Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2014 Sep 4.
Published in final edited form as: Mucosal Immunol. 2013 Apr 3;6(4):657–665. doi: 10.1038/mi.2013.15

Mucosal immunity in human and simian immunodeficiency lentivirus infections

JM Brenchley 1
PMCID: PMC4154146  NIHMSID: NIHMS620138  PMID: 23549448

Abstract

Overwhelming evidence indicates that distinct pathological phenomenon occurs within the gastrointestinal (GI) tract of progressively simian immunodeficiency virus (SIV)-infected Asian macaques and HIV-infected humans compared with other anatomical sites. Massive loss of GI tract lamina propria CD4 T cells, alteration in the profile of lymphocytic cytokine production, changes in the landscape of GI tract antigen-presenting cells, and variations to the structural barrier of the GI tract are hallmarks of progressive HIV/SIV infections. The pathology within the GI tract results in translocation of microbial products from the lumen of the intestine into peripheral circulation. These translocated microbial products directly stimulate the immune system and exacerbate immune activation and, thus, disease progression. Initiation of combination antiretroviral therapy (cART) does not restore completely the immunological abnormalities within the GI tract. This incomplete restoration within the GI tract may contribute to the increased mortality observed within HIV-infected individuals treated for decades with cART. Novel therapeutic interventions aimed at enhancing GI tract anatomy and physiology may improve the prognosis of HIV-infected individuals.

INTRODUCTION

Given that HIV and simian immunodeficiency virus (HIV/SIV) are cytopathic viruses and that they have tropism for leukocytes that express the receptor CD4 and one of a set of chemokine receptors, it follows that progressive loss of CD4 T cells is a hallmark of the disease and is, ultimately, associated with the development of AIDS.14 Although the virus has specific tropism for CD4 T cells and loss of these cells underpins susceptibility to opportunistic infections, cessation of viral replication does not appear to allow HIV/SIV-infected individuals to return to health completely. Indeed, the immunological pathology that occurs during the course of untreated HIV/SIV infections cannot be completely reversed by therapeutic administration of combination antiretroviral therapy (cART). The pathological occurrences during the course of untreated, progressive HIV/SIV infections are complex and multifaceted. In particular, there are specific pathological occurrences within the GI tract that are thought to be of critical importance in perpetuating disease progression.

The gastrointestinal (GI) tract is the predominant structural and immunological barrier against the microorganisms of the outside world. In addition, the enterocytes that form the single cell layer of the mucosal epithelium provide essential roles in nutrient absorption. Hence, pathology to the structural and immunological anatomy of the GI tract during progressive HIV/SIV infections results in multiple deleterious sequelae. Here I review the numerous alterations observed within the GI tracts of HIV-infected individuals and/or SIV-infected Asian macaques and how these are thought to influence the overall health of such individuals and therapeutic interventions that might restore the anatomy and physiology of the GI tract.

PATHOLOGY WITHIN THE GI TRACT

In 1983, HIV was defined as the infectious agent that causes AIDS.5,6 Only one year later, in 1984, Kotler et al.7 observed that HIV-infected individuals had histological abnormalities of the GI mucosa. These observed abnormalities included malabsorption and lymphocyte depletion and they concluded that: “The histologic findings suggest that a specific pathologic process occurs in the lamina propria of the small intestine and colon in some patients with the syndrome.” This finding was incredibly insightful in its anticipation of subsequent discoveries. Indeed, the term “HIV enteropathy” has been appreciated for as long as it has been known that HIV causes AIDS. The enteropathy that afflicts HIV-infected individuals can occur from the acute phase of the infection through to advanced disease. It involves diarrhea, increased GI inflammation, increased intestinal permeability, and malabsorption.810 Subsequently, considerable effort has investigated immunological abnormalities within the GI tract of progressively HIV/SIV-infected individuals, and overwhelming evidence suggests distinct immunological pathologies within the GI tract compared with other anatomical sites.

Loss of CD4 T cells

There is overwhelming evidence to suggest that CD4 T cells are disproportionately depleted from the lamina propria of the GI tract compared with peripheral blood of acutely HIV-infected humans and SIV-infected Asian macaques. Indeed, after the original observations of enteropathy and histological abnormalities in the HIV-infected GI tract, immunohistochemical analysis showed a proinflammatory infiltration of lymphocytes, yet concomitant with a striking absence of CD4 T cells.11,12 These findings lead to the suggestion that the intestinal mucosa could be a site of significant HIV replication and CD4 T-cell infection and consequent depletion.12 Subsequently, other studies also using immunohistochemistry confirmed that the GI tract was, indeed, significantly depleted of CD4 T cells.1315 The mechanism underlying this preferential loss of GI tract CD4 T cells is likely multifaceted; however, it seemed unlikely that HIV coincidentally has tropism for CD4 T cells and these were the very cells depleted. Studies in SIV-infected Asian macaques has shown significant viral replication within the GI tracts of acutely SIV-infected animals, suggesting much of the CD4 T-cell depletion is attributed to direct infection with SIV.16,17

Use of flow cytometric analysis has also allowed for quantitative examination and phenotypic assessment of GI CD4 T cells in HIV/SIV-infected individuals.1825 Several of these studies examined CD4/CD8 ratios in intestinal biopsies and peripheral blood to compare CD4 T-cell depletion between these two anatomical sites, and these studies also suggested that GI tract was preferentially depleted of CD4 T cells. Additional studies that concentrated on examining depletion of the specific cells targeted by HIV—CD4 T cells that express the HIV coreceptor CCR5 (chemokine (C-C motif) receptor 5)—have shown that these cells are almost completely depleted from GI tract tissues.2226 Indeed, phenotypic analysis of CD4 T cells in GI mucosa of healthy individuals demonstrated that the majority of T cells in mucosal tissues expressed CCR5 and were very permissive to in vitro infections with HIV.27,28

Altered functionality of GI tract lymphocytes

Although CD4 T-cell depletion is nearly complete in progressively HIV-infected humans and SIV-infected Asian macaques, it is important to note that CD4 T-cell depletion is not the only abnormality observed within the GI tract. Indeed, there are many other pathological phenomena that likely contribute to disease progression. In healthy individuals, GI tract lymphocytes contribute to antibacterial defenses and can promote maintenance of the structural barrier of the GI tract. In particular, specialized lymphocytic subsets that reside in the GI tract are capable of producing effector cytokines such as interleukin-17 (IL-17) and interleukin-22 (IL-22). These cytokines can recruit neutrophils and myeloid cells to effector sites by inducing granulocyte colony-stimulating factor,29 and are involved in epithelial regeneration in mucosal tissues.30 This IL-22/IL-17-mediated maintenance of the mucosal layers is associated with induction expression of claudins (which are protein components of epithelial tight junctions), defensins (which have antibacterial activities), and mucin.31 Moreover, IL-22 is, itself, a growth factor for epithelial cells as the majority of epithelial cells in the body express the IL-22 receptor and IL-22 can cause epithelial cells to divide in vitro.32

Several studies have shown that although CD4 T cells are massively depleted from the GI tracts of progressively HIV/SIV-infected individuals, CD4 T cells that produce the effector cytokine IL-17 are preferentially depleted.3335 Given the known roles for IL-17/IL-22 in maintenance of mucosal barriers, preferential loss of T helper type 17 (Th17) cells was hypothesized to provide mechanistic insights into damage to the structural barrier of the GI tract observed in chronically HIV/SIV-infected individuals.3639 However, although CD4 Th17 cells are classically considered to be the predominant source of IL-17 and IL-22, recent studies have clearly demonstrated that subsets of CD8 T cells and innate lymphocytes are also capable of producing these effector cytokines. Subsequent studies have demonstrated that the CD8 T cells and innate lymphocytes that produce IL-17 and IL-22 are, similar to Th17 cells, present at decreased frequencies within the GI tracts of HIV/SIV-infected individuals.4043 This immunological skewing away from GI tract lymphocytes capable of producing IL-17 is associated with systemic immune activation36,40 and focal damage to the structural barrier of the GI tract (discussed below).40 Taken together, these data suggest that immunological devastation within the GI tract is a critically important factor to consider for HIV/SIV disease progression. Understanding the mechanisms underlying this immunological abnormality may lead to novel therapeutic interventions to decrease systemic immune activation that predicts disease progression better than either plasma viral loads of peripheral blood CD4 T-cell counts.1

That IL-17/IL-22-producing CD8 T cells, CD4 T cells, and innate lymphocytes are present at decreased frequencies within the GI tracts of SIV-infected animals provides some insight into the mechanisms underlying this observed decrease. Indeed, as neither CD8 T cells nor innate lymphocytes express the CD4 receptor for HIV/SIV, it seems unlikely that preferential infection by HIV/SIV in progressively infected individuals underlies depletion of IL-17/IL-22-producing cells. Moreover, we previously used quantitative real time-PCR for HIV DNA and flow cytometric sorting of mitogenically stimulated lymphocytes from chronically HIV-infected individuals to determine the relative infection frequencies of Th17 and Th1 cells and found equal infection of Th1 and Th17 cells.33 Given the uniformity in loss of all IL-17/IL-22-producing lymphocytes and lack of preferential HIV/SIV infection of Th17 cells, several groups began examining the nature of the landscape of antigen-presenting cells (APCs) within the GI tract of SIV/HIV-infected individuals. These studies have uniformly demonstrated significant alterations to the APC landscape in progressively infected GI tracts. High numbers of APCs expressing the tryptophan-metabolizing enzyme indoleamine 2,3 dioxygenase (IDO) have been observed in chronically HIV-infected invididuals,44,45 and IDO activity (measured by serum levels of tryptophan and tryptophan metabolites) has been associated with low frequencies of Th17 cells in progressively HIV-infected individuals.46 Moreover, the metabolites produced by IDO directly inhibited IL-17-producing lymphocytes in vitro.42 The high levels of IDO+ APCs observed in HIV/SIV-infected individuals was associated with decreased frequencies of CD103+ APCs, which are thought to provide factors required for differentiation and maintenance of IL-17/IL-22-producing lymphocytes.40 CD103 + APCs were also excellent in inducing Th17 cells from naive CD4 T cells in vitro.40 Hence, alterations to the landscape of GI tract APCs may limit the ability of progressively HIV/SIV-infected individuals to maintain IL-17/IL-22-producing lymphocytes.

Structural alterations of the GI tract

Given that the majority of epithelial cells in the body express the IL-22 receptor,32 loss of lymphocytes producing IL-17 and IL-22 might be expected to have a detrimental effect on maintenance of the GI tract epithelial barrier. Indeed, the tight epithelial barrier is a single layer of cells that serves to separate the host from the enormous luminal bacterial microbiome. The epithelium also absorbs luminal nutrients. Several studies have indicated impaired integrity of the epithelial barrier of the GI tract in chronically HIV-infected individuals. Indeed, as previously stated, Kotler et al.7 originally observed malabsorption, and lymphocyte depletion in HIV-infected individuals. This enteropathy afflicting HIV-infected individuals could occur from the acute phase of the infection through to advanced disease and involved diarrhea, increased GI inflammation, increased intestinal permeability, and malabsorption.810 Demonstration of damage to the structural barrier of the GI tract has been provided by focal breaches in the barrier, massive apoptosis of enterocytes, decreased expression of epithelial repair tight junction genes, and increased GI tract permeability.10,4750 Indeed, focal breaches of the epithelial barrier were in spatial juxtaposition to infiltrating microbial products in vivo, and the extent of damage to the GI tract barrier during HIV/SIV was directly associated with the degree of microbial translocation, both locally within the GI tract and systemically (discussed below).4749

The mechanisms underlying damage to the structural barrier of the GI tract are likely numerous and complex. Given the data published, it is reasonable to suggest that the focal damage observed to the structural barrier of the GI tract is attributed, at least in part, to the alterations to the immunological environment within the GI tract. Indeed, the acute inflammatory response associated with acute HIV/SIV infection is a very likely culprit in initiating a cyclical process. Part of the acute inflammatory process observed in HIV infection involves production of tumor necrosis factor (TNF).51 Although production of TNF is a critical immunological response, overproduction of TNF can have deleterious effects on the epithelial barrier of the GI tract. Indeed, exposure to TNF could induce apoptosis in epithelial cells in vitro,52 and in vivo studies also suggested that excess production of TNF perturbs the tight epithelial barrier, in part by induction of enterocyte apoptosis (reviewed in ref. 53). Consistent with this premise, therapeutic interventions aimed solely at decreasing TNF levels in vivo resulted in improved integrity of the structural barrier of the GI tract in individuals with Crohn’s disease.54 There are also recent data demonstrating that in vivo treatment with anti-TNF therapies ameliorated some of the pathogenic consequences of acute SIV infection in Asian macaques.55 However, the efficacy of anti-TNF therapy in decreasing enterocyte apoptosis and focal damage to the structural barrier of the GI tract during progressive SIV infection is yet to be examined. After GI tract damage has been initiated, alterations in the functionality of local lymphocyte populations (discussed above) likely contributes to an inability to repair the damage considering that the amount of damage to the structural barrier of the GI tract is negatively associated with the degree to which IL-17/IL-22-producing lymphocytes are decreased.40

MUCOSAL HIV/SIV-SPECIFIC CELLULAR RESPONSES

The vast majority of HIV transmission occurs via introduction of the virus to a mucosal site.56 Moreover, after acquisition of the virus, the first cells that become productively infected, leading to viral spread, appear to be mucosal CD4 T cells.17 As such, there is significant effort in producing vaccines that stimulate HIV/SIV-specific immune responses within mucosal tissues.57 The immune response that is elicited during natural infection is, in general, insufficient to control viral replication. However, a small fraction (<0.5%) of HIV-infected individuals, termed elite controllers, spontaneously control viral replication to undetectable levels and many of these individuals express the HLA types HLA B57 or HLA B27.58 It is widely believed that cellular immune responses mounted against the virus by such elite controllers is responsible for control of viral replication.58 Thus, there has been significant attention given to understanding the types of HIV-specific and SIV-specific immune responses elicited in elite controllers. Similar frequencies of HIV-specific CD8 T cells are present within the mucosa of the GI tract in cohorts of HIV-infected individuals irrespective of their ability to control viral replication.22,59 Although the magnitude of the HIV-specific CD8 T-cell response does not appear to differ between elite controllers and individuals with progressive HIV disease, the functional quality of the HIV-specific CD8 T-cell response appears to be improved in elite controllers.60 In particular, HIV-specific CD8 T cells in elite controllers are able to better simultaneously produce several effector cytokines compared with HIV-specific CD8 T cells in individuals with detectable plasma viremia. Similar multifunctionality is also observed among HIV-specific CD4 T cells in the GI tract of elite controllers compared with individuals with detectable plasma viremia.61 This association between increased functionality of HIV-specific T cells and decreased pathology is also across different anatomical sites within individuals. Indeed, increased functionality of HIV-specific T cells in broncheoalveolar lavage is associated with increased frequencies of CD4 T cells in broncheoalveolar lavage compared with the GI tract.59

Although the functionality of virus-specific T cells has been associated with controlled viral replication in vivo, it is unclear whether the polyfunctional T cells control viral replication or the polyfunctional T cells result from low levels of viral replication. To help clarify a potential role for T cells in controlling viral replication, Hansen et al.62,63 developed a replication-competent cytomegalovirus (CMV) viral vector expressing SIV proteins. Infection with the CMV vector induced high frequencies of SIV-specific CD8 T cells with an effector memory phenotype and these effector memory CD8 T cells could protect rhesus macaques from mucosal SIV infection. The degree of protection against mucosal SIV infection was associated with the degree of SIV-specific effector memory CD8 T cells elicited by the CMV vectors.63 Hence, specific functional and phenotypic subsets of T-cell immunity against HIV/SIV appear to provide protection against viral replication in vivo. The superior protection against infection seen in animals vaccinated with CMV vectors compared with animals vaccinated with adenovirus vectors may be attributed to the increased differentiation status of the virus-specific CD8 T cells induced by CMV vectors compared with virus-specific CD8 T cells induced by adenovirus vectors that tend to be less differentiated and express surface markers such as CD27 and CD28.64

In addition to studies of virus-specific T-cell responses, much effort has gone into understanding humoral responses that neutralize broad stains of HIV (reviewed in ref. 65). Indeed, preventing initial infection events would likely require a mucosal humoral immune response. Consistent with this premise, application of SIV-specific neutralizing antibodies to the vaginal surface can prevent vaginal infection of rhesus macaques using a pathogenic strain of SIV.66 Moreover, evidence of protective humoral immunity in HIV comes from observations that individuals repetitively exposed to the virus and yet who remain uninfected produce HIV-specific immunoglobulin A antibodies at mucosal sites.67,68 However, the majority of chronically HIV-infected individuals do not mount vigorous HIV-specific immunoglobulin A antibody responses in mucosal sites or systemically.69 Thus, these findings suggest that induction of a functional HIV-specific immune response would aid in control of viral replication and may inhibit viral replication. However, vaccines that induce immunological responses that prevent acquisition of HIV do not currently exist and significant pathology within the GI tract occurs during progressive infection and the majority of HIV-infected individuals will succumb to AIDS if not treated with cART.

MICROBIAL TRANSLOCATION

That the human GI tract is colonized with approximately 1014 normal flora bacteria, the structural and immunological perturbations to the GI tracts in progressively HIV/SIV-infected individuals was hypothesized to result in microbial translocation.70 Microbial translocation during HIV infection was first described in 2006, where it was demonstrated that bioactive microbial products were significantly elevated in plasma from HIV-infected individuals and from progressively SIV-infected Asian macaques.71 Furthermore, the levels of lipopolysaccharide (LPS) in these individuals directly correlated with activation of both the adaptive and innate arms of the immune system.71 Subsequently, at least 50 groups corroborated the existence of microbial translocation during progressive HIV/SIV infections (reviewed in refs. 72 and 73). Hence, there is overwhelming evidence that the immunological and epithelial damage that occur in progressively HIV/SIV infected individuals lead to an inability of the host to restrict the microbiome and all of its immunostimulatory components to the lumen of the GI tract. Moreover, it is quite clear that the microbial products that translocate during progressive HIV/SIV infections are bioactive in vivo and exacerbate chronic immune activation. However, the degree to which microbial translocation contributes to the chronic immune activation is less clear. Indeed, microbial translocation is but one cause of immune activation in chronically HIV/SIV-infected individuals and other factors likely contribute. There has been only one study that did not observe microbial translocation in chronically HIV-infected individuals.74 Here the authors used repeatedly thawed plasma samples from longitudinally sampled Ugandans before infection and then into the chronic phase of HIV infection and failed to find elevated plasma LPS or soluble CD14 (sCD14). However, in a follow-up study using exactly the same plasma samples, the authors found elevated levels of plasma C-reactive protein that correlated with sCD14.75 It is unclear why these two reports from the same authors appear to contradict one another and why the findings of these authors were inconsistent with the numerous other reports of microbial translocation in chronic HIV infection, but factors such as nonfasting blood draws and repeated freeze thaws of plasma samples might have played a role.

Consequences of microbial translocation

To understand more completely the degree to which microbial translocation, as opposed to HIV/SIV replication, contribute to immune activation and disease progression, several groups have turned attention to chronically HIV/SIV-infected individuals in which the contribution of viral replication is diminished. Indeed, there are at least two cohorts of HIV-infected individuals in which viral replication is reduced to a minimum: elite controllers and cART-treated individuals. Although elite controllers have a significantly improved prognosis compared with viremic HIV-infected individuals, they tend to have higher levels of immune activation compared with HIV-uninfected individuals, and some of these individuals nevertheless lose peripheral blood CD4 T cells and some even progress to AIDS.4 Elevated levels of plasma LPS have been detected in elite controllers that positively correlated with the frequency of activated phenotype CD38+HLA-DR+ CD8 T cells and negatively with peripheral blood CD4 T-cell counts.4

Similar studies have been performed using samples from cART-treated individuals. Current era cART is sufficient to control HIV replication to undetectable levels for decades.76 However, recent studies have shown clearly that although cART can suppress plasma viral loads to undetectable levels, cART-treated individuals nevertheless have increased mortality and morbidity. This increased mortality was closely associated with residual inflammation, which was in turn associated with cardiovascular disease,77,78 osteopenia,79 and cognitive decline.80 Given that viral replication is decreased to very low levels in these individuals, it seems highly unlikely that the residual inflammation is directly attributable to ongoing viral replication. This increased mortality seemed to be linked to microbial translocation given that elevated plasma levels of sCD14 (a bacterial LPS receptor that is produced after LPS stimulation in vivo) independently predict increased mortality in cART-treated HIV-infected individuals.78 Moreover, plasma levels of sCD163 (a scavenger receptor) were associated with unstable noncalcified coronary plaques in cART-treated HIV-infected individuals.78,81 Consistent with the premise that microbial translocation contributes to immune activation and immunological pathology, microbial translocation has been suggested to play an important role in limiting immunological reconstitution of peripheral blood CD4 T cells after administration of cART.71,8287

Given the strength of the data suggesting microbial translocation plays a major role in disease pathogenesis of HIV-infected individuals treated with long-term cART, it is important to consider mechanisms underlying the increased microbial translocation and consequent immune activation in these individuals. Significant efforts have been taken to understand more completely the state of the GI tract in HIV-infected individuals treated for long periods of time with cART. From these studies, it is quite clear that significant pathology remains despite cART adequately controlling systemic viral replication to very low levels. Several studies of the small intestine of cART-treated, HIV-infected individuals have concurred that CD4 T-cell reconstitution is poor and occurs at a much slower rate than the CD4 T-cell reconstitution observed in peripheral blood.23,8890 Moreover, individuals treated with cART during the early stages of HIV infection have improved reconstituted CD4 T cells in the GI tract compared with individuals treated with cART during chronic HIV infection (who rarely reconstituted significant numbers of GI tract CD4 T cells).89,90 Continued depletion of GI tract CD4 T cells after administration of cART likely contributes to an inability to preserve the integrity of the GI tract that has been suggested from gene chip analysis of epithelial cells where decreased expression of GI tract repair genes was observed,90 which is also consistent with the premise that the local immunological environment influences the ability of the host to maintain epithelial integrity (discussed above). The observed inability of cART-treated, HIV-infected individuals to reconstitute healthy frequencies of GI tract CD4 T cells was related to inflammation-associated fibrosis within Peyer’s patches and lamina propria of the GI tract,91,92 suggesting that fibrosis to the GI tract prohibits healthy immunological responses. Fibrosis appears to limit bioavailability of IL-7 within the lymphoid tissue that might provide mechanistic insight into how fibrosis limits reconstitution of CD4 T cells.93

GI TRACT BACTERIOME/VIROME

Although it is clear that microbial translocation occurs during progressive HIV/SIV infections and contributes to immune activation, microbial translocation also occurs in other diseases associated with inflammation of the GI tract. In other diseases where microbial translocation has been implicated in pathogenesis, an altered balance in the composition of the commensal microflora (dysbiosis) has been suggested to play an important role in the disease.94,95 From this, several hypothesized that dysbiosis may facilitate microbial translocation and consequent immune activation in progressive HIV/SIV infections. Cross-sectional analysis of the fecal microbiome of different cohorts of individuals seemed to suggest that dysbiosis in HIV-infected individuals may contribute to disease progression. These studies tended to agree that HIV-infected individuals had increased levels of potentially pathogenic fecal bacteria including clostridia and proteobacter species and lower levels of probiotic organisms such as Lactobacilli and Bifidobacteria.9698 Moreover, higher levels of fecal proteobacteria were associated with local immune activation within the GI tract.99

The existence of the SIV, nonhuman primate model of HIV infection allowed for the longitudinal sampling of feces at different amounts of time after infection. This allowed for a more controlled examination of the microbiota while minimizing differences that might be attributed to lifestyle and diet. Three studies have used next-generation sequencing approaches to study the composition of the fecal micro-biota in SIV-infected and SIV-uninfected nonhuman primates.100102 Two of these studies examined the microbiome of cross-sectional cohorts of SIV-infected and SIV-uninfected animals,100,101 and one study examined the microbiome of several animals longitudinally, before and after SIV infection.102 There was agreement among all three studies regarding the major communities of fecal microbes within nonhuman primates and there was agreement among all three studies that SIV infection did not result in significant alteration in the fecal bacteriome of progressively SIV-infected Asian macaques.

The bacteriome is clearly the largest microbial community within the GI tract; however, other potentially pathogenic microorganisms can also contribute to the microbiome. To examine the composition of the microbiome of progressively HIV/SIV-infected individuals in a more unbiased approach, Handley et al.101 used 454 “shot gun” sequencing technology to analyze all DNA and RNA species present in fecal samples. The authors found a significant increase in the size of the fecal virome in SIV-infected Asian macaques and the virome contained potentially pathogenic viruses, and adenoviruses, in particular, were found to colocalize with areas of damage to the structural barrier of the GI tract. Importantly, these same fecal viruses were absent in SIV-infected natural hosts of SIV who manifest a nonprogressive infection with SIV. Hence, subclinical GI tract viral infections might contribute to microbial translocation and immune activation in progressive HIV/SIV infections.

Given the discrepancy between studies of HIV-infected humans and SIV-infected Asian macaques, it is unclear if the GI tract bacteriome is significantly altered during progressive HIV/SIV infections. One possible explanation for the discrepancy is inherent differences between the cohorts of individuals examined in the human studies and not the viral infection per se. With the discrepancies between data retrieved from nonhuman primates and humans, and the lack of comparable virome analysis in humans, and considering that is not entirely clear whether dysbioses cause or result from immune activation and immunodeficiency (or both), further studies are certainly warranted.

TREATMENTS

With the several studies indicating that microbial translocation plays an important role in exacerbating immune activation and, potentially, in comorbidities in cART-treated individuals, therapeutic interventions aimed at decreasing microbial translocation have been considered. The therapeutic intervention aimed at decreasing microbial translocation that has been investigated most thoroughly is probiotic bacteria. Probiotic bacteria are thought to impart benefits to the host via a variety of mechanisms including production of short-chain fatty acids (important for proliferation of epithelial cells), competition with potentially pathogenic bacteria for nutrients, and decrease in GI tract inflammation. Several studies have investigated potential benefits imparted to HIV-infected individuals by probiotic dietary supplementation. The majority of these studies have focused analyses on systemic effects of probiotic supplementation. Three studies have found modest improvement in reconstitution of peripheral blood CD4 T cells and decreased GI disorders after treatment with cART and probiotics.103106 A fourth study using two probiotic organisms found moderately decreased microbial translocation and immune activation after probiotic supplementation of cART.107 A fifth study aimed to alter the composition of the GI tract microbiota by supplementing cART with a unique oligosaccharide mixture of “prebiotics” that are thought to promote the growth of probiotic organisms.98 The prebiotic treatment was, indeed, associated with increased probiotic organisms and treated individuals had decreased markers of microbial translocation and systemic immune activation compared with placebo-treated individuals.98 Finally, a sixth study using a combination of four probiotic organisms and a combination of prebiotics found marginal decreases in immune activation in individuals treated with these “synbiotics” and cART.108 Importantly, probiotics were associated with some sort of immunological improvements in all five human studies. However, the mechanisms by which probiotics exerted these effects was unclear. Moreover, with the multitude of species of bacteria that have probiotic characteristics, it is unclear which probiotics might be most beneficial to cART-treated, HIV-infected individuals. Of the numerous species of bacteria that have been used as probiotics, the two formulations of probiotics that have been most exhaustively studied in other diseases characterized by microbial translocation are Lactobacillus rhamnosis GG and VSL-3.109112 VSL-3 is a combination of four strains of Lactobacilli, three strains of Bifidobacteria, and one strain of Streptococcus. A combination of Lactobacillus GG and VSL-3 was recently shown to impart an immunological benefit to cART-treated, SIV-infected Asian macaques.102 This probiotic treatment led to decreased fibrosis within GI tract lymphoid follicles that was associated with increased reconstitution of GI tract CD4 T cells. Indeed, in these animals, just 5 months of therapy was sufficient for reconstitution of GI tract CD4 T cells to near healthy levels.102 Hence, the immunological benefits that seem to be imparted by probiotic use likely involves restoration of GI tract physiology. It is important to note, however, that these published studies have, in general, involved fairly small sample sizes and the largest effects of the probiotic treatments tend to be within the GI tract itself. Moreover, there are numerous preparations of probiotic therapeutics and it is unclear if all probiotics will have similar effects. Hence, large placebo interventional trials are certainly warranted.

Probiotics are one of many therapeutic interventions that have been envisioned to decrease microbial translocation. Other therapeutic interventions aimed at decreasing microbial translocation attempt to remove microbial products from circulation. One study supplemented cART with oral hyper-immune bovine colostrum. The bovine colostrum comprised high levels of proteins thought to be capable of mediating clearance of microbial products from circulation. However, the colostrum-treated individuals did not have lower levels of immune activation or microbial translocation compared with placebo-treated individuals.113 These data suggested that the hyperimmune bovine colostrum was insufficient to clear microbial products from circulation and alternative modalities could be considered. A second study involved treating SIV-infected Asian macaques with sevelamer, a phosphate-binding molecule with anti-inflammatory properties thought to be attributed to LPS binding. Sevelamer-treated, SIV-infected animals had significantly reduced microbial translocation, immune activation, and cardiovascular comorbidities compared with control animals.114 A large, placebo-controlled study of sevelamer in HIV-infected humans is currently underway in the AIDS clinical trials group (A5296). Taken together, the data suggest that supplementation of cART with therapeutics aimed at decreasing microbial translocation imparts immunological benefits and may improve the prognosis of cART-treated, HIV-infected individuals.

CONCLUDING REMARKS

Dysfunction of the immunological and structural barriers of the GI tract are becoming pathognomonic with progressive HIV/SIV disease. There have been significant strides in understanding the mechanisms underlying damage to the GI tract, its consequences, and in developing therapeutic interventions aimed in restoring the function of the GI tract. Although the precipitating events that occur during the acute phase of HIV/SIV infections leading to gastrointestinal damage remain elusive, it is clear that local immune activation, loss of total CD4 T cells, altered functionality of GI tract lymphocytes and myeloid cells, fibrosis of lymphoid and lamina propria tissues, and increased turnover of GI tract epithelial cells all play an important role in GI tract dysfunction, allowing microbial products to translocate systemically. Preliminary data using therapeutic interventions aimed at decreasing microbial translocation and improving GI tract dysfunction in HIV/SIV-infected individuals are looking very promising, but effects of blocked microbial translocation on systemic immune activation need to be better parsed out. However, long-term studies will be required to determine whether or not the immunological benefits imparted to individuals treated with cART and microbial translocation-reducing therapeutics have decreased comorbidities associated with long-term cART treatment.

Acknowledgments

I thank the Cleveland Immunopathogenesis Consortium (BBC) for helpful discussions. This work was supported by the Intramural NIAID, US NIH program. 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 US Government.

Footnotes

DISCLOSURE

The author declared no conflict of interest.

References

  • 1.Giorgi JV, 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;179:859–870. doi: 10.1086/314660. [DOI] [PubMed] [Google Scholar]
  • 2.Salazar-Gonzalez JF, et al. Increased immune activation precedes the inflection point of CD4 Tcells and the increased serum virus load in human immunodeficiency virus infection. J Infect Dis. 1998;178:423–430. doi: 10.1086/515629. [DOI] [PubMed] [Google Scholar]
  • 3.Sodora DL, et al. Toward an AIDS vaccine: lessons from natural simian immunodeficiency virus infections of African nonhuman primate hosts. Nat Med. 2009;15:861–865. doi: 10.1038/nm.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Hunt PW, et al. Relationship between Tcell activation and CD4(+) Tcell count in HIV-seropositive individuals with undetectable plasma HIV RNA levels in the absence of therapy. J Infect Dis. 2008;197:126–133. doi: 10.1086/524143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Barre-Sinoussi F, et al. Isolation of a T-lympotropic retrovirus from a patient at risk for acquired immune deficiency syndrome (AIDS) Science. 1983;220:868–871. doi: 10.1126/science.6189183. [DOI] [PubMed] [Google Scholar]
  • 6.Gallo RC, et al. Isolation of human T-cell leukemia virus in acquired immune deficiency syndrome (AIDS) Science. 1983;220:865–867. doi: 10.1126/science.6601823. [DOI] [PubMed] [Google Scholar]
  • 7.Kotler DP, Gaetz HP, Lange M, Klein EB, Holt PR. Enteropathy associated with the acquired immunodeficiency syndrome. Ann Intern Med. 1984;101:421–428. doi: 10.7326/0003-4819-101-4-421. [DOI] [PubMed] [Google Scholar]
  • 8.Kapembwa MS, et al. Altered small-intestinal permeability associated with diarrhoea in human-immunodeficiency-virus-infected Caucasian and African subjects. Clin Sci (Lond) 1991;81:327–334. doi: 10.1042/cs0810327. [DOI] [PubMed] [Google Scholar]
  • 9.Bjarnason I, et al. Intestinal inflammation, ileal structure and function in HIV. AIDS. 1996;10:1385–1391. doi: 10.1097/00002030-199610000-00011. [DOI] [PubMed] [Google Scholar]
  • 10.Sharpstone D, et al. Small intestinal transit, absorption, and permeability in patients with AIDS with and without diarrhoea. Gut. 1999;45:70–76. doi: 10.1136/gut.45.1.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.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;90:552–558. doi: 10.1016/0016-5085(86)91108-x. [DOI] [PubMed] [Google Scholar]
  • 12.Clayton F, et al. Rectal mucosal pathology varies with human immunodeficiency virus antigen content and disease stage. Gastroenterology. 1992;103:919–933. doi: 10.1016/0016-5085(92)90026-u. [DOI] [PubMed] [Google Scholar]
  • 13.Ullrich R, Zeitz M, Riecken EO. Enteric immunologic abnormalities in human immunodeficiency virus infection. Semin Liver Dis. 1992;12:167–174. doi: 10.1055/s-2007-1007388. [DOI] [PubMed] [Google Scholar]
  • 14.Kotler DP, Reka S, Clayton F. Intestinal mucosal inflammation associated with human immunodeficiency virus infection. Dig Dis Sci. 1993;38:1119–1127. doi: 10.1007/BF01295730. [DOI] [PubMed] [Google Scholar]
  • 15.Clayton F, Snow G, Reka S, Kotler DP. Selective depletion of rectal lamina propria rather than lymphoid aggregate CD4 lymphocytes in HIV infection. Clin Exp Immunol. 1997;107:288–292. doi: 10.1111/j.1365-2249.1997.236-ce1111.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Mattapallil JJ, et al. Massive infection and loss of memory CD4+ Tcells in multiple tissues during acute SIV infection. Nature. 2005;434:1093–1097. doi: 10.1038/nature03501. [DOI] [PubMed] [Google Scholar]
  • 17.Li Q, et al. Peak SIV replication in resting memory CD4+ Tcells depletes gut lamina propria CD4+ T cells. Nature. 2005;434:1148–1152. doi: 10.1038/nature03513. [DOI] [PubMed] [Google Scholar]
  • 18.Lim SG, et al. Loss of mucosal CD4 lymphocytes is an early feature of HIV infection. Clin Exp Immunol. 1993;92:448–454. doi: 10.1111/j.1365-2249.1993.tb03419.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Schneider T, et al. Loss of CD4 T lymphocytes in patients infected with human immunodeficiency virus type 1 is more pronounced in the duodenal mucosa than in the peripheral blood. Berlin Diarrhea/Wasting Syndrome Study Group. Gut. 1995;37:524–529. doi: 10.1136/gut.37.4.524. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Smit-McBride Z, Mattapallil JJ, McChesney M, Ferrick D, Dandekar S. Gastrointestinal T lymphocytes retain high potential for cytokine responses but have severe CD4+ T-cell depletion at all stages of simian immunodeficiency virus infection compared to peripheral lymphocytes. J Virol. 1998;72:6646–6656. doi: 10.1128/jvi.72.8.6646-6656.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Guadalupe M, et al. Severe CD4+ T-cell depletion in gut lymphoid tissue during primary human immunodeficiency virus type 1 infection and substantial delay in restoration following highly active antiretroviral therapy. J Virol. 2003;77:11708–11717. doi: 10.1128/JVI.77.21.11708-11717.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Shacklett BL, et al. Trafficking of human immunodeficiency virus type 1-specific CD8+ Tcells to gut-associated lymphoid tissue during chronic infection. J Virol. 2003;77:5621–5631. doi: 10.1128/JVI.77.10.5621-5631.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Mehandru S, et al. Primary HIV-1 infection is associated with preferential depletion of CD4+ T lymphocytes from effector sites in the gastrointestinal tract. J Exp Med. 2004;200:761–770. doi: 10.1084/jem.20041196. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Brenchley JM, et al. CD4+ T cell depletion during all stages of HIV disease occurs predominantly in the gastrointestinal tract. J Exp Med. 2004;200:749–759. doi: 10.1084/jem.20040874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25.Picker LJ, et al. Insufficient production and tissue delivery of CD4+ memory T cells in rapidly progressive simian immunodeficiency virus infection. J Exp Med. 2004;200:1299–1314. doi: 10.1084/jem.20041049. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Schuitemaker H, et al. Biological phenotype of human immunodeficiency virus type 1 clones at different stages of infection: progression of disease is associated with a shift from monocytotropic to T-cell tropic virus populations. J Virol. 1992;66:1354–1360. doi: 10.1128/jvi.66.3.1354-1360.1992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Poles MA, Elliott J, Taing P, Anton PA, Chen IS. A preponderance of CCR5(+) CXCR4(+) mononuclear cells enhances gastrointestinal mucosal susceptibility to human immunodeficiency virus type 1 infection. J Virol. 2001;75:8390–8399. doi: 10.1128/JVI.75.18.8390-8399.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lapenta C, et al. Human intestinal lamina propria lymphocytes are naturally permissive to HIV-1 infection. Eur J Immunol. 1999;29:1202–1208. doi: 10.1002/(SICI)1521-4141(199904)29:04<1202::AID-IMMU1202>3.0.CO;2-O. [DOI] [PubMed] [Google Scholar]
  • 29.Ye P, et al. Requirement of interleukin 17 receptor signaling for lung CXC chemokine and granulocyte colony-stimulating factor expression, neutrophil recruitment, and host defense. J Exp Med. 2001;194:519–527. doi: 10.1084/jem.194.4.519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Brand S, et al. IL-22 is increased in active Crohn’s disease and promotes proinflammatory gene expression and intestinal epithelial cell migration. Am J Physiol Gastrointest Liver Physiol. 2006;290:G827–G838. doi: 10.1152/ajpgi.00513.2005. [DOI] [PubMed] [Google Scholar]
  • 31.Sugimoto K, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest. 2008;118:534–544. doi: 10.1172/JCI33194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Aujla SJ, Kolls JK. IL-22: a critical mediator in mucosal host defense. J Mol Med. 2009;87:451–454. doi: 10.1007/s00109-009-0448-1. [DOI] [PubMed] [Google Scholar]
  • 33.Brenchley JM, et al. Differential Th17 CD4 T-cell depletion in pathogenic and nonpathogenic lentiviral infections. Blood. 2008;112:2826–2835. doi: 10.1182/blood-2008-05-159301. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Cecchinato V, et al. Altered balance between Th17 and Th1 cells at mucosal sites predicts AIDS progression in simian immunodeficiency virus-infected macaques. Mucosal Immunol. 2008;1:279–288. doi: 10.1038/mi.2008.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Favre D, et al. Critical loss of the balance between Th17 and Tregulatory cell populations in pathogenic SIV infection. PLoS Pathog. 2009;5:e1000295. doi: 10.1371/journal.ppat.1000295. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Gordon SN, et al. Disruption of intestinal CD4+ T cell homeostasis is a key marker of systemic CD4+ Tcell activation in HIV-infected individuals. J Immunol. 2010;185:5169–5179. doi: 10.4049/jimmunol.1001801. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Klatt NR, Brenchley JM. Th17 cell dynamics in HIV infection. Curr Opin HIV AIDS. 2010;5:135–140. doi: 10.1097/COH.0b013e3283364846. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Chege D, et al. Sigmoid Th17 populations, the HIV latent reservoir, and microbial translocation in men on long-term antiretroviral therapy. AIDS. 2011;25:741–749. doi: 10.1097/QAD.0b013e328344cefb. [DOI] [PubMed] [Google Scholar]
  • 39.Hunt PW. Th17, gut, and HIV: therapeutic implications. Curr Opin HIV AIDS. 2010;5:189–193. doi: 10.1097/COH.0b013e32833647d9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Klatt NR, 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;5:646–657. doi: 10.1038/mi.2012.38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Xu H, et al. IL-17-producing innate lymphoid cells are restricted to mucosal tissues and are depleted in SIV-infected macaques. Mucosal Immunol. 2012;5:658–669. doi: 10.1038/mi.2012.39. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Reeves RK, et al. Gut inflammation and indoleamine deoxygenase inhibit IL-17 production and promote cytotoxic potential in NKp44 + mucosal NK cells during SIV infection. Blood. 2011;118:3321–3330. doi: 10.1182/blood-2011-04-347260. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Nigam P, Kwa S, Velu V, Amara RR. Loss of IL-17-producing CD8 T cells during late chronic stage of pathogenic simian immunodeficiency virus infection. J Immunol. 2010;186:745–753. doi: 10.4049/jimmunol.1002807. [DOI] [PubMed] [Google Scholar]
  • 44.Boasso A, et al. HIV inhibits CD4+ T-cell proliferation by inducing indoleamine 2,3-dioxygenase in plasmacytoid dendritic cells. Blood. 2007;109:3351–3359. doi: 10.1182/blood-2006-07-034785. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Manches O, et al. HIV-activated human plasmacytoid DCs induce Tregs through an indoleamine 2,3-dioxygenase-dependent mechanism. J Clin Invest. 2008;118:3431–3439. doi: 10.1172/JCI34823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Favre D, et al. Tryptophan catabolism by indoleamine 2,3-dioxygenase 1 alters the balance of TH17 to regulatory Tcells in HIV disease. Sci Transl Med. 2010;2:32r–36r. doi: 10.1126/scitranslmed.3000632. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Estes JD, et al. Damaged intestinal epithelial integrity linked to microbial translocation in pathogenic simian immunodeficiency virus infections. PLoS Pathog. 2010;6:e1001052. doi: 10.1371/journal.ppat.1001052. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Nazli A, et al. Exposure to HIV-1 directly impairs mucosal epithelial barrier integrity allowing microbial translocation. PLoS Pathog. 2010;6:e1000852. doi: 10.1371/journal.ppat.1000852. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.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;171:1952–1965. doi: 10.2353/ajpath.2007.070017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Li Q, et al. Simian immunodeficiency virus-induced intestinal cell apoptosis is the underlying mechanism of the regenerative enteropathy of early infection. J Infect Dis. 2008;197:420–429. doi: 10.1086/525046. [DOI] [PubMed] [Google Scholar]
  • 51.Barcellini W, et al. Cytokines and soluble receptor changes in the transition from primary to early chronic HIV type 1 infection. AIDS Res Hum Retroviruses. 1996;12:325–331. doi: 10.1089/aid.1996.12.325. [DOI] [PubMed] [Google Scholar]
  • 52.Ruemmele FM, et al. Lipopolysaccharide modulation of normal enterocyte turnover by toll-like receptors is mediated by endogenously produced tumour necrosis factor alpha. Gut. 2002;51:842–848. doi: 10.1136/gut.51.6.842. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Grunfeld C, Palladino MA. Jr Tumor necrosis factor: immunologic, antitumor, metabolic, and cardiovascular activities. Adv Intern Med. 1990;35:45–71. [PubMed] [Google Scholar]
  • 54.Tilg H, Kaser A. Antitumour necrosis factor therapy in Crohn’s disease. Expert Opin Biol Ther. 2002;2:715–721. doi: 10.1517/14712598.2.7.715. [DOI] [PubMed] [Google Scholar]
  • 55.Tabb B, et al. Reduced inflammation and lymphoid tissue immunopathology in rhesus macaques receiving anti-TNF treatment during primary SIV infection. J Infect Dis. 2012;207:880–892. doi: 10.1093/infdis/jis643. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.WHO-UNAIDS. UNAIDS Report on the Global AIDS Epidemic. 2010 [Google Scholar]
  • 57.Koup RA, Douek DC. Vaccine design for CD8 T lymphocyte responses. Cold Spring Harb Perspect Med. 2011;1:a007252. doi: 10.1101/cshperspect.a007252. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Migueles SA, Connors M. Long-term nonprogressive disease among untreated HIV-infected individuals: clinical implications of understanding immune control of HIV. JAMA. 2010;304:194–201. doi: 10.1001/jama.2010.925. [DOI] [PubMed] [Google Scholar]
  • 59.Brenchley JM, et al. High frequencies of polyfunctional HIV-specific T cells are associated with preservation of mucosal CD4 T cells in bronchoalveolar lavage. Mucosal Immunol. 2008;1:49–58. doi: 10.1038/mi.2007.5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Ferre AL, et al. Mucosal immune responses to HIV-1 in elite controllers: a potential correlate of immune control. Blood. 2009;113:3978–3989. doi: 10.1182/blood-2008-10-182709. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Ferre AL, et al. HIV controllers with HLA-DRB1*13 and HLA-DQB1*06 alleles have strong, polyfunctional mucosal CD4+ T-cell responses. J Virol. 2010;84:11020–11029. doi: 10.1128/JVI.00980-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Hansen SG, et al. Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature. 2011;473:523–527. doi: 10.1038/nature10003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Hansen SG, et al. Effector memory Tcell responses are associated with protection of rhesus monkeys from mucosal simian immunodeficiency virus challenge. Nat Med. 2009;15:293–299. doi: 10.1038/nm.1935. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Freel SA, et al. Phenotypic and functional profile of HIV-inhibitory CD8 T cells elicited by natural infection and heterologous prime/boost vaccination. J Virol. 2010;84:4998–5006. doi: 10.1128/JVI.00138-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Scheid JF, et al. Sequence and structural convergence of broad and potent HIV antibodies that mimic CD4 binding. Science. 2011;333:1633–1637. doi: 10.1126/science.1207227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 66.Veazey RS, et al. Prevention of virus transmission to macaque monkeys by a vaginally applied monoclonal antibody to HIV-1 gp120. Nat Med. 2003;9:343–346. doi: 10.1038/nm833. [DOI] [PubMed] [Google Scholar]
  • 67.Devito C, et al. Mucosal and plasma IgA from HIV-1-exposed uninfected individuals inhibit HIV-1 transcytosis across human epithelial cells. J Immunol. 2000;165:5170–5176. doi: 10.4049/jimmunol.165.9.5170. [DOI] [PubMed] [Google Scholar]
  • 68.Broliden K, et al. Functional HIV-1 specific IgA antibodies in HIV-1 exposed, persistently IgG seronegative female sex workers. Immunol Lett. 2001;79:29–36. doi: 10.1016/s0165-2478(01)00263-2. [DOI] [PubMed] [Google Scholar]
  • 69.Mestecky J, et al. Paucity of antigen-specific IgA responses in sera and external secretions of HIV-type 1-infected individuals. AIDS Res Hum Retroviruses. 2004;20:972–988. doi: 10.1089/aid.2004.20.972. [DOI] [PubMed] [Google Scholar]
  • 70.Brenchley JM, Price DA, Douek DC. HIV disease: fallout from a mucosal catastrophe? Nat Immunol. 2006;7:235–239. doi: 10.1038/ni1316. [DOI] [PubMed] [Google Scholar]
  • 71.Brenchley JM, et al. Microbial translocation is a cause of systemic immune activation in chronic HIV infection. Nat Med. 2006;12:1365–1371. doi: 10.1038/nm1511. [DOI] [PubMed] [Google Scholar]
  • 72.Klatt NR, Funderburg NT, Brenchley JM. Microbial translocation, immune activation, and HIV disease. Trends Microbiol. 2012;21:6–13. doi: 10.1016/j.tim.2012.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Brenchley JM, Douek DC. Microbial translocation across the GI tract. Annu Rev Immunol. 2012;30:149–173. doi: 10.1146/annurev-immunol-020711-075001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Redd AD, et al. Microbial translocation, the innate cytokine response, and HIV-1 disease progression in Africa. Proc Natl Acad Sci USA. 2009;106:6718–6723. doi: 10.1073/pnas.0901983106. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Redd AD, et al. C-reactive protein levels increase during HIV-1 disease progression in Rakai, Uganda, despite the absence of microbial translocation. J Acquir Immune Defic Syndr. 2010;54:556–559. doi: 10.1097/QAI.0b013e3181e0cdea. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Siliciano JD, Siliciano RF. Biomarkers of HIV replication. Curr Opin HIV AIDS. 2010;5:491–497. doi: 10.1097/COH.0b013e32833f206f. [DOI] [PubMed] [Google Scholar]
  • 77.Kuller LH, et al. Inflammatory and coagulation biomarkers and mortality in patients with HIV infection. PLoS Med. 2008;5:e203. doi: 10.1371/journal.pmed.0050203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Sandler NG, et al. Plasma levels of soluble CD14 independently predict mortality in HIV infection. J Infect Dis. 2011;203:780–790. doi: 10.1093/infdis/jiq118. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Arnsten JH, et al. Decreased bone mineral density and increased fracture risk in aging men with or at risk for HIV infection. AIDS. 2007;21:617–623. doi: 10.1097/QAD.0b013e3280148c05. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.McCutchan JA, et al. HIV suppression by HAART preserves cognitive function in advanced, immune-reconstituted AIDS patients. AIDS. 2007;21:1109–1117. doi: 10.1097/QAD.0b013e3280ef6acd. [DOI] [PubMed] [Google Scholar]
  • 81.Burdo TH, et al. Soluble CD163, a novel marker of activated macrophages, is elevated and associated with noncalcified coronary plaque in HIV-infected patients. J Infect Dis. 2011;204:1227–1236. doi: 10.1093/infdis/jir520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Anselmi A, et al. Immune reconstitution in human immunodeficiency virus type 1-infected children with different virological responses to anti-retroviral therapy. Clin Exp Immunol. 2007;150:442–450. doi: 10.1111/j.1365-2249.2007.03526.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Baroncelli S, et al. Microbial translocation is associated with residual viral replication in HAART-treated HIV + subjects with <50copies/ml HIV-1 RNA. J Clin Virol. 2009;46:367–370. doi: 10.1016/j.jcv.2009.09.011. [DOI] [PubMed] [Google Scholar]
  • 84.Cassol E, et al. Persistent microbial translocation and immune activation in HIV-1-infected South Africans receiving combination antiretroviral therapy. J Infect Dis. 2010;202:723–733. doi: 10.1086/655229. [DOI] [PubMed] [Google Scholar]
  • 85.Jiang W, et al. Plasma levels of bacterial DNA correlate with immune activation and the magnitude of immune restoration in persons with antiretroviral-treated HIV infection. J Infect Dis. 2009;199:1177–1185. doi: 10.1086/597476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Marchetti G, et al. Microbial translocation is associated with sustained failure in CD4+ T-cell reconstitution in HIV-infected patients on long-term highly active antiretroviral therapy. AIDS. 2008;22:2035–2038. doi: 10.1097/QAD.0b013e3283112d29. [DOI] [PubMed] [Google Scholar]
  • 87.Massanella M, et al. CD4 T-cell hyperactivation and susceptibility to cell death determine poor CD4 T-cell recovery during suppressive HAART. AIDS. 2010;24:959–968. doi: 10.1097/QAD.0b013e328337b957. [DOI] [PubMed] [Google Scholar]
  • 88.Miao YM, et al. 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 CD4 T cell densities. J Infect Dis. 2002;185:1043–1050. doi: 10.1086/340235. [DOI] [PubMed] [Google Scholar]
  • 89.Mehandru S, et al. Lack of mucosal immune reconstitution during prolonged treatment of acute and early HIV-1 infection. PLoS Med. 2006;3:e484. doi: 10.1371/journal.pmed.0030484. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Guadalupe M, et al. Viral suppression and immune restoration in the gastrointestinal mucosa of human immunodeficiency virus type 1-infected patients initiating therapy during primary or chronic infection. J Virol. 2006;80:8236–8247. doi: 10.1128/JVI.00120-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Schacker TW, et al. Amount of lymphatic tissue fibrosis in HIV infection predicts magnitude of HAART-associated change in peripheral CD4 cell count. AIDS. 2005;19:2169–2171. doi: 10.1097/01.aids.0000194801.51422.03. [DOI] [PubMed] [Google Scholar]
  • 92.Estes JD, et al. CD4 reconstitution of lymphoid tissues is dependent on earlier initiation of HAART. Conferences on Retroviruses and Opportunistic Infections; Los Angeles, CA. 2007. [Google Scholar]
  • 93.Zeng M, et al. Cumulative mechanisms of lymphoid tissue fibrosis and T cell depletion in HIV-1 and SIV infections. J Clin Invest. 2011;121:998–1008. doi: 10.1172/JCI45157. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Tannock GW. Molecular analysis of the intestinal microflora in IBD. Mucosal Immunol. 2008;1 (Suppl 1):S15–S18. doi: 10.1038/mi.2008.54. [DOI] [PubMed] [Google Scholar]
  • 95.Walker AW, et al. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiol. 2010;11:7. doi: 10.1186/1471-2180-11-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Gori A, et al. Early impairment of gut function and gut flora supporting a role for alteration of gastrointestinal mucosa in human immunodeficiency virus pathogenesis. J Clin Microbiol. 2008;46:757–758. doi: 10.1128/JCM.01729-07. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Wolf BW, Wheeler KB, Ataya DG, Garleb KA. Safety and tolerance of Lactobacillus reuteri supplementation to a population infected with the human immunodeficiency virus. Food Chem Toxicol. 1998;36:1085–1094. doi: 10.1016/s0278-6915(98)00090-8. [DOI] [PubMed] [Google Scholar]
  • 98.Gori A, 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;4:554–563. doi: 10.1038/mi.2011.15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ellis CL, et al. Molecular characterization of stool microbiota in HIV-infected subjects by panbacterial and order-level 16S ribosomal DNA (rDNA) quantification and correlations with immune activation. J Acquir Immune Defic Syndr. 2011;57:363–370. doi: 10.1097/QAI.0b013e31821a603c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.McKenna P, et al. The macaque gut microbiome in health, lentiviral infection, and chronic enterocolitis. PLoS Pathog. 2008;4:e20. doi: 10.1371/journal.ppat.0040020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Handley SA, et al. Pathogenic simian immunodeficiency virus infection is associated with expansion of the enteric virome. Cell. 2012;151:253–266. doi: 10.1016/j.cell.2012.09.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Klatt NR, et al. Probiotic/prebiotic supplementation of antiretrovirals improves gastrointestinal immunity in SIV-infected macaques. J Clin Invest. 2013;123:903–907. doi: 10.1172/JCI66227. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Hummelen R, et al. Effect of 25 weeks probiotic supplementation on immune function of HIV patients. Gut Microbes. 2011;2:80–85. doi: 10.4161/gmic.2.2.15787. [DOI] [PubMed] [Google Scholar]
  • 104.Hummelen R, et al. Lactobacillus rhamnosus GR-1 and L. reuteri RC-14 to prevent or cure bacterial vaginosis among women with HIV. Int J Gynaecol Obstet. 2010;111:245–248. doi: 10.1016/j.ijgo.2010.07.008. [DOI] [PubMed] [Google Scholar]
  • 105.Hummelen R, Vos AP, van’t Land B, van Norren K, Reid G. Altered host-microbe interaction in HIV: a target for intervention with pro- and prebiotics. Int Rev Immunol. 2010;29:485–513. doi: 10.3109/08830185.2010.505310. [DOI] [PubMed] [Google Scholar]
  • 106.Hummelen R, Hemsworth J, Reid G. Micronutrients, N-acetyl cysteine, probiotics and prebiotics, a review of effectiveness in reducing HIV progression. Nutrients. 2011;2:626–651. doi: 10.3390/nu2060626. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Gonzalez-Hernandez LA, 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. doi: 10.1186/1475-2891-11-90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Schunter M, et al. Randomized pilot trial of a synbiotic dietary supplement in chronic HIV-1 infection. BMC Complement Altern Med. 2012;12:84. doi: 10.1186/1472-6882-12-84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Bibiloni R, et al. VSL#3 probiotic-mixture induces remission in patients with active ulcerative colitis. Am J Gastroenterol. 2005;100:1539–1546. doi: 10.1111/j.1572-0241.2005.41794.x. [DOI] [PubMed] [Google Scholar]
  • 110.Sood A, et al. The probiotic preparation, VSL#3 induces remission in patients with mild-to-moderately active ulcerative colitis. Clin Gastroenterol Hepatol. 2009;7:1202–1209. doi: 10.1016/j.cgh.2009.07.016. [DOI] [PubMed] [Google Scholar]
  • 111.Zocco MA, et al. Efficacy of Lactobacillus GG in maintaining remission of ulcerative colitis. Aliment Pharmacol Ther. 2006;23:1567–1574. doi: 10.1111/j.1365-2036.2006.02927.x. [DOI] [PubMed] [Google Scholar]
  • 112.Gupta P, Andrew H, Kirschner BS, Guandalini S. Is lactobacillus GG helpful in children with Crohn’s disease? Results of a preliminary, open-label study. J Pediatr Gastroenterol Nutr. 2000;31:453–457. doi: 10.1097/00005176-200010000-00024. [DOI] [PubMed] [Google Scholar]
  • 113.Byakwaga H, et al. Intensification of antiretroviral therapy with raltegravir or addition of hyperimmune bovine colostrum in HIV-infected patients with suboptimal CD4+ T-cell response: a randomized controlled trial. J Infect Dis. 2011;204:1532–1540. doi: 10.1093/infdis/jir559. [DOI] [PubMed] [Google Scholar]
  • 114.Pandrea I, Apetrei C. Therapeutic interventions to reduce microbial translocation and immune activation. AIDS; Conference; 25 July 2012; Washington, DC. 2012. [Google Scholar]

RESOURCES