Massive infection and loss of memory CD4+ T cells in multiple tissues during acute SIV infection
.
Nature
2005
;
434
:
1093
7
.

21

Whitney
JB
,
Hill
AL
,
Sanisetty
S
et al. .
Rapid seeding of the viral reservoir prior to SIV viraemia in rhesus monkeys
.
Nature
2014
;
512
:
74
7
.

22

Barouch
DH
,
Ghneim
K
,
Bosche
WJ
et al. .
Rapid inflammasome activation following mucosal SIV infection of Rhesus ronkeys
.
Cell
2016
;
165
:
656
67
.

23

Gordon
SN
,
Klatt
NR
,
Bosinger
SE
et al. .
Severe depletion of mucosal CD4+ T cells in AIDS-free simian immunodeficiency virus-infected sooty mangabeys
.
J Immunol
2007
;
179
:
3026
34
.

24

Kovacs
SB
,
Sheikh
V
,
Thompson
WL
et al. .
T-cell depletion in the colonic mucosa of patients with idiopathic CD4+ lymphopenia
.
J Infect Dis
2015
;
212
:
1579
87
.

25

Schuetz
A
,
Deleage
C
,
Sereti
I
et al. .
Initiation of ART during early acute HIV infection preserves mucosal Th17 function and reverses HIV-related immune activation
.
PLoS Pathog
2014
;
10
:
e1004543
.

26

Brenchley
JM
.
Mucosal immunity in human and simian immunodeficiency lentivirus infections
.
Mucosal Immunol
2013
;
6
:
657
65
.

27

Ryan
ES
,
Micci
L
,
Fromentin
R
et al. .
Loss of function of intestinal IL-17 and IL-22 producing cells contributes to inflammation and viral persistence in SIV-infected rhesus macaques
.
PLoS Pathog
2016
;
12
:
e1005412
.

28

Monteiro
P
,
Gosselin
A
,
Wacleche
VS
et al. .
Memory CCR6+CD4+ T cells are preferential targets for productive HIV type 1 infection regardless of their expression of integrin beta7
.
J Immunol
2011
;
186
:
4618
30
.

29

El Hed
A
,
Khaitan
A
,
Kozhaya
L
et al. .
Susceptibility of human Th17 cells to human immunodeficiency virus and their perturbation during infection
.
J Infect Dis
2010
;
201
:
843
54
.

30

Alvarez
Y
,
Tuen
M
,
Shen
G
et al. .
Preferential HIV infection of CCR6+ Th17 cells is associated with higher levels of virus receptor expression and lack of CCR5 ligands
.
J Virol
2013
;
87
:
10843
54
.

31

Xu
H
,
Wang
X
,
Liu
DX
,
Moroney-Rasmussen
T
,
Lackner
AA
,
Veazey
RS
.
IL-17-producing innate lymphoid cells are restricted to mucosal tissues and are depleted in SIV-infected macaques
.
Mucosal Immunol
2012
;
5
:
658
69
.

32

Li
H
,
Richert-Spuhler
LE
,
Evans
TI
et al. .
Hypercytotoxicity and rapid loss of NKp44+ innate lymphoid cells during acute SIV infection
.
PLoS Pathog
2014
;
10
:
e1004551
.

33

Kloverpris
HN
,
Kazer
SW
,
Mjosberg
J
et al. .
innate lymphoid cells are depleted irreversibly during acute HIV-1 infection in the absence of viral suppression
.
Immunity
2016
;
44
:
391
405
.

34

Cosgrove
C
,
Ussher
JE
,
Rauch
A
et al. .
Early and nonreversible decrease of CD161++ /MAIT cells in HIV infection
.
Blood
2013
;
121
:
951
61
.

35

Vinton
C
,
Wu
F
,
Rossjohn
J
et al. .
Mucosa-Associated Invariant T Cells Are Systemically Depleted in Simian Immunodeficiency Virus-Infected Rhesus Macaques
.
J Virol
2016
;
90
:
4520
9
.

36

Brenchley
JM
,
Paiardini
M
,
Knox
KS
et al. .
Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections
.
Blood
2008
;
112
:
2826
35
.

37

Zhang
Z
,
Cheng
L
,
Zhao
J
et al. .
Plasmacytoid dendritic cells promote HIV-1-induced group 3 innate lymphoid cell depletion
.
J Clin Invest
2015
;
125
:
3692
703
.

38

Mellor
AL
,
Munn
DH
.
IDO expression by dendritic cells: tolerance and tryptophan catabolism
.
Nat Rev Immunol
2004
;
4
:
762
74
.

39

Favre
D
,
Mold
J
,
Hunt
PW
et al. .
Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 alters the balance of TH17 to regulatory T cells in HIV disease
.
Sci Transl Med
2010
;
2
:
32ra6
.

40

Kotler
DP
,
Gaetz
HP
,
Lange
M
,
Klein
EB
,
Holt
PR
.
Enteropathy associated with the acquired immunodeficiency syndrome
.
Ann Intern Med
1984
;
101
:
421
8
.

41

Bjarnason
I
,
Sharpstone
DR
,
Francis
N
et al. .
Intestinal inflammation, ileal structure and function in HIV
.
AIDS
1996
;
10
:
1385
91
.

42

Nazli
A
,
Chan
O
,
Dobson-Belaire
WN
et al. .
Exposure to HIV-1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation
.
PLoS Pathog
2010
;
6
:
e1000852
.

43

Mohan
M
,
Aye
PP
,
Borda
JT
,
Alvarez
X
,
Lackner
AA
.
Gastrointestinal disease in simian immunodeficiency virus-infected rhesus macaques is characterized by proinflammatory dysregulation of the interleukin-6-Janus kinase/signal transducer and activator of transcription3 pathway
.
Am J Pathol
2007
;
171
:
1952
65
.

44

Schulbin
H
,
Bode
H
,
Stocker
H
et al. .
Cytokine expression in the colonic mucosa of human immunodeficiency virus-infected individuals before and during 9 months of antiretroviral therapy
.
Antimicrob Agents Chemother
2008
;
52
:
3377
84
.

45

Kim
WK
,
Sun
Y
,
Do
H
et al. .
Monocyte heterogeneity underlying phenotypic changes in monocytes according to SIV disease stage
.
J Leukoc Biol
2010
;
87
:
557
67
.

46

Ancuta
P
,
Kamat
A
,
Kunstman
KJ
et al. .
Microbial translocation is associated with increased monocyte activation and dementia in AIDS patients
.
PLoS One
2008
;
3
:
e2516
.

47

Hao
XP
,
Lucero
CM
,
Turkbey
B
et al. .
Experimental colitis in SIV-uninfected rhesus macaques recapitulates important features of pathogenic SIV infection
.
Nat Commun
2015
;
6
:
8020
.

48

Funderburg
NT
,
Stubblefield Park
SR
,
Sung
HC
et al. .
Circulating CD4(+) and CD8(+) T cells are activated in inflammatory bowel disease and are associated with plasma markers of inflammation
.
Immunology
2013
;
140
:
87
97
.

49

Pasternak
BA
,
D'Mello
S
,
Jurickova
II
et al. .
Lipopolysaccharide exposure is linked to activation of the acute phase response and growth failure in pediatric Crohn's disease and murine colitis
.
Inflamm Bowel Dis
2010
;
16
:
856
69
.

50

Siliciano
JD
,
Siliciano
RF
.
Biomarkers of HIV replication
.
Curr Opin HIV AIDS
2010
;
5
:
491
7
.

51

Duprez
DA
,
Kuller
LH
,
Tracy
R
et al. .
Lipoprotein particle subclasses, cardiovascular disease and HIV infection
.
Atherosclerosis
2009
;
207
:
524
9
.

52

Lederman
MM
,
Calabrese
L
,
Funderburg
NT
et al. .
Immunologic failure despite suppressive antiretroviral therapy is related to activation and turnover of memory CD4 cells
.
J Infect Dis
2011
;
204
:
1217
26
.

53

Miao
YM
,
Hayes
PJ
,
Gotch
FM
,
Barrett
MC
,
Francis
ND
,
Gazzard
BG
.
Elevated mucosal addressin cell adhesion molecule-1 expression in acquired immunodeficiency syndrome is maintained during antiretroviral therapy by intestinal pathogens and coincides with increased duodenal CD4T cell densities
.
J Infect Dis
2002
;
185
:
1043
50
.

54

Guadalupe
M
,
Sankaran
S
,
George
MD
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
47
.

55

Chung
CY
,
Alden
SL
,
Funderburg
NT
,
Fu
P
,
Levine
AD
.
Progressive proximal-to-distal reduction in expression of the tight junction complex in colonic epithelium of virally- suppressed HIV+ individuals
.
PLoS Pathog
2014
;
10
:
e1004198
.

56

Serrano-Villar
S
,
Sainz
T
,
Ma
ZM
et al. .
Effects of Combined CCR5/Integrase Inhibitors- Based Regimen on Mucosal Immunity in HIV-Infected Patients Naive to Antiretroviral Therapy: A Pilot Randomized Trial
.
PLoS Pathog
2016
;
12
:
e1005381
.

57

Sandler
NG
,
Wand
H
,
Roque
A
et al. .
Plasma levels of soluble CD14 independently predict mortality in HIV infection
.
J Infect Dis
2011
;
203
:
780
90
.

58

Hunt
PW
,
Shive
C
,
Clagett
B
,
Funderburg
N
,
Nattat
MV
et al. .
Gut epithelial barrier dysfunction, inflammation, and coagulation predict higher mortality during treated HIV/AIDS [abstract 278]
. In
: Program and Abstracts from the 19th Conference on Retroviruses and Opportunistic Infections
,
Seattle, Washington
,
2012
.

59

Tenorio
A
,
Zheng
E
,
Bosch
R
et al. .
Soluble markers of inflammation and coagulation, but not T-cell activation, predict nonAIDS- defining events during suppressive antiretroviral therapy [abstract 790]
. In:
Program and Abstracts from the 19th Conference on Retroviruses and Opportunistic Infections
,
Atlanta, Georgia,
2013
.

60

Tenorio
AR
,
Zheng
Y
,
Bosch
RJ
et al. .
Soluble markers of inflammation and coagulation but not T-cell activation predict non-AIDS-defining morbid events during suppressive antiretroviral treatment
.
J Infect Dis
2014
;
210
:
1248
59
.

61

Zhang
ZQ
,
Notermans
DW
,
Sedgewick
G
et al. .
Kinetics of CD4+ T cell repopulation of lymphoid tissues after treatment of HIV-1 infection
.
Proc Natl Acad Sci U S A
1998
;
95
:
1154
9
.

62

Estes
J
,
Baker
JV
,
Brenchley
JM
et al. .
Collagen deposition limits immune reconstitution in the gut
.
J Infect Dis
2008
;
198
:
456
64
.

63

Kim
CJ
,
McKinnon
LR
,
Kovacs
C
et al. .
Mucosal Th17 cell function is altered during HIV infection and is an independent predictor of systemic immune activation
.
J Immunol
2013
;
191
:
2164
73
.

64

Chun
TW
,
Nickle
DC
,
Justement
JS
et al. .
Persistence of HIV in gut-associated lymphoid tissue despite long-term antiretroviral therapy
.
J Infect Dis
2008
;
197
:
714
20
.

65

Tilg
H
,
Moschen
A
,
Kaser
A
.
Mode of function of biological anti-TNF agents in the treatment of inflammatory bowel diseases
.
Expert Opin Biol Ther
2007
;
7
:
1051
9
.

66

Hold
GL
,
Smith
M
,
Grange
C
,
Watt
ER
,
El-Omar
EM
,
Mukhopadhya
I
.
Role of the gut microbiota in inflammatory bowel disease pathogenesis: what have we learnt in the past 10 years?
World J Gastroenterol
2014
;
20
:
1192
210
.

67

Vujkovic-Cvijin
I
,
Dunham
RM
,
Iwai
S
et al. .
Dysbiosis of the gut microbiota is associated with HIV disease progression and tryptophan catabolism
.
Sci Transl Med
2013
;
5
:
193ra91
.

68

Dillon
SM
,
Lee
EJ
,
Kotter
CV
et al. .
An altered intestinal mucosal microbiome in HIV-1 infection is associated with mucosal and systemic immune activation and endotoxemia
.
Mucosal Immunol
2014
;
7
:
983
94
.

69

Mutlu
EA
,
Keshavarzian
A
,
Losurdo
J
et al. .
A compositional look at the human gastrointestinal microbiome and immune activation parameters in HIV infected subjects
.
PLoS Pathog
2014
;
10
:
e1003829
.

70

Lozupone
CA
,
Li
M
,
Campbell
TB
et al. .
Alterations in the gut microbiota associated with HIV-1 infection
.
Cell Host Microbe
2013
;
14
:
329
39
.

71

Vazquez-Castellanos
JF
,
Serrano-Villar
S
,
Latorre
A
et al. .
Altered metabolism of gut microbiota contributes to chronic immune activation in HIV-infected individuals
.
Mucosal Immunol
2015
;
8
:
760
72
.

72

Yu
G
,
Fadrosh
D
,
Ma
B
,
Ravel
J
,
Goedert
JJ
.
Anal microbiota profiles in HIV-positive and HIV-negative MSM
.
AIDS
2014
;
28
:
753
60
.

73

Noguera-Julian
M
,
Rocafort
M
,
Guillen
Y
et al. .
Gut microbiota linked to sexual preference and HIV infection
.
EBioMedicine
2016
;
5
:
135
46
.

74

Le Chatelier
E
,
Nielsen
T
,
Qin
J
et al. .
Richness of human gut microbiome correlates with metabolic markers
.
Nature
2013
;
500
:
541
6
.

75

Manichanh
C
,
Borruel
N
,
Casellas
F
,
Guarner
F
.
The gut microbiota in IBD
.
Nat Rev Gastroenterol Hepatol
2012
;
9
:
599
608
.

76

Nowak
P
,
Troseid
M
,
Avershina
E
et al. .
Gut microbiota diversity predicts immune status in HIV-1 infection
.
AIDS
2015
;
29
:
2409
18
.

77

McKenna
P
,
Hoffmann
C
,
Minkah
N
et al. .
The macaque gut microbiome in health, lentiviral infection, and chronic enterocolitis
.
PLoS Pathog
2008
;
4
:
e20
.

78

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
66
.

79

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
7
.

80

Monaco
CL
,
Gootenberg
DB
,
Zhao
G
et al. .
Altered Virome and Bacterial Microbiome in Human Immunodeficiency Virus-Associated Acquired Immunodeficiency Syndrome
.
Cell Host Microbe
2016
;
19
:
311
22
.

81

Klase
Z
,
Ortiz
A
,
Deleage
C
et al. .
Dysbiotic bacteria translocate in progressive SIV infection
.
Mucosal Immunol
2015
;
8
:
1009
20
.

82

Kartalija
M
,
Sande
MA
.
Diarrhea and AIDS in the era of highly active antiretroviral therapy
.
Clin Infect Dis
1999
;
28
:
701
5
;
quiz 6-7
.

83

Kok
A
,
Hocqueloux
L
,
Hocini
H
et al. .
Early initiation of combined antiretroviral therapy preserves immune function in the gut of HIV-infected patients
.
Mucosal Immunol
2015
;
8
:
127
40
.

84

Kristoff
J
,
Haret-Richter
G
,
Ma
D
et al. .
Early microbial translocation blockade reduces SIV-mediated inflammation and viral replication
.
J Clin Invest
2014
;
124
:
2802
6
.

85

Sandler
NG
,
Zhang
X
,
Bosch
RJ
et al. .
Sevelamer does not decrease lipopolysaccharide or soluble CD14 levels but decreases soluble tissue factor, low-density lipoprotein (LDL) cholesterol, and oxidized LDL cholesterol levels in individuals with untreated HIV infection
.
J Infect Dis
2014
;
210
:
1549
54
.

86

Somsouk
M
,
Dunham
RM
,
Cohen
M
et al. .
The immunologic effects of mesalamine in treated HIV-infected individuals with incomplete CD4+ T cell recovery: a randomized crossover trial
.
PLoS One
2014
;
9
:
e116306
.

87

Tenorio
AR
,
Chan
ES
,
Bosch
RJ
et al. .
Rifaximin has a marginal impact on microbial translocation, T-cell activation and inflammation in HIV-positive immune non-responders to antiretroviral therapy - ACTG A5286
.
J Infect Dis
2015
;
211
:
780
90
.

88

Pandrea
I
,
Xu
C
,
Stock
JL
et al. .
Antibiotic and Antiinflammatory Therapy Transiently Reduces Inflammation and Hypercoagulation in Acutely SIV-Infected Pigtailed Macaques
.
PLoS Pathog
2016
;
12
:
e1005384
.

89

Stiksrud
B
,
Nowak
P
,
Nwosu
FC
et al. .
Reduced Levels of D-dimer and Changes in Gut Microbiota Composition After Probiotic Intervention in HIV-Infected Individuals on Stable ART
.
J Acquir Immune Defic Syndr
2015
;
70
:
329
37
.

90

Villar-Garcia
J
,
Hernandez
JJ
,
Guerri-Fernandez
R
et al. .
Effect of probiotics (Saccharomyces boulardii) on microbial translocation and inflammation in HIV-treated patients: a double-blind, randomized, placebo-controlled trial
.
J Acquir Immune Defic Syndr
2015
;
68
:
256
63
.

91

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
.

92

Gori
A
,
Rizzardini
G
,
Van't Land
B
et al. .
Specific prebiotics modulate gut microbiota and immune activation in HAART-naive HIV-infected adults: results of the “COPA” pilot randomized trial
.
Mucosal Immunol
2011
;
4
:
554
63
.

93

Gonzalez-Hernandez
LA
,
Jave-Suarez
LF
,
Fafutis-Morris
M
et al. .
Synbiotic therapy decreases microbial translocation and inflammation and improves immunological status in HIV-infected patients: a double-blind randomized controlled pilot trial
.
Nutr J
2012
;
11
:
90
.

94

Li
SS
,
Zhu
A
,
Benes
V
et al. .
Durable coexistence of donor and recipient strains after fecal microbiota transplantation
.
Science
2016
;
352
:
586
9
.

95

Somsouk
M
,
Vujkovic-Cvijin
I
,
Pao
M
,
Hunt
PW
,
Lynch
SV
,
Mccune
JM
.
Fecal microbial transplantation: safety and engraftment during treated HIV infection [abstract 264]
. In:
Program and Abstracts from the 23rd Conference on Retroviruses and Opportunistic Infections
,
Boston, Massachusetts
,
2016
.

96

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
9
.

97

Micci
L
,
Ryan
ES
,
Fromentin
R
et al. .
Interleukin-21 combined with ART reduces inflammation and viral reservoir in SIV-infected macaques
.
J Clin Invest
2015
;
125
:
4497
513
.

98

Thiebaut
R
,
Jarne
A
,
Routy
JP
et al. .
Repeated Cycles of Recombinant Human Interleukin 7 in HIV-Infected Patients With Low CD4 T-Cell Reconstitution on Antiretroviral Therapy: Results of 2 Phase II Multicenter Studies
.
Clin Infect Dis
2016
;
62
:
1178
85
.